3rd EFMC Young Medicinal Symposium

September 1-2, 2016 | Manchester | United Kingdom

Book of Abstracts

Content

Welcome 2

Sponsors 3

Exhibitors’ Company Profile 5

Programme 6

Flash Poster Presentations List 9

Keynote Lectures 11

Winners of the National Medicinal Chemist Meeting 15

Oral Communications 31

Flash Poster Presentations 37

Posters Abstracts 59

List of abstracts 119

List of participants 125

1 Welcome

Dear participant

On behalf of the European Federation for Medicinal Chemistry (EFMC) and the Organizing Committee we warmly welcome you to Manchester for the 3rd edition of the EFMC Young Medicinal Chemist Symposium (EFMC-YMCS).

This edition has been organised on behalf of the EFMC in connection with the XXIV EFMC International Symposium on Medicinal Chemistry (EFMC-ISMC 2016) in keeping with the initial aim of: • Creating a network of young European investigators in Medicinal Chemistry • Stimulating young European investigators in Medicinal Chemistry to share their scientific work with peers and inspiring leaders in the field • Creating competition and excellence in Medicinal Chemistry within Europe by selecting the European Champion in Medicinal Chemistry

Over 125 scientists coming from 30 nations will gather at the Manchester Institute of Biotechnology for this exciting mini-symposium. The 2 keynote lectures, 14 oral communications by invited prize winners from national young medicinal chemist meetings in Europe, 4 additional selected oral communications, 20 Flash Poster Presentations and more than 70 poster presentations will lead you through the latest drug discovery advances in the major therapeutic areas, including neglected diseases, CNS disorders, inflammation, metabolic disorders, and oncology.

During the closing Award ceremony the following prizes will be awarded to the European Champions in Medicinal Chemistry: • The EFMC-YMCS Presentation Prize, sponsored by EFMC & Actelion • The EFMC-YMCS Poster Prize, sponsored by the Sir Simon Campbell & MedChemComm • The EFMC-YMCS Participant’s Prize, sponsored by Roche

We thank our sponsors (Actelion, Evotec, Merck, Roche, RSC, and Sir Simon Campbell), the Manchester Institute for Biotechnology and all the participating organisations for their support and look forward to your active participation!

Prof. Koen Augustijns Prof. Mike Waring EFMC President Symposium Chairman

2 Sponsors

Organised by

With the support of

Participating Organisations

Sponsors

Sir Simon Campbell

3 Sponsors

Award Sponsors

Sir Simon Campbell

Exhibitors

Media Partners

4 Exhibitors’ Company Profile

Chemspeed is the leading provider of high-throughput and high-output research & development workflow-solutions from single bench-top / standalone automated workstations (powder dispensing - sample preparation- synthesis - process development - formulation - application - testing) up to complete and integrated product development workflows for the entire product development processes in the chemical, material science, renewables & energy, pharmaceutical, agrochemical, specialty chemical, home care, cosmetics and nutrition industries, as well as academia.

Contact details: Chemspeed Technologies AG Rheinstrasse 32 CH-4302 Augst, Switzerland +41 61 816 95 09 - [email protected] - www.chemspeed.com

Merck Millipore and Sigma-Aldrich come together, as Merck, to solve the toughest problems in life science by collaborating with the global scientific community. The life science business of Merck has a global network spanning more than 60 countries, approximately 70 manufacturing sites, 19,000 employees and over 1 million customers. The Company’s portfolio of over 300,000 products can be viewed online - for more information, visit merckmillipore.com and sigma-aldrich.com Looking forward to meeting you on our booth.

Contact Details: Sigma-Aldrich Chimie Sarl 80 rue de Luzais 38297 St Quentin Fallavier, France [email protected] - www.sigma-aldrich.com

5 Programme

Thursday September 1, 2016

14:15 Registration

15:00 Welcome and opening

15:05 Case Histories of Fragment-Based Drug Discovery (KL01) Dr David REES (ASTEX PHARMACEUTICALS, Cambridge, United Kingdom)

15:35 Heterocycles and Medicinal Chemistry: the Importance of Innovative Synthesis (KL02) Dr Anthony WOOD (PFIZER, Cambridge, United States)

YMCS Competition Presentation Session I

16:05 New Avenues in Protein Labeling - Exploiting Dna for Directing Chemical Reactions Winner of the young medicinal chemist meeting in Denmark (CP01) Dr Anne Louise Bank KODAL (NOVO NORDISK, Bagsværd, Denmark)

16:25 Drug Design, Synthesis and Biological Evaluation of Antiretroviral and Anticancer Agents Winner of the young medicinal chemist meeting in Italy (CP02) Dr Valeria FAMILGLINI (UNIVERSITY OF ROME, Rome, Italy)

16:45 Development of Potent, Cell-Active Inhibitors of the Mycobacterium Tuberculosis Oxidase Cyp125 (OC01) Ms Madeline KAVANAGH (, Cambridge, United Kingdom)

17:05 Flash Poster Presentations (see list on page 9)

17:45 Poster presentation - session 1 (odd numbers) & Networking drink

19:00 End of the day

6 Programme

Friday September 2, 2016

YMCS Competition Presentation Session II

08:30 Antimalarial Drug Discovery: Exploring the Mep Pathway Winner of the young medicinal chemist meeting in the United Kingdom (CP03) Ms Kathryn PRICE (UNIVERSITY OF LIVERPOOL, Liverpool, United Kingdom)

08:50 Drugging the Undrugguble: Targeting Challenging E3 Ligases Winner of the young medicinal chemist meeting in Spain (CP04) Dr Carles GALDEANO (UNIVERSITAT DE BARCELONA, Barcelona, Spain)

09:10 Development of Highly Selective and Reversible Diacylglycerol Lipase Inhibitors Winner of the young medicinal chemist meeting in The Netherlands (CP05) Mr Janssen FREEK (LEIDEN UNIVERSITY, leiden , The Netherlands)

09:30 Functional Lipidomics-Based Lead Discovery Reveals Novel Role of Phospholipids in A Signaling Winner of the young medicinal chemist meeting in Germany (CP06) Dr Andreas KOEBERLE (FRIEDRICH SCHILLER UNIVERSITY JENA, Jena, Germany)

09:50 Design, Synthesis and Biological Evaluation of Selective CDK Inhibitors based on Flavopiridol Structure Winner of the young medicinal chemist meeting in Greece (CP07) Dr Nikitia MEXIA (UNIVERSITY OF ATHENS, Athens, Greece)

10:10 Coffee break

YMCS Competition Presentation Session III

10:35 Design and Synthesis of 2,2’-Diindolylmethanes to Selectively Target Certain G-Quadruplex DNA Structures Winner of the young medicinal chemist meeting in Sweden (CP08) Dr Madeleine LIVENDAHL (UMEA UNIVERSITY, Umeå, Sweden)

10:55 Stable, Effective, and Selective Anticancer Phenolato Titanium(Iv) Complexes Winner of the young medicinal chemist meeting in Israel (CP09) Mrs Sigalit MEKER (THE HEBREW UNIVERSITY OF JERUSALEM, Jerusalem, Israel)

11:15 Glyco-Conjugated Photosensitizers: From Synthesis to Validation in Cancer Photodynamic Therapy Winner of the young medicinal chemist meeting in Portugal (CP10) Ms Patricia PEREIRA (UNIVERSIDADE DE AVEIRO, Aveiro, Portugal)

11:35 Discovery of Cct251921: A Potent, Selective and Orally Bioavailable Small Molecule Modulator of the Mediator Complex-Associated Kinases Cdk8 and Cdk19 (OC02) Dr Aurelie MALLINGER (INSTITUTE OF CANCER RESEARCH, Sutton, London, United Kingdom)

11:55 Lunch and Poster presentation - session 2 (even numbers)

7 Programme

YMCS Competition Presentation Session IV

13:15 How to Design Cell Permeable Non-Peptidic Macrocycles (OC03) Dr Björn OVER (MACROCYCLE ADME, BEYOND RO5 AND NATURAL PRODUCT-FRAGMENTS, Landvetter, Sweden)

13:35 New 3d-Scaffolds for Fragment-Based Lead Discovery (Fbld): Exploration of Chemical Space Using Functionalized Spirohydantoins Winner of the young medicinal chemist meeting in France (CP11) Mr Hugues PREVET (LABORATORY U1177 - UNIVERSITY OF LILLE, Lille, France)

13:55 Discovery Of Novel Atpase Inhibitors Of Dna Gyrase And Topoisomerase Iv Starting From Marine Sponge Alkaloid Oroidin Winner of the young medicinal chemist meeting in Slovenia (CP12) Dr Tihomir TOMASIC (UNIVERSITY OF LJUBLJANA, Ljubljana, Slovenia)

14:15 Purine Mimicking 3’-Ethynylribofuranose Nucleosides: New Opportunities for an Old Sugar Modification Winner of the young medicinal chemist meeting in Belgium (KVCV) (CP13) Mr Fabian HULPIA (UGENT, Ghent, Belgium)

14:35 Experimental Platform for Characterization and High-Throughput Screening of Acetylcholinesterase Inhibitors Winner of the young medicinal chemist meeting in Poland (CP14) Dr Piotr DRACZKOWSKI (UNIVERSITY OF LUBLIN, Lublin, Poland)

14:55 Novel Derivatives of the Tspo Ligand Dpa-714 For in Vivo Pet Imaging of Neuroinflammation (OC04) Mrs Fanny CACHEUX (CEA, Orsay, France)

15:15 Voting Break

15:30 Closing and award ceremony

8 Flash Poster Presentations

• FP01 - Toward Libraries of Artificial Macrocycles Mrs Eman ABD ELRAHEEM (GRONINGEN UNIVERSITY, Groningen, The Netherlands) • FP02 - Discovery, Structural Exploration and Biological Profiling of a Novel Class of Antimycobacterial Dpre1 Inhibitors Ms Olga BALABON (UNIVERSITY OF ANTWERP, Wilrijk, Belgium) • FP03 - Orally Bioavailable Antimalarial 4(1h)-Quinolone Prodrugs with Single-Dose Cures Dr Fabian BROCKMEYER (NORTHEASTERN UNIVERSITY, Boston, United States) • FP04 - Developement of Synthesis Methodology for Dna-Encoded Small Molecule Screening Libraries Mrs Mateja KLIKA SKOPIC (TECHNISCHE UNIVERSITÄT DORTMUND, Dortmund, Germany) • FP05 - Fishing for an Off-Target”: Deconvoluting the Molecular Mechanism of Gpcr-Induced Macrophage Chemotaxis” Dr Maria CHATZOPOULOU (UNIVERSITY OF OXFORD, Oxford, United Kingdom) • FP06 - Design, Synthesis and Pharmacological Evaluation of Fluorescent Tools for Studying Cannabinoid Type 2 Receptor Ms Anna COOPER (UNIVERSITY OF OTAGO, Dunedin, New Zealand) • FP07 - Development of Novel, Selective and Irreversible Pi3kδ Inhibitors Mr Sam DALTON (UNIVERSITY OF STRATHCLYDE, Glasgow, United Kingdom) • FP08 - Identification of Novel Non-Natural Supramolecular Ligands as Stabilizers of 14-3-3? Protein-Protein Interactions Dr Alba GIGANTE (DUISBURG ESSEN UNIVERSITY, Essen, Germany) • FP09 - Targets in Orphan Diseases and Their Repurposing Candidates - Analysis with a Knime Workflow Using Open Phacts Ms Jana GURINOVA (UNIVERSITY OF VIENNA, Vienna, Austria) • FP10 - Selectivity Profiling of the Human Monoamine Transporters: A Case Study on Cathinone Analogs Ms Eva HELLSBERG (UNIVERSITY OF VIENNA, Vienna, Austria) • FP11 - Photoswitchable 4-(Piperidin-4-Yl)-1-Hydroxypyrazole Analogues for the Gaba Type A Receptors Ms Henriette KIHL (UNIVERSITY OF COPENHAGEN, Copenhagen, Denmark) • FP12 - Extending Sar of Bile Acids as Fxr/Tgr5 Modulators: Synthesis and Biological Activity of Ring C Modified Cdca Derivatives Ms Serena MOSTARDA (UNIVERSITÀ DEGLI STUDI DI PERUGIA, Perugia, Italy) • FP13 - Lead Optimisation of a Novel Class of Trypanosomacidal Agents Ms Harriet NEWSON (UNIVERSITY OF WESTERN AUSTRALIA, Crawley, Australia) • FP14 - Synthesis and Biological Evaluation of Diaromatic Guanidinium-Like Derivatives as Protein Kinase Inhibitors for the Treatment of Cancer Ms Viola PREVITALI (TRINITY COLLEGE DUBLIN, Dublin, Ireland) • FP15 - Development of Novel 1,4- as Antitrypanosomal Agents Dr Santo PREVITI (UNIVERSITY OF MESSINA, Messina, Italy) • FP16 - Determining the Binding Mode of 3-(1-Alkyl-1h-Imidazol-5-Yl)-1h-Indoles Mr Adam HOGENDORF (INSTITUTE OF PHARMACOLOGY, Kraków, Poland) • FP17 - Targeting the Human Dihydroorotate Dehydrogenase (Hdhodh) by a Scaffold Hopping Bioisosteric Approach Using Hydroxylated Pentaatomic Heterocycles Mr Stefano SAINAS (UNIVERSITA DEGLI STUDI DI TORINO, Torino, Italy) • FP18 - Towards the Elucidation of the Mechanism of Action of Small Molecule Upregulators of Utrophin Using Chemical Proteomics Ms Aini VUORINEN (UNIVERSITY OF OXFORD, Oxford, United Kingdom) • FP19 - Development of an Albumin-Binding Ligand for Prolonging the Plasma Half-Life of Peptide Therapeutics Mr Alessandro ZORZI (ECOLE POLYTECHNIQUE FÉDÉRALE DE LAUSANNE , Lausanne, Switzerland) • FP20 - Near-Infrared (Nir) Photothermal Therapy Using Biocompatible Palladium Nanoparticles Dr Belen RUBIO RUIZ (UNIVERSITY OF EDINBURGH, Edinburgh, United Kingdom)

9 Notes

______

10 Keynote Lectures

11 KL01 KL02 CASE HISTORIES OF FRAGMENT-BASED DRUG DISCOVERY HETEROCYCLES AND MEDICINAL CHEMISTRY: THE IMPORTANCE OF INNOVATIVE SYNTHESIS David Rees Anthony Wood Astex Pharmaceuticals 436 Cambridge Science Park Cambridge, CB4 0QA SVP and Head of Worldwide Medicinal Chemistry, Pfizer United Kingdom [email protected] [email protected]

Heterocyclic organic chemistry lies at the heart of modern medicinal chemistry design and synthesis. The lecture Fragment Based Drug Discovery (FBDD) is used in many academic and industrial laboratories. Over 20 will use some brief examples from recent projects at Pfizer to illustrate the importance of novel heterocycle FBDD-derived compounds have been progressed into clinical trials and two oncology drugs launched. synthesis as a means of accessing target molecules with appropriate properties. Examples will include compounds synthesized for SGLT2, GPR119, ACC, PDE9, BACE and ALK projects. I will also describe a The talk will describe FBDD at Astex including a fragment-to-clinic example. FBDD uses X-ray crystallography framework to evaluate the importance of complexity, including chirality, in the design and synthesis of target and other biophysical screening techniques (eg NMR, ITC) to identify initial fragment hits against a variety of molecules and screening libraries. protein targets (eg kinases, proteases, PPIs). Typically these fragments have MW = 120-220 and binding affinities between mM - uM.

12 KL01 KL02 CASE HISTORIES OF FRAGMENT-BASED DRUG DISCOVERY HETEROCYCLES AND MEDICINAL CHEMISTRY: THE IMPORTANCE OF INNOVATIVE SYNTHESIS David Rees Anthony Wood Astex Pharmaceuticals 436 Cambridge Science Park Cambridge, CB4 0QA SVP and Head of Worldwide Medicinal Chemistry, Pfizer United Kingdom [email protected] [email protected]

Heterocyclic organic chemistry lies at the heart of modern medicinal chemistry design and synthesis. The lecture Fragment Based Drug Discovery (FBDD) is used in many academic and industrial laboratories. Over 20 will use some brief examples from recent projects at Pfizer to illustrate the importance of novel heterocycle FBDD-derived compounds have been progressed into clinical trials and two oncology drugs launched. synthesis as a means of accessing target molecules with appropriate properties. Examples will include compounds synthesized for SGLT2, GPR119, ACC, PDE9, BACE and ALK projects. I will also describe a The talk will describe FBDD at Astex including a fragment-to-clinic example. FBDD uses X-ray crystallography framework to evaluate the importance of complexity, including chirality, in the design and synthesis of target and other biophysical screening techniques (eg NMR, ITC) to identify initial fragment hits against a variety of molecules and screening libraries. protein targets (eg kinases, proteases, PPIs). Typically these fragments have MW = 120-220 and binding affinities between mM - uM.

13 Notes

______

14 Winners of the National Medicinal Chemist Meeting

15 CP01 CP02 NEW AVENUES IN PROTEIN LABELING - EXPLOITING DNA FOR DRUG DESIGN, SYNTHESIS AND BIOLOGICAL EVALUATION OF DIRECTING CHEMICAL REACTIONS ANTIRETROVIRAL AND ANTICANCER AGENTS

Anne Louise B. Kodal, Christian B. Rosen, Thomas Tørring, Kurt V. Gothelf Famiglini Valeria

Center for DNA Nanotechnology, Department of Chemistry and iNANO, Aarhus University, Aarhus C, Denmark Istituto Pasteur Italy - Fondazione Cenci Bolognetti, Dipartimento di Chimica e Tecnologie del Farmaco, Sapienza Università di Roma, Piazzale Aldo Moro 5, I-00185 Roma, Italy

In recent years biopharmaceuticals have become an integral and rapidly expanding part of the pharmaceutical industry. The sophistication of biological molecules facilitates access to previously undruggable or challenging The investigation into the principle causes of mortality worldwide must always take account of the profound targets and their chemical modification allows introduction of tailor-made properties. Protein labeling has been differences between low and high income countries. In rich countries, the majority of the population die of used to extend in vivo half-life using PEGylation, and more recently facilitated directed delivery of therapeutics deseases like cancer and cardiovascular complications. In the poorest countries the causes of death are above all through antibody conjugation. HIV and other infections. Despite the good results achieved to date, limitations caused by high toxicity, drug As proteins are derived from 20 common amino acids, site-specific chemical modification poses a significant resistance, complex formulation and limited bioavailability underline the need to develop new agents. Therefore, challenge. Residue specific protein labeling often results in a heterogeneous mixture of products compromising pathologies like AIDS and cancer represent key objectives for research that require enormous economic characterization, purification, and protein activity. Alternatively, genetic engineering allows introduction of resources and a continued commitment to research into new drugs. My thesis, arising from this background site-specific labeling tags or in specialist cases unnatural amino acids. This process can be laborious and context, planned to synthesize and optimize new potential molecules with antiretroviral and anticancer activity. technically challenging. This thesis contains two sections: the first part focused on the development of new indolylarylsulfones (IASs) as Here, we demonstrate a new method for creating site-selective protein conjugates without employing genetic highly potent and broad spectrum HIV-1 non-nucleoside reverse transcriptase inhibitors (Figure 1A);1,3 the engineering. We merge concepts from affinity probe technology and DNA-templated synthesis to guide a second section reported the results of new arylthioindole (ATI) derivatives with tubulin polymerization chemical reaction to the vicinity of a metal-binding site of the protein (Figure 1). The method is termed inhibiting activity as anticancer agents (Figure 1B).2 DNA-templated protein conjugation (DTPC) and was shown to be general for metal-binding proteins including His6-tagged proteins, wild-type metalloproteins, and IgG1 antibodies1,2. The DNA label provides a unique handle to identify, functionalize, or manipulate proteins. We employed DNA-transferrin conjugates to achieve intracellular delivery of a DNA nanostructure3. We furthermore demonstrated site-selective introduction of a single bioorthogonal handle in proteins using a cleavable linker in the reactive DNA labeling strand (Figure 1). This provides a general handle for further conjugation and enabled quantitative labeling with PEG4.

The present project highlight the therapeutic potential of the IAS and ATI classes as antiretroviral and anticancerr agents, respectively, and prompt their further development. We improved the biological activity and the biovailability features of these compounds. References 1) Rosen*, C. B., Kodal*, A. L. B., Nielsen, J. S., Schaffert, D. S., Scavenius, C., Okholm, A. H., Voigt, N. V., Enghild, J. J., Kjems, J., Tørring, T. & Gothelf, K. V. Template-directed covalent conjugation of DNA to native antibodies, transferrin and References other metal-binding proteins. Nat Chem 6, 804–809 (2014) 1) Famiglini, V.; La Regina, G.; Coluccia A.; et al. J. Med Chem. 2014. 57, 9945-9957. 2) Gothelf, K. V., Tørring, T., Rosen, C. B. & Kodal, A. L. B. Site-selective conjugation of an oligonucleotide conjugate or a 2) La Regina, G.; Bai, R.; Rensen, WM.; et al. J.Med. Chem. 2013, 56,123-49. small molecule to a metal binding protein. PD10112PC00 3) La Regina, G.; Gatti, V.; Famiglini, V.; et al. ACS Comb. Sci. (ex J. Comb. Chem.) 2012, 14, 258-62. 3) Schaeffert, D. H., Okholm, A. H., Sørensen, R. S., Nielsen, J. S., Tørring, T. Rosen, C. B., Kodal, A. L. B., Mortensen, M. R., Gothelf, K. V. and Kjems, J. Intracellular delivery of a planar DNA origami structure by the transferrin-receptor internalization pathway. Small 12, 2634-2640 (2016) 4) Kodal*, A. L. B., Rosen*, C. B., Mortensen, M. M., Tørring, T. and Gothelf, K. V. DNA-templated introduction of an aldehyde handle in proteins. Chembiochem, 17, 1338-1342 (2016)

16 CP01 CP02 NEW AVENUES IN PROTEIN LABELING - EXPLOITING DNA FOR DRUG DESIGN, SYNTHESIS AND BIOLOGICAL EVALUATION OF DIRECTING CHEMICAL REACTIONS ANTIRETROVIRAL AND ANTICANCER AGENTS

Anne Louise B. Kodal, Christian B. Rosen, Thomas Tørring, Kurt V. Gothelf Famiglini Valeria

Center for DNA Nanotechnology, Department of Chemistry and iNANO, Aarhus University, Aarhus C, Denmark Istituto Pasteur Italy - Fondazione Cenci Bolognetti, Dipartimento di Chimica e Tecnologie del Farmaco, Sapienza Università di Roma, Piazzale Aldo Moro 5, I-00185 Roma, Italy

In recent years biopharmaceuticals have become an integral and rapidly expanding part of the pharmaceutical industry. The sophistication of biological molecules facilitates access to previously undruggable or challenging The investigation into the principle causes of mortality worldwide must always take account of the profound targets and their chemical modification allows introduction of tailor-made properties. Protein labeling has been differences between low and high income countries. In rich countries, the majority of the population die of used to extend in vivo half-life using PEGylation, and more recently facilitated directed delivery of therapeutics deseases like cancer and cardiovascular complications. In the poorest countries the causes of death are above all through antibody conjugation. HIV and other infections. Despite the good results achieved to date, limitations caused by high toxicity, drug As proteins are derived from 20 common amino acids, site-specific chemical modification poses a significant resistance, complex formulation and limited bioavailability underline the need to develop new agents. Therefore, challenge. Residue specific protein labeling often results in a heterogeneous mixture of products compromising pathologies like AIDS and cancer represent key objectives for research that require enormous economic characterization, purification, and protein activity. Alternatively, genetic engineering allows introduction of resources and a continued commitment to research into new drugs. My thesis, arising from this background site-specific labeling tags or in specialist cases unnatural amino acids. This process can be laborious and context, planned to synthesize and optimize new potential molecules with antiretroviral and anticancer activity. technically challenging. This thesis contains two sections: the first part focused on the development of new indolylarylsulfones (IASs) as Here, we demonstrate a new method for creating site-selective protein conjugates without employing genetic highly potent and broad spectrum HIV-1 non-nucleoside reverse transcriptase inhibitors (Figure 1A);1,3 the engineering. We merge concepts from affinity probe technology and DNA-templated synthesis to guide a second section reported the results of new arylthioindole (ATI) derivatives with tubulin polymerization chemical reaction to the vicinity of a metal-binding site of the protein (Figure 1). The method is termed inhibiting activity as anticancer agents (Figure 1B).2 DNA-templated protein conjugation (DTPC) and was shown to be general for metal-binding proteins including His6-tagged proteins, wild-type metalloproteins, and IgG1 antibodies1,2. The DNA label provides a unique handle to identify, functionalize, or manipulate proteins. We employed DNA-transferrin conjugates to achieve intracellular delivery of a DNA nanostructure3. We furthermore demonstrated site-selective introduction of a single bioorthogonal handle in proteins using a cleavable linker in the reactive DNA labeling strand (Figure 1). This provides a general handle for further conjugation and enabled quantitative labeling with PEG4.

The present project highlight the therapeutic potential of the IAS and ATI classes as antiretroviral and anticancerr agents, respectively, and prompt their further development. We improved the biological activity and the biovailability features of these compounds. References 1) Rosen*, C. B., Kodal*, A. L. B., Nielsen, J. S., Schaffert, D. S., Scavenius, C., Okholm, A. H., Voigt, N. V., Enghild, J. J., Kjems, J., Tørring, T. & Gothelf, K. V. Template-directed covalent conjugation of DNA to native antibodies, transferrin and References other metal-binding proteins. Nat Chem 6, 804–809 (2014) 1) Famiglini, V.; La Regina, G.; Coluccia A.; et al. J. Med Chem. 2014. 57, 9945-9957. 2) Gothelf, K. V., Tørring, T., Rosen, C. B. & Kodal, A. L. B. Site-selective conjugation of an oligonucleotide conjugate or a 2) La Regina, G.; Bai, R.; Rensen, WM.; et al. J.Med. Chem. 2013, 56,123-49. small molecule to a metal binding protein. PD10112PC00 3) La Regina, G.; Gatti, V.; Famiglini, V.; et al. ACS Comb. Sci. (ex J. Comb. Chem.) 2012, 14, 258-62. 3) Schaeffert, D. H., Okholm, A. H., Sørensen, R. S., Nielsen, J. S., Tørring, T. Rosen, C. B., Kodal, A. L. B., Mortensen, M. R., Gothelf, K. V. and Kjems, J. Intracellular delivery of a planar DNA origami structure by the transferrin-receptor internalization pathway. Small 12, 2634-2640 (2016) 4) Kodal*, A. L. B., Rosen*, C. B., Mortensen, M. M., Tørring, T. and Gothelf, K. V. DNA-templated introduction of an aldehyde handle in proteins. Chembiochem, 17, 1338-1342 (2016)

17 CP03 CP04 ANTIMALARIAL DRUG DISCOVERY: EXPLORING THE MEP DRUGGING THE UNDRUGGABLE: TARGETING CHALLENGING E3 PATHWAY LIGASES

Kathryn Price (1), Christopher Armstrong (2), Leah Imlay (2), Dana Hodge (2), C Pidathala (1), Galdeano Carles (1,2) Alexandre Lawrenson (1), Neil Berry (1), Paul O'Neill (1), Audrey Odom (2) 1) Institut de Biomedicina de la Universitat de Barcelona (IBUB) and Departament de Fisicoquímica, Facultat de Farmàcia, 1) The University of Liverpool, Department of Chemistry, Liverpool, L69 7ZD, UK Universitat de Barcelona, Av. Joan XXIII s/n, 08028 Barcelona, Spain 2) Washington University School of Medicine, St Louis, MO 63110, USA 2) 2 Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dow Street, Dundee, DD1 5EH, Scotland, UK

The development of effective antimalarial chemotherapeutics remains one of the major challenges within drug discovery. Malaria is still a major threat to global health with an estimated 198 million cases and 584 thousand Despite a clear opportunity for the development of a new class of therapeutic agents, the scientific community fatalities resulting from malaria infection in 2013.1 Despite a 30% decreased global incidence of malaria has been slow to invest significant resources and efforts on targeting components of the Ubiquitin-Proteasome between 2000 and 2013 through upscaling of malaria interventions, it is the adept ability of the Plasmodium System (UPS) and this field is still in its infancy. This has been due to the historical lack of foundational tools parasite to develop drug resistance which is so significantly hindering the fight against this entirely preventable and approaches and the significant complexity inherent in this biological system. disease. With 91 countries registering malaria transmission in 2014, the development of new antimalarial drugs Protein ubiquitination occurs through a cascade of enzymatic reactions, involving an E1 ubiquitin-activating and battle to overcome parasite resistance has never been more crucial. enzyme, an E2 ubiquitin-conjugating enzyme and an E3 ubiquitin ligase. E3 ubiquitin ligases confer substrate Through the intensified pursuit of new antimalarial agents and novel drug targets, the spotlight has fallen on the specificity to protein ubiquitination pathways, and are very attractive drug targets for therapeutic intervention, chemical manipulation of isoprenoid . Isoprenoids represent the oldest and largest class of natural even more in the view of the relevance that personalised medicine will have in the near future. However, to date, products which play vital roles in many cellular functions.2 In contrast to mammals; malaria-causing efforts to target and validate E3 ligases using small-molecules have been rewarded with limited success. 3 Plasmodium parasites use the non-mevalonate (MEP) biosynthetic pathway to generate isoprenoid precursors. In this talk, I will elaborate my adventures (and disadventures) developing chemical probes to unlock and Such biological selectivity makes the MEP pathway highly attractive as an antimalarial chemotherapeutic target. validate two enticing, challenging and medically relevant E3 protein ligases, the VHL and the FBW7 E3 ligases, Fosmidomycin, currently the most potent and best-characterised inhibitor of the MEP pathway, targeting the as a potential therapeutic target. VHL recruits for degradation the crucial transcription factor in hypoxia, 4 enzyme IspC, has validated the pathway as a viable target for antimalarial drug discovery and led to our HIF-1alpha. FBW7 is one of the most commonly deregulated UPS proteins in human cancers, which targets a objective to deliver a lead candidate molecule suitable for clinical development, targeting P.falciparum IspD (Pf range of substrates for degradation, including some key human oncoproteins including cyclin-E, MYC, Notch IspD). We seek to answer the question: Can arresting the MEP pathway through PfIspD inhibition cause and Junk. parasite death via disruption of isoprenoid precursor generation? Following a high throughput screen identifying the 1,2-benzoisothiazolone (BITZ) chemotype as a promising Pf IspD inhibitor, with the hit compound displaying an inhibitory IC50 of 484nM5, organic synthesis has explored structural modifications to the side chain of the BITZ core. Resulting inhibitors have been used to develop structure-activity relationship (SAR) around the PfIspD active site. Molecular modelling and machine learning techniques have been used to propose a mechanism of enzyme inhibition and guide the selection of new inhibitor analogues through modelling predictions. Molecular and techniques have been employed to study the effect of the BITZ inhibitors on the IspD enzyme and MEP pathway. Enzymatic and whole cell assays have been performed to determine inhibitory activity at PfIspD and the whole cell; site directed mutagenesis has assessed mutation effects on the behaviour and activity of inhibitors and fluorescent microscopy has evaluated localisation of the BITZ motif within the cell and its effects on morphology. This work has resulted in the generation of the three most potent PfIspD inhibitors to date, exhibiting enzymatic inhibition of 0.07-0.27μM, alongside impressive whole cell activity of 0.6-1.0μM. Site directed mutagenesis and structural manipulations have confirmed a covalent interaction between the BITZ core and cysteine residue in the PfIspD active site essential in achieving enzyme inhibition. We seek to progress the BITZ inhibitor series into lead optimisation, enhancing pharmacokinetics and ultimately generate an optimised lead targeting PfIspD, progressing the development of novel antimalarial agents. Alongside this, we are also exploring a second chemical series of non-covalent PfIspD inhibitors, based around the tetrahydro-β-carboline chemotype, identified in a phenotypic screen by GSK and made available to researchers by the Medicines for Malaria Venture (MMV) as part of the Malaria Box. Early analogues of this series display low nanomolar activity at Pf IspD and provide a contrasting chemical construct, SAR and mechanism of inhibition to the BITZ analogues explored to date.6

References 1) World Health Organization, World Malaria Report, 2014. 2) W. N. Hunter, J Biol Chem, 2007, 282, 21573-21577. 3) B. M. Lange, T. Rujan, W. Martin and R. Croteau, P Natl Acad Sci USA, 2000, 97, 13172-13177. 4) A. R. Odom and W. C. Van Voorhis, Mol Biochem Parasit, 2010, 170, 108-111. 5) A. S. Lawrenson and N. G. Berry, University of Liverpool, 2012. 6) L. S. Imlay et al., ACS Infect. Dis. 2015, 1, 157–167.

18 CP03 CP04 ANTIMALARIAL DRUG DISCOVERY: EXPLORING THE MEP DRUGGING THE UNDRUGGABLE: TARGETING CHALLENGING E3 PATHWAY LIGASES

Kathryn Price (1), Christopher Armstrong (2), Leah Imlay (2), Dana Hodge (2), C Pidathala (1), Galdeano Carles (1,2) Alexandre Lawrenson (1), Neil Berry (1), Paul O'Neill (1), Audrey Odom (2) 1) Institut de Biomedicina de la Universitat de Barcelona (IBUB) and Departament de Fisicoquímica, Facultat de Farmàcia, 1) The University of Liverpool, Department of Chemistry, Liverpool, L69 7ZD, UK Universitat de Barcelona, Av. Joan XXIII s/n, 08028 Barcelona, Spain 2) Washington University School of Medicine, St Louis, MO 63110, USA 2) 2 Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dow Street, Dundee, DD1 5EH, Scotland, UK

The development of effective antimalarial chemotherapeutics remains one of the major challenges within drug discovery. Malaria is still a major threat to global health with an estimated 198 million cases and 584 thousand Despite a clear opportunity for the development of a new class of therapeutic agents, the scientific community fatalities resulting from malaria infection in 2013.1 Despite a 30% decreased global incidence of malaria has been slow to invest significant resources and efforts on targeting components of the Ubiquitin-Proteasome between 2000 and 2013 through upscaling of malaria interventions, it is the adept ability of the Plasmodium System (UPS) and this field is still in its infancy. This has been due to the historical lack of foundational tools parasite to develop drug resistance which is so significantly hindering the fight against this entirely preventable and approaches and the significant complexity inherent in this biological system. disease. With 91 countries registering malaria transmission in 2014, the development of new antimalarial drugs Protein ubiquitination occurs through a cascade of enzymatic reactions, involving an E1 ubiquitin-activating and battle to overcome parasite resistance has never been more crucial. enzyme, an E2 ubiquitin-conjugating enzyme and an E3 ubiquitin ligase. E3 ubiquitin ligases confer substrate Through the intensified pursuit of new antimalarial agents and novel drug targets, the spotlight has fallen on the specificity to protein ubiquitination pathways, and are very attractive drug targets for therapeutic intervention, chemical manipulation of isoprenoid biosynthesis. Isoprenoids represent the oldest and largest class of natural even more in the view of the relevance that personalised medicine will have in the near future. However, to date, products which play vital roles in many cellular functions.2 In contrast to mammals; malaria-causing efforts to target and validate E3 ligases using small-molecules have been rewarded with limited success. 3 Plasmodium parasites use the non-mevalonate (MEP) biosynthetic pathway to generate isoprenoid precursors. In this talk, I will elaborate my adventures (and disadventures) developing chemical probes to unlock and Such biological selectivity makes the MEP pathway highly attractive as an antimalarial chemotherapeutic target. validate two enticing, challenging and medically relevant E3 protein ligases, the VHL and the FBW7 E3 ligases, Fosmidomycin, currently the most potent and best-characterised inhibitor of the MEP pathway, targeting the as a potential therapeutic target. VHL recruits for degradation the crucial transcription factor in hypoxia, 4 enzyme IspC, has validated the pathway as a viable target for antimalarial drug discovery and led to our HIF-1alpha. FBW7 is one of the most commonly deregulated UPS proteins in human cancers, which targets a objective to deliver a lead candidate molecule suitable for clinical development, targeting P.falciparum IspD (Pf range of substrates for degradation, including some key human oncoproteins including cyclin-E, MYC, Notch IspD). We seek to answer the question: Can arresting the MEP pathway through PfIspD inhibition cause and Junk. parasite death via disruption of isoprenoid precursor generation? Following a high throughput screen identifying the 1,2-benzoisothiazolone (BITZ) chemotype as a promising Pf IspD inhibitor, with the hit compound displaying an inhibitory IC50 of 484nM5, organic synthesis has explored structural modifications to the side chain of the BITZ core. Resulting inhibitors have been used to develop structure-activity relationship (SAR) around the PfIspD active site. Molecular modelling and machine learning techniques have been used to propose a mechanism of enzyme inhibition and guide the selection of new inhibitor analogues through modelling predictions. Molecular and chemical biology techniques have been employed to study the effect of the BITZ inhibitors on the IspD enzyme and MEP pathway. Enzymatic and whole cell assays have been performed to determine inhibitory activity at PfIspD and the whole cell; site directed mutagenesis has assessed mutation effects on the behaviour and activity of inhibitors and fluorescent microscopy has evaluated localisation of the BITZ motif within the cell and its effects on morphology. This work has resulted in the generation of the three most potent PfIspD inhibitors to date, exhibiting enzymatic inhibition of 0.07-0.27μM, alongside impressive whole cell activity of 0.6-1.0μM. Site directed mutagenesis and structural manipulations have confirmed a covalent interaction between the BITZ core and cysteine residue in the PfIspD active site essential in achieving enzyme inhibition. We seek to progress the BITZ inhibitor series into lead optimisation, enhancing pharmacokinetics and ultimately generate an optimised lead targeting PfIspD, progressing the development of novel antimalarial agents. Alongside this, we are also exploring a second chemical series of non-covalent PfIspD inhibitors, based around the tetrahydro-β-carboline chemotype, identified in a phenotypic screen by GSK and made available to researchers by the Medicines for Malaria Venture (MMV) as part of the Malaria Box. Early analogues of this series display low nanomolar activity at Pf IspD and provide a contrasting chemical construct, SAR and mechanism of inhibition to the BITZ analogues explored to date.6

References 1) World Health Organization, World Malaria Report, 2014. 2) W. N. Hunter, J Biol Chem, 2007, 282, 21573-21577. 3) B. M. Lange, T. Rujan, W. Martin and R. Croteau, P Natl Acad Sci USA, 2000, 97, 13172-13177. 4) A. R. Odom and W. C. Van Voorhis, Mol Biochem Parasit, 2010, 170, 108-111. 5) A. S. Lawrenson and N. G. Berry, University of Liverpool, 2012. 6) L. S. Imlay et al., ACS Infect. Dis. 2015, 1, 157–167.

19 CP05 CP06 DEVELOPMENT OF HIGHLY SELECTIVE AND REVERSIBLE FUNCTIONAL LIPIDOMICS-BASED LEAD DISCOVERY REVEALS DIACYLGLYCEROL LIPASE INHIBITORS NOVEL ROLE OF PHOSPHOLIPIDS IN VITAMIN A SIGNALING

Freek Janssen (1), Marc Baggelaar (1), Ming Jiang (1), Herman Overkleeft (2), Mario van der Stelt (1) Andreas Koeberle

1) Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden, The Netherlands Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University, 07743 Jena, Germany 2) Department of Bio-organic Synthesis, Leiden Institute of Chemistry, Leiden, The Netherlands

Functional lipidomics combines comprehensive lipid profiling with target identification and lead discovery to Diacylglycerol lipase (DAGL)-α and -β are enzymes responsible for the biosynthesis of the endocannabinoid reveal novel strategies for pharmacological intervention. Using this approach, we identified phosphatidylcholines 2-arachidonoylglycerol (2-AG). Selective and reversible inhibitors are required to study the exact function of with polyunsaturated fatty acids (PUFA-PC) that oscillate during the cell cycle. PUFA-PC suppresses cell DAGLs in neuronal cells, however they are not available to date. Previously we identified LEI104, which proliferation by inhibiting membrane binding and thus activation of the survival kinase Akt [1] and are 1 belongs to the class of α-ketoheterocycles, as the first reversible inhibitor for DAGL-α. LEI104 was, however, apparently part of a feed-forward mechanism of apoptosis. By screening a library of nutrients, natural products weakly active in a cellular assay and was not selective over FAAH, the enzyme responsible for the metabolism and drugs for selective effects on the phospholipidome and lipid mediator production, we identified PUFA-PC as of another endocannabinoid, anandamide. Here, we present a full account of our hit optimization efforts, critical mediator for the long-term regulation of Akt by vitamin A. The pleiotropic effects of retinol (vitamin A) 2,3 including screening of a 1000+ membered α-keto heterocycles library. We developed FJ609, which displayed on adult physiology and embryonic development are mediated through the active metabolite all-trans retinoic similar lipophilicity and was ~60-fold more potent compared to LEI104. Importantly, FJ609 was also highly acid (RA). Bound to retinoic acid receptors (RARs), RA controls transcription but also fine-tunes the expression 4 selective over FAAH. It is envisioned that the α-ketoheterocycles class of DAGL inhibitors will provide an of RA target genes by activating kinases such as Akt. The mechanisms for long-term regulation of Akt by excellent chemical series to develop small molecule therapies for diseases, such as obesity, related metabolic vitamin A are incompletely understood. Here, we show by lipidomic profiling that retinol and RA deplete disorders and neuroinflammation, in which excessive 2-AG signalling or its metabolites play a crucial role. NIH-3T3 fibroblasts from phosphatidylcholines (PC) with polyunsaturated fatty acids, in particular linoleic acid (18:2), and concomitantly induce long-term Akt activation. We demonstrate that the cellular ratio of 18:2-PC References determines the activation state of Akt, and ascribe the effects of vitamin A on lipid composition and Akt signaling to retinoic acid X receptor (RXR) activation by using selective agonists. Administration of vitamin A 1) Baggelaar, Janssen et al., (2013), Angew. Chem. Int. Ed., 52, 12081-12085 2) Baggelaar, Janssen et al., (2015), J. Am. Chem. Soc., 137, 8851-8857 to mice decreased 18:2-PC levels in brain (but not in other tissues) and in parallel enhanced basal Akt activation, 3) Janssen, Baggelaar et al., (2015), J. Med. Chem. 58, 9742-9753 which was attributed to astrocytes rather than to neurons in dissociated hippocampal cultures. Our study reveals 4) Janssen & Baggelaar et al., manuscript in preparation how vitamin A is likely to regulate long-term Akt signaling: binding to nuclear receptors, modulating the membrane lipid composition and subsequently enhancing Akt membrane translocation and thus activation. We anticipate this cascade to be key for brain homeostasis in light of the well-established roles of vitamin A, polyunsaturated fatty acids as well as Akt for brain physiology.

References 1) Koeberle et al., Proc. Natl. Acad. Sci. 2013, 110, 2546

20 CP05 CP06 DEVELOPMENT OF HIGHLY SELECTIVE AND REVERSIBLE FUNCTIONAL LIPIDOMICS-BASED LEAD DISCOVERY REVEALS DIACYLGLYCEROL LIPASE INHIBITORS NOVEL ROLE OF PHOSPHOLIPIDS IN VITAMIN A SIGNALING

Freek Janssen (1), Marc Baggelaar (1), Ming Jiang (1), Herman Overkleeft (2), Mario van der Stelt (1) Andreas Koeberle

1) Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden, The Netherlands Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University, 07743 Jena, Germany 2) Department of Bio-organic Synthesis, Leiden Institute of Chemistry, Leiden, The Netherlands

Functional lipidomics combines comprehensive lipid profiling with target identification and lead discovery to Diacylglycerol lipase (DAGL)-α and -β are enzymes responsible for the biosynthesis of the endocannabinoid reveal novel strategies for pharmacological intervention. Using this approach, we identified phosphatidylcholines 2-arachidonoylglycerol (2-AG). Selective and reversible inhibitors are required to study the exact function of with polyunsaturated fatty acids (PUFA-PC) that oscillate during the cell cycle. PUFA-PC suppresses cell DAGLs in neuronal cells, however they are not available to date. Previously we identified LEI104, which proliferation by inhibiting membrane binding and thus activation of the survival kinase Akt [1] and are 1 belongs to the class of α-ketoheterocycles, as the first reversible inhibitor for DAGL-α. LEI104 was, however, apparently part of a feed-forward mechanism of apoptosis. By screening a library of nutrients, natural products weakly active in a cellular assay and was not selective over FAAH, the enzyme responsible for the metabolism and drugs for selective effects on the phospholipidome and lipid mediator production, we identified PUFA-PC as of another endocannabinoid, anandamide. Here, we present a full account of our hit optimization efforts, critical mediator for the long-term regulation of Akt by vitamin A. The pleiotropic effects of retinol (vitamin A) 2,3 including screening of a 1000+ membered α-keto heterocycles library. We developed FJ609, which displayed on adult physiology and embryonic development are mediated through the active metabolite all-trans retinoic similar lipophilicity and was ~60-fold more potent compared to LEI104. Importantly, FJ609 was also highly acid (RA). Bound to retinoic acid receptors (RARs), RA controls transcription but also fine-tunes the expression 4 selective over FAAH. It is envisioned that the α-ketoheterocycles class of DAGL inhibitors will provide an of RA target genes by activating kinases such as Akt. The mechanisms for long-term regulation of Akt by excellent chemical series to develop small molecule therapies for diseases, such as obesity, related metabolic vitamin A are incompletely understood. Here, we show by lipidomic profiling that retinol and RA deplete disorders and neuroinflammation, in which excessive 2-AG signalling or its metabolites play a crucial role. NIH-3T3 fibroblasts from phosphatidylcholines (PC) with polyunsaturated fatty acids, in particular linoleic acid (18:2), and concomitantly induce long-term Akt activation. We demonstrate that the cellular ratio of 18:2-PC References determines the activation state of Akt, and ascribe the effects of vitamin A on lipid composition and Akt signaling to retinoic acid X receptor (RXR) activation by using selective agonists. Administration of vitamin A 1) Baggelaar, Janssen et al., (2013), Angew. Chem. Int. Ed., 52, 12081-12085 2) Baggelaar, Janssen et al., (2015), J. Am. Chem. Soc., 137, 8851-8857 to mice decreased 18:2-PC levels in brain (but not in other tissues) and in parallel enhanced basal Akt activation, 3) Janssen, Baggelaar et al., (2015), J. Med. Chem. 58, 9742-9753 which was attributed to astrocytes rather than to neurons in dissociated hippocampal cultures. Our study reveals 4) Janssen & Baggelaar et al., manuscript in preparation how vitamin A is likely to regulate long-term Akt signaling: binding to nuclear receptors, modulating the membrane lipid composition and subsequently enhancing Akt membrane translocation and thus activation. We anticipate this cascade to be key for brain homeostasis in light of the well-established roles of vitamin A, polyunsaturated fatty acids as well as Akt for brain physiology.

References 1) Koeberle et al., Proc. Natl. Acad. Sci. 2013, 110, 2546

21 CP07 CP08 DESIGN, SYNTHESIS AND BIOLOGICAL EVALUATION OF DESIGN AND SYNTHESIS OF 2,2'-DIINDOLYLMETHANES TO SELECTIVE CDK INHIBITORS BASED ON FLAVOPIRIDOL SELECTIVELY TARGET CERTAIN G-QUADRUPLEX DNA STRUCTURE STRUCTURES

Nikitia Mexia (1), Danai Spanou (1), Photini Tsiripillou (1), Sandrine Ruchaud (2), Nam Sangkil (3), Madeleine Livendahl (1), Jan Jamroskovic (2), Svetlana Ivanova (1), Peter Demirel (1), Nasim Sabouri (2), Nektarios Aligiannis (1), Nicolas Gaboriaud-Kolar (1), Alexios-Leandros Skaltsounis (1) Erik Chorell (1)

1) Department of Pharmacognosy and Natural Products Chemistry, Faculty of harmacy, University of Athens, 1) Department of Chemistry, Umeå University, Umeå, Sweden Panepistimiopolis, Athens, 15771, Greece 2) Department of Medical and Biophysics, Umeå University, Umeå, Sweden 2) USR3151 CNRS/UPMC, Plate-forme de Criblage KISSf (Kinase Inhibitor Specialized Screening Facility), Station Biologique de Roscoff, F-29688 Roscoff, France 3) Molecular Medicine, Beckman Research Institute, City of Hope Comprehensive Cancer Center, 1500 East Duarte Road, Duarte, CA 91010, USA G-quadruplexes (G4) are secondary DNA structures that are evolutionary conserved and enriched at e.g. telomeres, promotor regions, and ribosomal DNA. G4 DNA structures are important in many biological systems for example in genetic regulation, control of the latency of viruses as well as the protection of the telomere ends Flavopiridol, a hemisynthetic flavone inspired from the natural product rohitukine, is the first CDK inhibitor to from degrading. However, there are still big gaps in the knowledge of the biological interplay of these systems. enter clinical trials and has recently been granted an Orphan Drug designation for the treatment of patients with G4-stabilizing molecules are thus of interest both as chemical research tools and because they hold great Acute Myeloid Leukemia [1,2,3]. The unique structural characteristics of flavopiridol have exerted the interest therapeutic potential for the treatment of cancer and infectious diseases. We have developed a set of new for the application of several modifications during the past decades, aiming to ameliorate the compound’s G4-stabilizing ligands, to be used as chemical tools.1 Our main structure is inspired by one of the most biological properties. However, the synthesized derivatives had more than one features altered simultaneously frequently used G4-ligands, Phen-DC3.2 Our strategy was to replace the central phenantroline part in Phen-DC3 and they were less active than flavopiridol itself. This observation led us to design a series of analogues with with a 2,2’-diindolyl motif to increase the flexibility of the ligand. The ligands were first evaluated for their only one structural alteration each time, bearing either a differentiated alkaloid moiety or different substitution G4-binding properties through our in vitro assay based on Thioflavin T fluorescence.3 The assay showed a dose on the flavone’s aromatic ring. After dealing successfully with a demanding synthetic route for the first dependent binding with all of the synthesized ligands. For three of the ligands, the binding was comparative to, analogues, we tried to improve some crucial steps of this synthesis, establishing thus a more efficient and less or even better than, the binding of Phen-DC3. Interestingly, the ligands showed selectivity between the different expensive pathway that we followed for the rest of the compounds. All the analogues, as well as some of the key G4 structures in this assay with up to 11-fold preference for one of the G4 structures compared to the others. The synthetic intermediates, have been evaluated for their ability to selectively inhibit Cyclin Dependent Kinases ligand binding has also been investigated using circular dichroism (CD), which showed a less intrusive binding among a panel of protein kinases. Most of the analogues were found to be potent and selective inhibitors of with our ligands when compared to the binding of Phen-DC3. Additionally, the ligands ability to stabilize G4 CDK9 whilst one inhibits selectively CDK2. Additionally, the same compounds have been evaluated for their structures was also investigated using CD, which confirmed their selectivity for certain G4 structures. cytotoxic activity on several cancer cell lines, including leukemia cells.

References 1) Naik et al. (1988) Tetrahedron 44, 7, 2081-2086 2) Senderowicz (1999) Investig. New Drugs 17, 313-320 3) OT News (2014) Oncol. Times 36, 10, 91

References 1) M. Livendahl, J. Jamroskovic, S. Ivanova, P. Demirel, N. Sabouri, E. Chorell, Chem. Eur. J. (minor revisions) 2016. 2) A. De Cian, E. DeLemos, J.-L. Mergny, M.-P. Teulade- Fichou, D. Monchaud J. Am. Chem. Soc. 2007, 129, 1856-1857. 3) M. Wallgren, J. B. Mohammad, K.-P. Yan, P. Pourbozorgi- Langroudi, M. Ebrahimi, N. Sabouri, Nucleic Acids Res. 2016, doi: 10.1093/nar/gkw349

22 CP07 CP08 DESIGN, SYNTHESIS AND BIOLOGICAL EVALUATION OF DESIGN AND SYNTHESIS OF 2,2'-DIINDOLYLMETHANES TO SELECTIVE CDK INHIBITORS BASED ON FLAVOPIRIDOL SELECTIVELY TARGET CERTAIN G-QUADRUPLEX DNA STRUCTURE STRUCTURES

Nikitia Mexia (1), Danai Spanou (1), Photini Tsiripillou (1), Sandrine Ruchaud (2), Nam Sangkil (3), Madeleine Livendahl (1), Jan Jamroskovic (2), Svetlana Ivanova (1), Peter Demirel (1), Nasim Sabouri (2), Nektarios Aligiannis (1), Nicolas Gaboriaud-Kolar (1), Alexios-Leandros Skaltsounis (1) Erik Chorell (1)

1) Department of Pharmacognosy and Natural Products Chemistry, Faculty of harmacy, University of Athens, 1) Department of Chemistry, Umeå University, Umeå, Sweden Panepistimiopolis, Athens, 15771, Greece 2) Department of Medical Biochemistry and Biophysics, Umeå University, Umeå, Sweden 2) USR3151 CNRS/UPMC, Plate-forme de Criblage KISSf (Kinase Inhibitor Specialized Screening Facility), Station Biologique de Roscoff, F-29688 Roscoff, France 3) Molecular Medicine, Beckman Research Institute, City of Hope Comprehensive Cancer Center, 1500 East Duarte Road, Duarte, CA 91010, USA G-quadruplexes (G4) are secondary DNA structures that are evolutionary conserved and enriched at e.g. telomeres, promotor regions, and ribosomal DNA. G4 DNA structures are important in many biological systems for example in genetic regulation, control of the latency of viruses as well as the protection of the telomere ends Flavopiridol, a hemisynthetic flavone inspired from the natural product rohitukine, is the first CDK inhibitor to from degrading. However, there are still big gaps in the knowledge of the biological interplay of these systems. enter clinical trials and has recently been granted an Orphan Drug designation for the treatment of patients with G4-stabilizing molecules are thus of interest both as chemical research tools and because they hold great Acute Myeloid Leukemia [1,2,3]. The unique structural characteristics of flavopiridol have exerted the interest therapeutic potential for the treatment of cancer and infectious diseases. We have developed a set of new for the application of several modifications during the past decades, aiming to ameliorate the compound’s G4-stabilizing ligands, to be used as chemical tools.1 Our main structure is inspired by one of the most biological properties. However, the synthesized derivatives had more than one features altered simultaneously frequently used G4-ligands, Phen-DC3.2 Our strategy was to replace the central phenantroline part in Phen-DC3 and they were less active than flavopiridol itself. This observation led us to design a series of analogues with with a 2,2’-diindolyl motif to increase the flexibility of the ligand. The ligands were first evaluated for their only one structural alteration each time, bearing either a differentiated alkaloid moiety or different substitution G4-binding properties through our in vitro assay based on Thioflavin T fluorescence.3 The assay showed a dose on the flavone’s aromatic ring. After dealing successfully with a demanding synthetic route for the first dependent binding with all of the synthesized ligands. For three of the ligands, the binding was comparative to, analogues, we tried to improve some crucial steps of this synthesis, establishing thus a more efficient and less or even better than, the binding of Phen-DC3. Interestingly, the ligands showed selectivity between the different expensive pathway that we followed for the rest of the compounds. All the analogues, as well as some of the key G4 structures in this assay with up to 11-fold preference for one of the G4 structures compared to the others. The synthetic intermediates, have been evaluated for their ability to selectively inhibit Cyclin Dependent Kinases ligand binding has also been investigated using circular dichroism (CD), which showed a less intrusive binding among a panel of protein kinases. Most of the analogues were found to be potent and selective inhibitors of with our ligands when compared to the binding of Phen-DC3. Additionally, the ligands ability to stabilize G4 CDK9 whilst one inhibits selectively CDK2. Additionally, the same compounds have been evaluated for their structures was also investigated using CD, which confirmed their selectivity for certain G4 structures. cytotoxic activity on several cancer cell lines, including leukemia cells.

References 1) Naik et al. (1988) Tetrahedron 44, 7, 2081-2086 2) Senderowicz (1999) Investig. New Drugs 17, 313-320 3) OT News (2014) Oncol. Times 36, 10, 91

References 1) M. Livendahl, J. Jamroskovic, S. Ivanova, P. Demirel, N. Sabouri, E. Chorell, Chem. Eur. J. (minor revisions) 2016. 2) A. De Cian, E. DeLemos, J.-L. Mergny, M.-P. Teulade- Fichou, D. Monchaud J. Am. Chem. Soc. 2007, 129, 1856-1857. 3) M. Wallgren, J. B. Mohammad, K.-P. Yan, P. Pourbozorgi- Langroudi, M. Ebrahimi, N. Sabouri, Nucleic Acids Res. 2016, doi: 10.1093/nar/gkw349

23 CP09 CP10 STABLE, EFFECTIVE, AND SELECTIVE ANTICANCER GLYCO-CONJUGATED PHOTOSENSITIZERS: FROM SYNTHESIS PHENOLATO TITANIUM(IV) COMPLEXES TO VALIDATION IN CANCER PHOTODYNAMIC THERAPY

Sigalit Meker, Edit Y. Tshuva Patrícia M. R. Pereira (1,2,3), Charles M. Drain (3,4), Rosa Fernandes (2,5,6), João P. C. Tomé (1,7)

The Institute of Chemistry, The Hebrew University of Jerusalem, Jerusalem 91904 Israel 1) QOPNA, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal. 2) IBILI, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal. 3) Department of Chemistry and Biochemistry, Hunter College and Graduate Center of the City University of New York (CUNY), 695 Park Avenue, New York, NY 10065, USA. 4) The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA. Cisplatin is a widely used platinum-based metallo-chemotherapeutic drug that is considered an efficient 5) CNC.IBILI, University of Coimbra, 3004-504 Coimbra, Portugal. treatment mainly for testicular and ovarian cancers. However, its narrow activity range and severe side effects 6) Center of Investigation in Environment, Genetics and Oncobiology, 3001-201 Coimbra, Portugal. 7) CQE, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal. trigger studies of other potent transition metal complexes. Titanium(IV) complexes based on Cp and diketonato ligands have been studied extensively as anticancer agents due to their high activity toward various cancer cells and reduced side effects; however, they have not yet been utilized in the clinic due to water instability and formation of unidentified aggregates in aquatic solutions. The combination of a photosensitizing molecule (Photosensitizer, PS) and light to induce toxicity by Reactive We introduced the highly cytotoxic salan titanium(IV) complexes, which exhibit slow and defined hydrolysis to Oxygen Species (ROS) production is very attractive in the treatment of cancer by Photodynamic Therapy form stable oxo-bridged ligand-bound polynuclear compounds. These polynuclear hydrolysis products were (PDT).Unfortunately, the PSs used in clinic for the treatment of cancer have low cancer selectivity and poor inactive when administered directly, due to solubility and cellular penetration limitations. Conversion of these water solubility. The success of new PDT agentsin cancer treatment requires a strong collaboration between polynuclear compounds into nanoparticles enabled overcoming these limitations and thus such nanoformulated for the synthesis of the porphyrinoid molecule and conjugation with a cancer-targeting motif, physics complexes demonstrated cytotoxicity toward human and murine cancer cells. Their activity indicates that inert for the development of appropriate light irradiation devices, biochemists for understanding cellular responses, compounds lacking labile ligands may act as active species, and that labile ligands are not a necessity for and clinicians for the clinical studies. biological activity. Recently, we have combined the knowledge of chemists and biochemists to develop new PSs by conjugating Herein we present the synthesis and characterization of rationally designed anticancer Ti(IV) complexes that them with specificgalactose motifs to be used in the treatment of cancer by PDT. These new glyco-PSs were lack labile ligands and bear bis- and tetrakis-phenolato hexadentate ligands. These complexes are highly potent designed for a specific target such asa galactose-binding protein overexpressed in cancer cells, and their anticancer agents and are extremely stable in aquatic solutions compared with other Ti(IV) complexes; in photophysical properties evaluated. We have tested these PSs in different biological models in order to validate particular they maintain their activity after long periods of exposure to aquatic solutions and blood serum, which their photodynamic potential. significantly contributes to their therapeutic potential. Their hydrolytic behaviour, anti-tumor activity in vivo and The aim of this communication is to provide a thorough overview of experimental approaches and methods by in vitro, and selectivity toward cancerous cells will be discussed, as well as preliminary mechanistic insights. which novel glyco-PSs can be tested in vitro for applications in PDT. We presentresults of(1) photo-physical/-chemical methods including ROS generation, photostability, fluorescence properties, (2) water solubility, (3)in vitro biochemical assays including interactions with specific proteins, PS uptake, intracellular localization, dark toxicity, (4) photodynamic assays using cancer cell lines growing in monolayers and as spheroids, (5)in vivo studies in xenograft tumor models and ex vivo studies in organotyopic cultures of tumors. Thanks are due to FCT/MEC for the financial support to QOPNA (FCTUID/QUI/00062/2013), IBILI (FCTUID/NEU/04539/2013) and CQE (FCTUID/QUI/0100/2013) research units, through national funds and where applicable cofinanced by the FEDER, within the PT2020 Partnership Agreement. This work was also supported by ACIMAGO (ref. 12/12). The authors also acknowledge FCT for the doctoral research grant SFRH/BD/85941/2012 (to PMRP).

References 1) Liu, F. T.; Rabinovich, G. A. Nat. Rev. Cancer 2005, 5, 29–41. 2) Singh, S.; Aggarwal, A.; Bhupathiraju, N. V. S. D. K.; Arianna, G.; Tiwari, K.; Drain, C. M. Chem. Rev. 2015, 115, 10261–10306. 3) Pereira, P. M. R.; Silva, S.; Cavaleiro, J. A. S.; Ribeiro, C. A. F.; Tomé, J. P. C.; Fernandes, R. PLoS One 2014, 9, e95529. 4) Silva, S.; Pereira, P. M. R.; Silva, P.; Paz, F. A. A.; Faustino, M. A. F.; Cavaleiro, J. A. S.; Tomé, J. P. C.; Chemical Communications2012, 48, 3608-3610. References 1) S. Meker; O. Braitbard; M. D. Hall; J. Hochman; E. Y. Tshuva; Chem – Eur. J. (2016), in press. Selected for front cover. 2) S. Meker; K. Margulis-Goshen; E. Weiss; S. Magdassi; E. Y. Tshuva; Angew. Chem. Int. Ed. (2012), 51, 10515; Selected for Back Cover. Selected as “Hot Paper”.

24 CP09 CP10 STABLE, EFFECTIVE, AND SELECTIVE ANTICANCER GLYCO-CONJUGATED PHOTOSENSITIZERS: FROM SYNTHESIS PHENOLATO TITANIUM(IV) COMPLEXES TO VALIDATION IN CANCER PHOTODYNAMIC THERAPY

Sigalit Meker, Edit Y. Tshuva Patrícia M. R. Pereira (1,2,3), Charles M. Drain (3,4), Rosa Fernandes (2,5,6), João P. C. Tomé (1,7)

The Institute of Chemistry, The Hebrew University of Jerusalem, Jerusalem 91904 Israel 1) QOPNA, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal. 2) IBILI, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal. 3) Department of Chemistry and Biochemistry, Hunter College and Graduate Center of the City University of New York (CUNY), 695 Park Avenue, New York, NY 10065, USA. 4) The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA. Cisplatin is a widely used platinum-based metallo-chemotherapeutic drug that is considered an efficient 5) CNC.IBILI, University of Coimbra, 3004-504 Coimbra, Portugal. treatment mainly for testicular and ovarian cancers. However, its narrow activity range and severe side effects 6) Center of Investigation in Environment, Genetics and Oncobiology, 3001-201 Coimbra, Portugal. 7) CQE, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal. trigger studies of other potent transition metal complexes. Titanium(IV) complexes based on Cp and diketonato ligands have been studied extensively as anticancer agents due to their high activity toward various cancer cells and reduced side effects; however, they have not yet been utilized in the clinic due to water instability and formation of unidentified aggregates in aquatic solutions. The combination of a photosensitizing molecule (Photosensitizer, PS) and light to induce toxicity by Reactive We introduced the highly cytotoxic salan titanium(IV) complexes, which exhibit slow and defined hydrolysis to Oxygen Species (ROS) production is very attractive in the treatment of cancer by Photodynamic Therapy form stable oxo-bridged ligand-bound polynuclear compounds. These polynuclear hydrolysis products were (PDT).Unfortunately, the PSs used in clinic for the treatment of cancer have low cancer selectivity and poor inactive when administered directly, due to solubility and cellular penetration limitations. Conversion of these water solubility. The success of new PDT agentsin cancer treatment requires a strong collaboration between polynuclear compounds into nanoparticles enabled overcoming these limitations and thus such nanoformulated chemists for the synthesis of the porphyrinoid molecule and conjugation with a cancer-targeting motif, physics complexes demonstrated cytotoxicity toward human and murine cancer cells. Their activity indicates that inert for the development of appropriate light irradiation devices, biochemists for understanding cellular responses, compounds lacking labile ligands may act as active species, and that labile ligands are not a necessity for and clinicians for the clinical studies. biological activity. Recently, we have combined the knowledge of chemists and biochemists to develop new PSs by conjugating Herein we present the synthesis and characterization of rationally designed anticancer Ti(IV) complexes that them with specificgalactose motifs to be used in the treatment of cancer by PDT. These new glyco-PSs were lack labile ligands and bear bis- and tetrakis-phenolato hexadentate ligands. These complexes are highly potent designed for a specific target such asa galactose-binding protein overexpressed in cancer cells, and their anticancer agents and are extremely stable in aquatic solutions compared with other Ti(IV) complexes; in photophysical properties evaluated. We have tested these PSs in different biological models in order to validate particular they maintain their activity after long periods of exposure to aquatic solutions and blood serum, which their photodynamic potential. significantly contributes to their therapeutic potential. Their hydrolytic behaviour, anti-tumor activity in vivo and The aim of this communication is to provide a thorough overview of experimental approaches and methods by in vitro, and selectivity toward cancerous cells will be discussed, as well as preliminary mechanistic insights. which novel glyco-PSs can be tested in vitro for applications in PDT. We presentresults of(1) photo-physical/-chemical methods including ROS generation, photostability, fluorescence properties, (2) water solubility, (3)in vitro biochemical assays including interactions with specific proteins, PS uptake, intracellular localization, dark toxicity, (4) photodynamic assays using cancer cell lines growing in monolayers and as spheroids, (5)in vivo studies in xenograft tumor models and ex vivo studies in organotyopic cultures of tumors. Thanks are due to FCT/MEC for the financial support to QOPNA (FCTUID/QUI/00062/2013), IBILI (FCTUID/NEU/04539/2013) and CQE (FCTUID/QUI/0100/2013) research units, through national funds and where applicable cofinanced by the FEDER, within the PT2020 Partnership Agreement. This work was also supported by ACIMAGO (ref. 12/12). The authors also acknowledge FCT for the doctoral research grant SFRH/BD/85941/2012 (to PMRP).

References 1) Liu, F. T.; Rabinovich, G. A. Nat. Rev. Cancer 2005, 5, 29–41. 2) Singh, S.; Aggarwal, A.; Bhupathiraju, N. V. S. D. K.; Arianna, G.; Tiwari, K.; Drain, C. M. Chem. Rev. 2015, 115, 10261–10306. 3) Pereira, P. M. R.; Silva, S.; Cavaleiro, J. A. S.; Ribeiro, C. A. F.; Tomé, J. P. C.; Fernandes, R. PLoS One 2014, 9, e95529. 4) Silva, S.; Pereira, P. M. R.; Silva, P.; Paz, F. A. A.; Faustino, M. A. F.; Cavaleiro, J. A. S.; Tomé, J. P. C.; Chemical Communications2012, 48, 3608-3610. References 1) S. Meker; O. Braitbard; M. D. Hall; J. Hochman; E. Y. Tshuva; Chem – Eur. J. (2016), in press. Selected for front cover. 2) S. Meker; K. Margulis-Goshen; E. Weiss; S. Magdassi; E. Y. Tshuva; Angew. Chem. Int. Ed. (2012), 51, 10515; Selected for Back Cover. Selected as “Hot Paper”.

25 CP11 CP12 NEW 3D-SCAFFOLDS FOR FRAGMENT-BASED LEAD DISCOVERY DISCOVERY OF NOVEL ATPase INHIBITORS OF DNA GYRASE AND (FBLD): EXPLORATION OF CHEMICAL SPACE USING TOPOISOMERASE IV STARTING FROM MARINE SPONGE FUNCTIONALIZED SPIROHYDANTOINS. ALKALOID OROIDIN

Hugues Prevet (1), Marion Flipo (1), Baptiste Villemagne (1), Asahi Kawana (1), Nicolas Renault (2), Tihomir Tomašič (1), Nace Zidar (1), Janez Ilaš (1), Päivi Tammela (2), Danijel Kikelj (1) Benoit Deprez (1), Nicolas Willand (1) 1) University of Ljubljana, Faculty of Pharmacy, 1000 Ljubljana, Slovenia 1) Univ. Lille, Inserm, Institut Pasteur of Lille, U1177 - Drugs and Molecules for living Systems, F-59000 Lille, France 2) University of Helsinki, Faculty of Pharmacy, Centre for Drug Research, FI-00014 Helsinki, Finland 2) Institut of Chimie Pharmaceutique Albert Lespagnol, Lille Inflammation Research International Center, UFR Pharmacie, F-59000 Lille, France

Bacterial DNA gyrase and topoisomerase IV (topoIV) are heterotetrameric proteins consisting of two GyrA or ParC subunits, which are involved in DNA transit, and two GyrB or ParE subunits containing the ATPase Fragment-based lead discovery (FBLD) has emerged as a powerful tool for the identification of small molecules domains, respectively. They are well-known and validated targets in the discovery of antibacterial drugs; that bind weakly but efficiently to biological targets. FBLD has already been successfully applied to a large set however, inhibitors of their ATP-binding subunits GyrB and ParE, have so far not reached clinical use. Growing (1, 2) of targets but a series of challenges remains to be overcome especially in the design of new chemical resistance against the fluoroquinolones, that target the GyrA/ParC subunits, limits their therapeutic potential and (3) libraries . This is indeed considered as a pivotal element in fragment-based lead discovery as many stimulates the search for novel inhibitor classes targeting the GyrB/ParE ATP-binding sites. Several recent commercially available fragment libraries still contain mostly planar aromatic scaffolds. This lack of geometrical publications and patent applications on bacterial DNA gyrase and topoIV inhibitors emphasize the attractiveness and chemical diversity jeopardizes the success in discovering hits for more challenging targets, such as of these two enzymes for antibacterial drug discovery.1 protein-protein interactions (4). Recently, we have identified that the 4,5-dibromo-1H-pyrrole-2-carboxamide moiety, which is present in Agelas In this work we developed the concept of privileged fragment based on a spirohydantoin motif as the minimal oroides marine sponge alkaloid oroidin, is an important moiety for binding to the hydrophobic pocket of the rigid central scaffold that can donate or accept H-bonds. The presence of a spiranic carbon atom allows to Escherichia coli GyrB ATP-binding site.2 Structure-based optimization of initial low micromolar hits based on provide rigidity and sphericity and to distribute pharmacophores in the three dimensions of space. We optimized the 4,5,6,7-tetrahydrobenzo[1,2-d]thiazole scaffold resulted in the low nanomolar E. coli and submicromolar (5) a microwave-assisted procedure for the rapid synthesis of a set of analogues . We then developed chemical Staphylococcus aureus DNA gyrase inhibitors possessing also micromolar inhibitory activity of E. coli and S. pathways that allowed the selective N-monoalkylations of the hydantoin ring to provide access to fragment-like aureus topoIV. Compounds also displayed modest antibacterial activity against Gram positive S. aureus and or more complex lead-like structures. Finally, a virtual focused library of 500 000 N-alkylated spirohydantoins Enterococcus faecalis, while they were found to be efflux pump substrates in E. coli, which most likely leads to was generated using 120 commercially available alkylating agents. Remarkably this set of compounds was able their inactivity against Gram negative bacterial strains.2 Replacement of the 4,5,6,7-tetrahydrobenzo[1,2-d in silico to fully cover the 3D chemical space. As a proof of concept, this library was screened on MDM2 and ]thiazole scaffold by the benzothiazole core resulted in almost equipotent E. coli DNA gyrase inhibitors; in silico hits were synthesized. The results of the screening will be discussed. however, some of the compounds possessed balanced dual DNA gyrase/topoIV inhibitory activities. High resolution co-crystal structure of the E. coli GyrB in complex with a benzothiazole inhibitor revealed details of its interactions within the ATP-binding site and provided basis for further structure-based optimization. In addition, starting from the 4,5,6,7-tetrahydrobenzo[1,2-d]thiazole series of potent E. coli DNA gyrase inhibitors, a new structural class of N-phenylpyrrolamides was designed and through several optimisation cycles resulted in the low nanomolar E. coli DNA gyrase inhibitors. The binding mode of this structural class to the ATP-binding site of E. coli GyrB was revealed by a high-resolution crystal structure of the enzyme in complex with a N-phenyl-4,5-dibromopyrrolamide-based inhibitor.3,4 Potent inhibition and observed antibacterial activity highlight these novel structural classes of DNA gyrase and topoIV inhibitors as promising starting points for structure-based design of inhibitors with improved antibacterial activity.

References 1) Bisacchi, G. S.; Manchester, J.I. A new-class antibacterial-almost. Lessons in drug discovery and development: A critical analysis of more than 50 years of effort toward ATPase inhibitors of DNA gyrase and topoisomerase IV. ACS Infect. Dis. References 2014, 1, 4-41. 2) Tomašič, T.; Katsamakas, S.; Hodnik, Ž.; Ilaš, J.; Brvar, M.; Šolmajer, T.; Montalvao, S.; Tammela, P.; Banjanac, M.; 1) Baker, M. Fragment-based lead discovery grows up. Nat. Rev. Drug Discov. 12, 5-7 (2013) Ergović, G.; Anderluh, M.; Mašič, L.P.; Kikelj, D. Discovery of 4,5,6,7-tetrahydrobenzo[1,2-d]thiazoles as novel DNA 2) Scott, D. E. et al. Fragments-based approaches in drug discovery and chemical biology. Biochemistry 51, 4990-5003 gyrase inhibitors targeting the ATP-binding site. J. Med. Chem. 2015, 58, 5501-5521. (2012) 3) Zidar, N.; Macut, H.; Tomašič, T.; Brvar, M.; Montalvao, S.; Tammela, P.; Šolmajer, T.; Mašič, L.P.; Ilaš, J.; Kikelj, D. 3) Murray, C. W. et al. Opportunity Knocks: Organic Chemistry for Fragment-Based Drug Discovery (FBDD). Angew. N-Phenyl-4,5-dibromopyrrolamides and N-phenylindolamides as ATP competitive DNA gyrase B inhibitors: Design, Chem. Int. Ed. 54, 2-7 (2015) synthesis and evaluation. J. Med. Chem. 2015, 58, 6179-6194. 4) Morley A. D. et al. Fragment-based hit identification: thinking in 3D. Drug Discov. Today. 18, 1221-1227 (2013) 4) Zidar, N.; Tomašič, T.; Macut, H.; Sirc, A.; Brvar, M.; Montalvao, S.; Tammela, P.; Ilaš, J.; Kikelj, D. New 5) Prevet H. et al. Microwave-assisted synthesis of functionalized spirohydantoins as 3-D privileged fragments for scouting N-phenyl-4,5-dibromopyrrolamides and N-phenylindolamides as ATPase inhibitors of DNA gyrase. Eur. J. Med. Chem. the chemical space. Tet. Lett., doi:10.1016/j.tetlet.2016.05.065 2016, 117, 197-211.

26 CP11 CP12 NEW 3D-SCAFFOLDS FOR FRAGMENT-BASED LEAD DISCOVERY DISCOVERY OF NOVEL ATPase INHIBITORS OF DNA GYRASE AND (FBLD): EXPLORATION OF CHEMICAL SPACE USING TOPOISOMERASE IV STARTING FROM MARINE SPONGE FUNCTIONALIZED SPIROHYDANTOINS. ALKALOID OROIDIN

Hugues Prevet (1), Marion Flipo (1), Baptiste Villemagne (1), Asahi Kawana (1), Nicolas Renault (2), Tihomir Tomašič (1), Nace Zidar (1), Janez Ilaš (1), Päivi Tammela (2), Danijel Kikelj (1) Benoit Deprez (1), Nicolas Willand (1) 1) University of Ljubljana, Faculty of Pharmacy, 1000 Ljubljana, Slovenia 1) Univ. Lille, Inserm, Institut Pasteur of Lille, U1177 - Drugs and Molecules for living Systems, F-59000 Lille, France 2) University of Helsinki, Faculty of Pharmacy, Centre for Drug Research, FI-00014 Helsinki, Finland 2) Institut of Chimie Pharmaceutique Albert Lespagnol, Lille Inflammation Research International Center, UFR Pharmacie, F-59000 Lille, France

Bacterial DNA gyrase and topoisomerase IV (topoIV) are heterotetrameric proteins consisting of two GyrA or ParC subunits, which are involved in DNA transit, and two GyrB or ParE subunits containing the ATPase Fragment-based lead discovery (FBLD) has emerged as a powerful tool for the identification of small molecules domains, respectively. They are well-known and validated targets in the discovery of antibacterial drugs; that bind weakly but efficiently to biological targets. FBLD has already been successfully applied to a large set however, inhibitors of their ATP-binding subunits GyrB and ParE, have so far not reached clinical use. Growing (1, 2) of targets but a series of challenges remains to be overcome especially in the design of new chemical resistance against the fluoroquinolones, that target the GyrA/ParC subunits, limits their therapeutic potential and (3) libraries . This is indeed considered as a pivotal element in fragment-based lead discovery as many stimulates the search for novel inhibitor classes targeting the GyrB/ParE ATP-binding sites. Several recent commercially available fragment libraries still contain mostly planar aromatic scaffolds. This lack of geometrical publications and patent applications on bacterial DNA gyrase and topoIV inhibitors emphasize the attractiveness and chemical diversity jeopardizes the success in discovering hits for more challenging targets, such as of these two enzymes for antibacterial drug discovery.1 protein-protein interactions (4). Recently, we have identified that the 4,5-dibromo-1H-pyrrole-2-carboxamide moiety, which is present in Agelas In this work we developed the concept of privileged fragment based on a spirohydantoin motif as the minimal oroides marine sponge alkaloid oroidin, is an important moiety for binding to the hydrophobic pocket of the rigid central scaffold that can donate or accept H-bonds. The presence of a spiranic carbon atom allows to Escherichia coli GyrB ATP-binding site.2 Structure-based optimization of initial low micromolar hits based on provide rigidity and sphericity and to distribute pharmacophores in the three dimensions of space. We optimized the 4,5,6,7-tetrahydrobenzo[1,2-d]thiazole scaffold resulted in the low nanomolar E. coli and submicromolar (5) a microwave-assisted procedure for the rapid synthesis of a set of analogues . We then developed chemical Staphylococcus aureus DNA gyrase inhibitors possessing also micromolar inhibitory activity of E. coli and S. pathways that allowed the selective N-monoalkylations of the hydantoin ring to provide access to fragment-like aureus topoIV. Compounds also displayed modest antibacterial activity against Gram positive S. aureus and or more complex lead-like structures. Finally, a virtual focused library of 500 000 N-alkylated spirohydantoins Enterococcus faecalis, while they were found to be efflux pump substrates in E. coli, which most likely leads to was generated using 120 commercially available alkylating agents. Remarkably this set of compounds was able their inactivity against Gram negative bacterial strains.2 Replacement of the 4,5,6,7-tetrahydrobenzo[1,2-d in silico to fully cover the 3D chemical space. As a proof of concept, this library was screened on MDM2 and ]thiazole scaffold by the benzothiazole core resulted in almost equipotent E. coli DNA gyrase inhibitors; in silico hits were synthesized. The results of the screening will be discussed. however, some of the compounds possessed balanced dual DNA gyrase/topoIV inhibitory activities. High resolution co-crystal structure of the E. coli GyrB in complex with a benzothiazole inhibitor revealed details of its interactions within the ATP-binding site and provided basis for further structure-based optimization. In addition, starting from the 4,5,6,7-tetrahydrobenzo[1,2-d]thiazole series of potent E. coli DNA gyrase inhibitors, a new structural class of N-phenylpyrrolamides was designed and through several optimisation cycles resulted in the low nanomolar E. coli DNA gyrase inhibitors. The binding mode of this structural class to the ATP-binding site of E. coli GyrB was revealed by a high-resolution crystal structure of the enzyme in complex with a N-phenyl-4,5-dibromopyrrolamide-based inhibitor.3,4 Potent inhibition and observed antibacterial activity highlight these novel structural classes of DNA gyrase and topoIV inhibitors as promising starting points for structure-based design of inhibitors with improved antibacterial activity.

References 1) Bisacchi, G. S.; Manchester, J.I. A new-class antibacterial-almost. Lessons in drug discovery and development: A critical analysis of more than 50 years of effort toward ATPase inhibitors of DNA gyrase and topoisomerase IV. ACS Infect. Dis. References 2014, 1, 4-41. 2) Tomašič, T.; Katsamakas, S.; Hodnik, Ž.; Ilaš, J.; Brvar, M.; Šolmajer, T.; Montalvao, S.; Tammela, P.; Banjanac, M.; 1) Baker, M. Fragment-based lead discovery grows up. Nat. Rev. Drug Discov. 12, 5-7 (2013) Ergović, G.; Anderluh, M.; Mašič, L.P.; Kikelj, D. Discovery of 4,5,6,7-tetrahydrobenzo[1,2-d]thiazoles as novel DNA 2) Scott, D. E. et al. Fragments-based approaches in drug discovery and chemical biology. Biochemistry 51, 4990-5003 gyrase inhibitors targeting the ATP-binding site. J. Med. Chem. 2015, 58, 5501-5521. (2012) 3) Zidar, N.; Macut, H.; Tomašič, T.; Brvar, M.; Montalvao, S.; Tammela, P.; Šolmajer, T.; Mašič, L.P.; Ilaš, J.; Kikelj, D. 3) Murray, C. W. et al. Opportunity Knocks: Organic Chemistry for Fragment-Based Drug Discovery (FBDD). Angew. N-Phenyl-4,5-dibromopyrrolamides and N-phenylindolamides as ATP competitive DNA gyrase B inhibitors: Design, Chem. Int. Ed. 54, 2-7 (2015) synthesis and evaluation. J. Med. Chem. 2015, 58, 6179-6194. 4) Morley A. D. et al. Fragment-based hit identification: thinking in 3D. Drug Discov. Today. 18, 1221-1227 (2013) 4) Zidar, N.; Tomašič, T.; Macut, H.; Sirc, A.; Brvar, M.; Montalvao, S.; Tammela, P.; Ilaš, J.; Kikelj, D. New 5) Prevet H. et al. Microwave-assisted synthesis of functionalized spirohydantoins as 3-D privileged fragments for scouting N-phenyl-4,5-dibromopyrrolamides and N-phenylindolamides as ATPase inhibitors of DNA gyrase. Eur. J. Med. Chem. the chemical space. Tet. Lett., doi:10.1016/j.tetlet.2016.05.065 2016, 117, 197-211.

27 CP13 CP14 PURINE MIMICKING 3'-ETHYNYLRIBOFURANOSE NUCLEOSIDES: EXPERIMENTAL PLATFORM FOR CHARACTERIZATION AND NEW OPPORTUNITIES FOR AN OLD SUGAR MODIFICATION HIGH-THROUGHPUT SCREENING OF ACETYLCHOLINESTERASE INHIBITORS. Fabian Hulpia (1), Jan Balzarini (2), Dominique Schols (2), Serge Van Calenbergh (1) Piotr Draczkowski (1), Manuela Bartolini (2), Vincenza Andrisano (3), Dariusz Matosiuk (1), Krzysztof 1) Laboratory for Medicinal Chemistry, Ghent University, Ottergemsesteenweg 460, B-9000, Ghent, Belgium Jozwiak (1) 2) KU Leuven, Rega Institute for Medical Research, Minderbroedersstraat 10, B-3000, Leuven, Belgium 1) Faculty of Pharmacy, Medical University of Lublin, ul. W. Chodzki 4a, 20-093 Lublin, Poland 2) Alma Mater Studiorum University of Bologna, Department of Pharmacy and Biotechnology, Bologna, Italy 3) Alma Mater Studiorum University of Bologna, Department for Life Quality Studies, Rimini, Italy Nucleoside analogues can be considered as cornerstones in the treatment of cancer and viral infections, with research dating back more than 50 years. Despite this fact, agents with intriguing and improved efficacy, tolerability etc. have still been discovered over the past decade.1 Recently, research in our laboratory was initiated to re-evaluate certain nucleoside scaffolds and to uncover untapped potential. This presentation will Acetylcholinesterase (AChE) enzyme as a part of the cholinergic neuronal pathways is involved in learning and discuss a library subset comprising nucleosides that combine a fixed 3-ethynyl-D-ribofuranose moiety2 with memory. As such, AChE is utilized as a drug target in treatment of neurodegenerative diseases such as purine-mimicking pyrazoles3 as nucleobase surrogates. Alzheimer’s disease (AD). The disease is officially listed as the sixth-leading cause of death. Due to lack of effective therapy and demographic trends it is expected that the number of patients with AD will continue to The central ribofuranose moiety was prepared according to literature procedures. Glycosylation via a one-pot increase dramatically. Since the AChE inhibitors currently used in treatment of AD have limited effectiveness Vorbrüggen reaction,3 followed by deprotection and carboxamide formation gave the final products. Initial there is a great need for new therapeutics. This, however, creates a demand for techniques that will allow rapid screening of a small subset of derivatives delivered a low micromolar hit against RSV virus, with a good and effective selection of new drug candidates and facilitate the development of new drugs. Here we present selectivity index (>10). Follow-up series with a modified carboxamide moiety, additional C-4 & C-5 substitution novel experimental platform which combines affinity chromatography (AC) and isothermal titration calorimetry and the parent ribofuranose analogue were prepared and profiled. (ITC) for comprehensive characterization of new AChE inhibitors. A small library of purine-mimicking C-3’-ethynylribofuranose nucleosides was successfully synthesized via We propose the AC as a tool for rapid selection of drug candidates acting on AChE from large chemical Vorbrüggen glycosylation as the key step. Screening of the prepared analogues showed interesting activity libraries. For this, human recombinant AChE was immobilized on a chromatographic support, i.e. against RSV-virus. Preliminary structure-activity exploration of this series points to a unique antiviral profile of ethylenediamine monolithic convective interaction media (CIM) disk. The obtained micro-immobilized-enzyme our initial hit. reactor (IMER) was connected to a standard HPLC system. The system was validated and obtained results showed very good correlation with the data from conventional method. The optimized analysis protocols allowed for rapid on-line screening of AChE inhibitors, determination of their activity with simultaneous evaluation of their binding kinetics. Moreover, immobilized enzyme showed increased stability compared to the in-solution form. Consequently, assay costs were reduced and data reproducibility was increased. We manage also to reduce influence of any nonspecific interactions between the screened compounds and the chromatographic support. The described instrumental set-up can be easily automated making the method suitable for high-throughput screening. The ITC technique constitutes the second component of the platform, which facilitates further development of References the selected drug candidates. The ITC experimental protocols were tuned up for comprehensive and tailored 1) Jordheim, L. P.; Durantel, D.; Zoulim, F.; Dumontet, C. Nat. Rev. Drug Discov. 2013, 12, 447. characterization of AChE-inhibitor interaction, including determination of AChE kinetics, potency and affinity 2) Hattori, H.; Tanaka, M.; Fukushima, M.; Sasaki, T.; Matsuda, A. J. Med. Chem. 1996, 39, 5005. evaluation of AChE inhibitors, determination of their mechanism of action and stoichiometry of the interaction. 3) Manfredini, S.; Bazzanini, R.; Baraldi, P. G.; Guarneri, M.; Simoni, D.; Marongiu, M. E.; Pani, A.; La Colla, P.; Tramontano, E. J. Med. Chem. 1992, 35, 917. For the first time, we also reported thermodynamic parameters of AChE-inhibitor interaction, which are crucial for rational drug design and can be used as a predictor of the drug selectivity. The method allowed to study interactions in conditions resembling physiological ones with macromolecules in their native form. Since the method excluded the need for secondary reactions and protein chemical modifications (i.e. labelling) we manage to increase the reproducibility of the inhibitor potency assay and eliminate usual false positive results. The developed experimental protocols can be easily modified to study other inhibitor-enzyme systems and considered as a universal methodology. Enzymes are targets of 47% marketed small-molecule drugs. They also represent the second largest group of novel target structures. As such, enzymes appear to be one of the most pharmacologically important class of biomolecules. Therefore, obtained results besides application in AChE-inhibitor interaction studies can have a broader impact in the field of drug development.

28 CP13 CP14 PURINE MIMICKING 3'-ETHYNYLRIBOFURANOSE NUCLEOSIDES: EXPERIMENTAL PLATFORM FOR CHARACTERIZATION AND NEW OPPORTUNITIES FOR AN OLD SUGAR MODIFICATION HIGH-THROUGHPUT SCREENING OF ACETYLCHOLINESTERASE INHIBITORS. Fabian Hulpia (1), Jan Balzarini (2), Dominique Schols (2), Serge Van Calenbergh (1) Piotr Draczkowski (1), Manuela Bartolini (2), Vincenza Andrisano (3), Dariusz Matosiuk (1), Krzysztof 1) Laboratory for Medicinal Chemistry, Ghent University, Ottergemsesteenweg 460, B-9000, Ghent, Belgium Jozwiak (1) 2) KU Leuven, Rega Institute for Medical Research, Minderbroedersstraat 10, B-3000, Leuven, Belgium 1) Faculty of Pharmacy, Medical University of Lublin, ul. W. Chodzki 4a, 20-093 Lublin, Poland 2) Alma Mater Studiorum University of Bologna, Department of Pharmacy and Biotechnology, Bologna, Italy 3) Alma Mater Studiorum University of Bologna, Department for Life Quality Studies, Rimini, Italy Nucleoside analogues can be considered as cornerstones in the treatment of cancer and viral infections, with research dating back more than 50 years. Despite this fact, agents with intriguing and improved efficacy, tolerability etc. have still been discovered over the past decade.1 Recently, research in our laboratory was initiated to re-evaluate certain nucleoside scaffolds and to uncover untapped potential. This presentation will Acetylcholinesterase (AChE) enzyme as a part of the cholinergic neuronal pathways is involved in learning and discuss a library subset comprising nucleosides that combine a fixed 3-ethynyl-D-ribofuranose moiety2 with memory. As such, AChE is utilized as a drug target in treatment of neurodegenerative diseases such as purine-mimicking pyrazoles3 as nucleobase surrogates. Alzheimer’s disease (AD). The disease is officially listed as the sixth-leading cause of death. Due to lack of effective therapy and demographic trends it is expected that the number of patients with AD will continue to The central ribofuranose moiety was prepared according to literature procedures. Glycosylation via a one-pot increase dramatically. Since the AChE inhibitors currently used in treatment of AD have limited effectiveness Vorbrüggen reaction,3 followed by deprotection and carboxamide formation gave the final products. Initial there is a great need for new therapeutics. This, however, creates a demand for techniques that will allow rapid screening of a small subset of derivatives delivered a low micromolar hit against RSV virus, with a good and effective selection of new drug candidates and facilitate the development of new drugs. Here we present selectivity index (>10). Follow-up series with a modified carboxamide moiety, additional C-4 & C-5 substitution novel experimental platform which combines affinity chromatography (AC) and isothermal titration calorimetry and the parent ribofuranose analogue were prepared and profiled. (ITC) for comprehensive characterization of new AChE inhibitors. A small library of purine-mimicking C-3’-ethynylribofuranose nucleosides was successfully synthesized via We propose the AC as a tool for rapid selection of drug candidates acting on AChE from large chemical Vorbrüggen glycosylation as the key step. Screening of the prepared analogues showed interesting activity libraries. For this, human recombinant AChE was immobilized on a chromatographic support, i.e. against RSV-virus. Preliminary structure-activity exploration of this series points to a unique antiviral profile of ethylenediamine monolithic convective interaction media (CIM) disk. The obtained micro-immobilized-enzyme our initial hit. reactor (IMER) was connected to a standard HPLC system. The system was validated and obtained results showed very good correlation with the data from conventional method. The optimized analysis protocols allowed for rapid on-line screening of AChE inhibitors, determination of their activity with simultaneous evaluation of their binding kinetics. Moreover, immobilized enzyme showed increased stability compared to the in-solution form. Consequently, assay costs were reduced and data reproducibility was increased. We manage also to reduce influence of any nonspecific interactions between the screened compounds and the chromatographic support. The described instrumental set-up can be easily automated making the method suitable for high-throughput screening. The ITC technique constitutes the second component of the platform, which facilitates further development of References the selected drug candidates. The ITC experimental protocols were tuned up for comprehensive and tailored 1) Jordheim, L. P.; Durantel, D.; Zoulim, F.; Dumontet, C. Nat. Rev. Drug Discov. 2013, 12, 447. characterization of AChE-inhibitor interaction, including determination of AChE kinetics, potency and affinity 2) Hattori, H.; Tanaka, M.; Fukushima, M.; Sasaki, T.; Matsuda, A. J. Med. Chem. 1996, 39, 5005. evaluation of AChE inhibitors, determination of their mechanism of action and stoichiometry of the interaction. 3) Manfredini, S.; Bazzanini, R.; Baraldi, P. G.; Guarneri, M.; Simoni, D.; Marongiu, M. E.; Pani, A.; La Colla, P.; Tramontano, E. J. Med. Chem. 1992, 35, 917. For the first time, we also reported thermodynamic parameters of AChE-inhibitor interaction, which are crucial for rational drug design and can be used as a predictor of the drug selectivity. The method allowed to study interactions in conditions resembling physiological ones with macromolecules in their native form. Since the method excluded the need for secondary reactions and protein chemical modifications (i.e. labelling) we manage to increase the reproducibility of the inhibitor potency assay and eliminate usual false positive results. The developed experimental protocols can be easily modified to study other inhibitor-enzyme systems and considered as a universal methodology. Enzymes are targets of 47% marketed small-molecule drugs. They also represent the second largest group of novel target structures. As such, enzymes appear to be one of the most pharmacologically important class of biomolecules. Therefore, obtained results besides application in AChE-inhibitor interaction studies can have a broader impact in the field of drug development.

29 Notes

______

30 Oral Communications

31 OC01 OC02 DEVELOPMENT OF POTENT, CELL-ACTIVE INHIBITORS OF THE DISCOVERY OF CCT251921: A POTENT, SELECTIVE AND ORALLY MYCOBACTERIUM TUBERCULOSIS CHOLESTEROL OXIDASE BIOAVAILABLE SMALL MOLECULE MODULATOR OF THE CYP125 MEDIATOR COMPLEX-ASSOCIATED KINASES CDK8 AND CDK19

Madeline E. Kavanagh (1), Anthony G. Coyne (1), Sophie H. Gilbert (1), Cecilia Amadi (2), Kirsty J. Aurélie Mallinger (1), Kai Schiemann (2), Christian Rink (1), Frank Stieber (2), Michel Calderini (2), McLean (2), Helena Boshoff (3), Clifton Barry (3), Andrew W. Munro (2), (1) Mark Stubbs (1), Oliver Poeschke (2), Michael Busch (2), Paul Czodrowski (2), Djordje Musil (2), Daniel Schwarz (2), Maria-Jesus Ortiz-Ruiz (1), Richard Schneider (2), Melanie Valenti (1), Alexis de Haven 1) Department of Chemistry, University of Cambridge, UK Brandon (1), (1), Trevor Dale (3), Dirk Wienke (2), Paul Clarke (1), Christina Esdar (2), 2) Manchester Institute of Biotechnology, Faculty of Life Sciences, The University of Manchester, UK Florence Raynaud (1), Suzanne Eccles (1), Felix Rohdich (2), Julian Blagg (1) 3) Tuberculosis Research Section, National Institutes of Health, Bethesda, Maryland, USA 1) Cancer Research UK Cancer Therapeutics Unit at The Institute of Cancer Research, London, SW7 3RP 2) Merck KGaA, Darmstadt, Germany 3) School of Bioscience, Cardiff University, Cardiff, UK Tuberculosis is a re-emerging global epidemic. The development of antibiotic resistant strains of M. tuberculosis (Mtb) and co-morbidity of the disease with HIV/AIDS means that new drugs with novel mechanisms of action are urgently required.1 The unique ability of Mtb to utilise host cholesterol as a source of energy and biosynthetic 1 precursors contributes significantly to the pathogen’s survival. It is essential for the bacteria to infect host The Mediator complex-associated kinase CDK8 has been implicated in human disease, particularly in colorectal 2 macrophages, for long-term persistence and for the synthesis of virulence factors.2–4 Cholesterol oxidation is cancer where CDK8 has been reported as a putative oncogene. We previously reported the discovery of catalysed by the Mtb cytochrome P450 enzyme CYP125 and also the functionally redundant enzyme CYP142 in CCT251545, a potent, orally bioavailable small molecule inhibitor of Wnt signalling from a cell-based pathway 3 4 certain Mtb strains.5–7 Knockout of the genes involved in cholesterol catabolism attenuates bacterial growth, screen. We identified protein kinase paralogs CDK8 and CDK19 as the primary targets of this chemical series. results in the accumulation of toxic metabolic intermediates and increases the sensitivity of Mtb to killing with Protein X-ray crystallography studies of the chemical probe CCT251545 in complex with CDK8/cyclin C other antibiotics. CYP125/142 share low similarity with human P450 enzymes and thus, developing chemical revealed an unusual protein binding conformation invoking a C-terminal loop insertion into the ATP binding site inhibitors of these enzymes could provide a new therapeutic option for tuberculosis. and enabled the design of improved CDK8/19-selective compounds. We optimized the metabolic stability and aqueous solubility of the chemical probe CCT251545 in order to facilitate further in vivo evaluation of CDK8/19 To approach this aim, a focused library of heme binding fragments was compiled and screened against a panel of pharmacology and progression into preclinical in vivo studies. Here we describe the medicinal chemistry Mtb P450 enzymes. A common fragment hit was identified to bind to both CYP125 and CYP142 with optimisation leading to CCT251921, a potent, highly selective and orally bioavailable dual CDK8/19 ligand with micromolar affinity (KD CYP125 = 150 uM). An X-ray crystal structure of this benzyl pyridine fragment was excellent translation to cell-based activity and improved pharmacokinetic and pharmaceutical properties.5 obtained in complex with CYP142 and comparative structural analysis between CYP125 and CYP142 was used Demonstration of in vivo activity following oral dosing in solid human tumor xenograft model will also be to guide several rounds of synthetic elaboration to optimise affinity and selectivity. A 3000-fold improvement in shown. Scaffold hopping and biochemical HTS versus CDK8 led to additional compound series with a Type I binding affinity over the original fragment was achieved in a number of different chemical series. Parallel binding mode and resulted in structurally differentiated back-up candidate with equivalent pharmacological screening of the inhibitors established key structure activity relationships which were used to develop dual profile to CCT251921.6-8 inhibitors of CYP125 and CYP142, as well as potent compounds with selectivity for CYP125.

Biochemical assays demonstrate that the compounds efficiently inhibit the catalytic activity of CYP125 and References CYP142 in vitro and cellular studies indicate low micromolar MIC values against two different clinical strains of Mtb. 1) Porter, D.C.; Farmaki, E. et al. Proc. Natl. Acad. Sci. USA 2012, 109, 13799-13804 These inhibitors provide the tools necessary to interrogate the importance of cholesterol metabolism for 2) Firestein, R.; Bass, A. J. et al. Nature 2008, 455, 547-551 Mtb virulence and additional synthetic optimisation is in progress to further optimise their cellular activity. 3) Mallinger, A.; Crumpler, S et al. J. Med. Chem. 2015, 58, 1717-1735 4) Dale, T.; Clarke, P. A. et al. Nat. Chem. Biol. 2015, 11, 973-980 5) Mallinger, A.; Schiemann K. et al J. Med. Chem, 2016, 59, 1078-1101 6) Schiemann, K.; Mallinger, A. et al Bioorg. Med. Chem. Lett. 2016, 26, 1443–1451 7) Mallinger, A.; Schiemann, K. et al ACS Med. Chem. Lett. DOI: 10.1021/acsmedchemlett.6b00022 8) Manuscripts in preparation

References 1) World Health Organization. WHO Global Tuberculosis Report; 2015; (2) Pandey, A. K. and Sassetti, C. M. Proc. Natl. Acad. Sci. 2008, 105, 4376–4380; (3) Chang, J. C. et al., J. Bacteriol. 2009, 191, 5232–5239; (4) Ouellet, H. et al., Trends Microbiol. 2011, 19 (11), 530–539; (5) Capyk, J. K. et al., J. Biol. Chem. 2009, 284, 35534–35542; (6) McLean, K. J. et al., J. Biol. Chem. 2009, 284, 35524–35533; (7) Driscoll, M. D. et al., J. Biol. Chem. 2010, 285 (49), 38270–38282.

32 OC01 OC02 DEVELOPMENT OF POTENT, CELL-ACTIVE INHIBITORS OF THE DISCOVERY OF CCT251921: A POTENT, SELECTIVE AND ORALLY MYCOBACTERIUM TUBERCULOSIS CHOLESTEROL OXIDASE BIOAVAILABLE SMALL MOLECULE MODULATOR OF THE CYP125 MEDIATOR COMPLEX-ASSOCIATED KINASES CDK8 AND CDK19

Madeline E. Kavanagh (1), Anthony G. Coyne (1), Sophie H. Gilbert (1), Cecilia Amadi (2), Kirsty J. Aurélie Mallinger (1), Kai Schiemann (2), Christian Rink (1), Frank Stieber (2), Michel Calderini (2), McLean (2), Helena Boshoff (3), Clifton Barry (3), Andrew W. Munro (2), Chris Abell (1) Mark Stubbs (1), Oliver Poeschke (2), Michael Busch (2), Paul Czodrowski (2), Djordje Musil (2), Daniel Schwarz (2), Maria-Jesus Ortiz-Ruiz (1), Richard Schneider (2), Melanie Valenti (1), Alexis de Haven 1) Department of Chemistry, University of Cambridge, UK Brandon (1), Paul Workman (1), Trevor Dale (3), Dirk Wienke (2), Paul Clarke (1), Christina Esdar (2), 2) Manchester Institute of Biotechnology, Faculty of Life Sciences, The University of Manchester, UK Florence Raynaud (1), Suzanne Eccles (1), Felix Rohdich (2), Julian Blagg (1) 3) Tuberculosis Research Section, National Institutes of Health, Bethesda, Maryland, USA 1) Cancer Research UK Cancer Therapeutics Unit at The Institute of Cancer Research, London, SW7 3RP 2) Merck KGaA, Darmstadt, Germany 3) School of Bioscience, Cardiff University, Cardiff, UK Tuberculosis is a re-emerging global epidemic. The development of antibiotic resistant strains of M. tuberculosis (Mtb) and co-morbidity of the disease with HIV/AIDS means that new drugs with novel mechanisms of action are urgently required.1 The unique ability of Mtb to utilise host cholesterol as a source of energy and biosynthetic 1 precursors contributes significantly to the pathogen’s survival. It is essential for the bacteria to infect host The Mediator complex-associated kinase CDK8 has been implicated in human disease, particularly in colorectal 2 macrophages, for long-term persistence and for the synthesis of virulence factors.2–4 Cholesterol oxidation is cancer where CDK8 has been reported as a putative oncogene. We previously reported the discovery of catalysed by the Mtb cytochrome P450 enzyme CYP125 and also the functionally redundant enzyme CYP142 in CCT251545, a potent, orally bioavailable small molecule inhibitor of Wnt signalling from a cell-based pathway 3 4 certain Mtb strains.5–7 Knockout of the genes involved in cholesterol catabolism attenuates bacterial growth, screen. We identified protein kinase paralogs CDK8 and CDK19 as the primary targets of this chemical series. results in the accumulation of toxic metabolic intermediates and increases the sensitivity of Mtb to killing with Protein X-ray crystallography studies of the chemical probe CCT251545 in complex with CDK8/cyclin C other antibiotics. CYP125/142 share low similarity with human P450 enzymes and thus, developing chemical revealed an unusual protein binding conformation invoking a C-terminal loop insertion into the ATP binding site inhibitors of these enzymes could provide a new therapeutic option for tuberculosis. and enabled the design of improved CDK8/19-selective compounds. We optimized the metabolic stability and aqueous solubility of the chemical probe CCT251545 in order to facilitate further in vivo evaluation of CDK8/19 To approach this aim, a focused library of heme binding fragments was compiled and screened against a panel of pharmacology and progression into preclinical in vivo studies. Here we describe the medicinal chemistry Mtb P450 enzymes. A common fragment hit was identified to bind to both CYP125 and CYP142 with optimisation leading to CCT251921, a potent, highly selective and orally bioavailable dual CDK8/19 ligand with micromolar affinity (KD CYP125 = 150 uM). An X-ray crystal structure of this benzyl pyridine fragment was excellent translation to cell-based activity and improved pharmacokinetic and pharmaceutical properties.5 obtained in complex with CYP142 and comparative structural analysis between CYP125 and CYP142 was used Demonstration of in vivo activity following oral dosing in solid human tumor xenograft model will also be to guide several rounds of synthetic elaboration to optimise affinity and selectivity. A 3000-fold improvement in shown. Scaffold hopping and biochemical HTS versus CDK8 led to additional compound series with a Type I binding affinity over the original fragment was achieved in a number of different chemical series. Parallel binding mode and resulted in structurally differentiated back-up candidate with equivalent pharmacological screening of the inhibitors established key structure activity relationships which were used to develop dual profile to CCT251921.6-8 inhibitors of CYP125 and CYP142, as well as potent compounds with selectivity for CYP125.

Biochemical assays demonstrate that the compounds efficiently inhibit the catalytic activity of CYP125 and References CYP142 in vitro and cellular studies indicate low micromolar MIC values against two different clinical strains of Mtb. 1) Porter, D.C.; Farmaki, E. et al. Proc. Natl. Acad. Sci. USA 2012, 109, 13799-13804 These inhibitors provide the tools necessary to interrogate the importance of cholesterol metabolism for 2) Firestein, R.; Bass, A. J. et al. Nature 2008, 455, 547-551 Mtb virulence and additional synthetic optimisation is in progress to further optimise their cellular activity. 3) Mallinger, A.; Crumpler, S et al. J. Med. Chem. 2015, 58, 1717-1735 4) Dale, T.; Clarke, P. A. et al. Nat. Chem. Biol. 2015, 11, 973-980 5) Mallinger, A.; Schiemann K. et al J. Med. Chem, 2016, 59, 1078-1101 6) Schiemann, K.; Mallinger, A. et al Bioorg. Med. Chem. Lett. 2016, 26, 1443–1451 7) Mallinger, A.; Schiemann, K. et al ACS Med. Chem. Lett. DOI: 10.1021/acsmedchemlett.6b00022 8) Manuscripts in preparation

References 1) World Health Organization. WHO Global Tuberculosis Report; 2015; (2) Pandey, A. K. and Sassetti, C. M. Proc. Natl. Acad. Sci. 2008, 105, 4376–4380; (3) Chang, J. C. et al., J. Bacteriol. 2009, 191, 5232–5239; (4) Ouellet, H. et al., Trends Microbiol. 2011, 19 (11), 530–539; (5) Capyk, J. K. et al., J. Biol. Chem. 2009, 284, 35534–35542; (6) McLean, K. J. et al., J. Biol. Chem. 2009, 284, 35524–35533; (7) Driscoll, M. D. et al., J. Biol. Chem. 2010, 285 (49), 38270–38282.

33 OC03 OC04 HOW TO DESIGN CELL PERMEABLE NON-PEPTIDIC NOVEL DERIVATIVES OF THE TSPO LIGAND DPA-714 FOR IN MACROCYCLES VIVO PET IMAGING OF NEUROINFLAMMATION

Björn Over (1), Pär Matsson (3), Christian Tyrchan (1), Constanze Hilgendorf (1), Stephen E. Johnston Fanny Cacheux (1), Vincent Médran-Navarrete (1), Annelaure Damont (1), Géraldine Pottier (1), (2), Richard J. Lewis (1), Matthew W.D. Perry (1), Jeremy R. Duvall (2), Jan Kihlberg (3) Bertrand Kuhnast (1), Frank Marguet (2), Frédéric Puech (2), Raphaël Boisgard (1), Frédéric Dollé (1)

1) IMED Biotech Unit, AstraZeneca Gothenburg, Sweden 1) CEA, I2BM, Service Hospitalier Frédéric Joliot, Orsay, France. 2) Broad Institute, Cambridge, Massachusetts, United States 2) Sanofi, LGCR ChemBio, Chilly-Mazarin, France. 3) BMC, Uppsala University, Sweden

Objectives: The translocator protein 18 kDa (TSPO) is today a validated target for a number of therapeutic Macrocycles, drug design, cell permeability, efflux, stereospecific permeability prediction applications, but also a well-recognized diagnostic biomarker for related-disease state and progression, prompting the development of specific and dedicated ligands worldwide. The pyrazolo[1,5-a]pyrimidine acetamide core (Alpidem (Ananxyl®) azaisoster) is of particular interest, with notably the development of alkoxy- derivatives1, including DPA-7142, a fluorine-18-labelled Positron Emission Tomography (PET) probe of microglial activation and neuroinflammation linked to neurodegenerative diseases. Herein, the synthesis and in vitro characterisation of novel DPA-714 analogues – exploring the effects of alkyl substitutions at the acetamide moiety, together with the replacement of the oxygen atom bridging the phenyl ring of the core structure and the fluorinated moiety to prevent defluorination issues encountered in vivo3 – is reported. Fluorine-18-labelling of one candidate is also described as well as its preliminary in vivo evaluation as a PET imaging agent.

Methods: Fluorinated derivatives 3a-n were prepared from the key-ester 2 (synthesized from 1) and commercially available amines. In vitro TSPO binding affinities (Ki) were determined using membrane homogenates from rat heart and screening against [3H]PK11195. TSPO versus Central Receptor

(CBR) selectivities were determined using membrane homogenates from rat cerebral cortex and screening BACKGROUND: Macrocycles are of increasing interest as chemical probes and drugs for intractable targets like against [3H]flunitrazepam. Lipophilicities (LogD7.4) were determined using a validated, standardized HPLC protein-protein-interactions, but determinants of their cell permeability and oral bioavailability are poorly method. Metabolic stabilities were evaluated using hepatic microsomal fractions from different species (human, understood.1 rat and mouse) and data expressed as intrinsic clearances (InC). The tosyloxy derivative 5 was prepared from 4 (synthesized from 1 too). No-carrier-added radiofluorination of [18F]-3b was carried on using standard OBJECTIVE: We aim to create an understanding of the properties that govern cell permeability, efflux and conditions. PET-imaging studies were conducted on an AMPA rat model of neuroinflammation. solubility of macrocycles, to incorporate our learnings into guidelines describing an orally bioavailable property space for macrocycles.2 Results: Compounds 2 and 4 were obtained in 11 % and 23 % overall yields, respectively. Amides 3a-n were obtained from 2 in moderate to good yields (23 to 81 %), and high purities (> 95 %). Compound 5 was obtained METHODS: We generated an extensive dataset by measuring cell permeability (Caco-2) and efflux ratios as well from 4 in 50 % yield. With one exception, all derivatives displayed nanomolar to subnanomolar affinity for the as aqueous solubility, LogD7.4 and pKa values under consistent experimental conditions for >200, non-peptidic, TSPO and also high selectivity versus CBR (Ki(CBR)/Ki(TSPO) > 103). Among this series, four compounds (3c de novo-designed macrocycles from the Broad Institute’s diversity-oriented screening collection, including (R1,R2 = Me,Pr), 3d (R1,R2 = Pr,Pr), 3k (R1,R2 = Me,Bn), and 3b (R1,R2 = Et,Et) showed better Ki values structurally diverse sets of matched pairs of stereo- and regioisomers. (respectively 0.25, 0.26, 0.30, 0.37 nM) than that of DPA-714 (0.91 nM), and favorable lipophilicity for brain RESULTS: This analysis revealed how specific functional groups, substituents and molecular properties impact penetration (3.5 < logD7.4 < 4.4). Compound 3b and 3c also exhibited similar metabolic stability when compared cell permeability. Analysis of energy minimized structures for stereo- and regioisomeric sets, combined with to DPA-714 in mouse, rat and human microsomes (InC value of 241 and 321 µL/min/mg protein, respectively, NMR structure verification, provided fundamental insight into how, sometimes dynamic, intramolecular versus 68). Radiochemically pure (> 95 %) [18F]-3b was obtained in 50 ± 11 % radiochemical yields and with interactions in the 3D conformations of macrocycles are linked to physicochemical properties and permeability. specific activity as high as 75 GBq/µmol. PET demonstrated a marked contrast between the lesioned area and the corresponding area in the intact contralateral hemisphere of the rat brain after 60 min with calculated IMPACT: Combined use of quantitative structure-permeability modeling and the procedure for conformational ipsilateral-to-contralateral ratio for [18F]-3b of 3.57 ± 0.48, comparable to the one reported for [18F]DPA-714 analysis now, for the first time, provides chemists with a rational approach to design cell-permeable and orally (3.71 ± 0.39), reflecting a high in vivo specific binding for the TSPO. bioavailable non-peptidic macrocycles.3 Conclusion: Fourteen new derivatives of the TSPO ligand DPA-714 have been synthesized and in vitro characterized. One selected derivative has also been labelled with fluorine-18 and stands out as a promising References candidate for use as a PET probe for in vivo neuroinflammation imaging. 1) Doak, B.C., Over, B., Giordanetto, F., Kihlberg, J. “Extent of oral druggable space beyond the rule of 5: Learnings from drugs and clinical candidates”, Chemistry & Biology, 2014, 21, p. 1115-1142 Acknowledgments: INMiND (HEALTH-F2-2011-278850) and FLI (ANR-11-INBS-0006). 2) Matsson, P., Doak, B. C., Over, B. & Kihlberg, J. Cell permeability beyond the rule of 5. Advanced Drug Delivery Reviews 101, 42-61, (2016) 3) Over, B., et al. “Understanding macrocycle cell permeability.” 2016, manuscript under review References 1) Selleri S, et al. (2001), Bioorg. Med. Chem., 9, 2661 2) Damont A, et al. (2008), J. Label. Compds Radiopharm., 51, 286 3) Peyronneau M-A, et al. (2013), Drug. Metab. Dispos., 41, 122 34 OC03 OC04 HOW TO DESIGN CELL PERMEABLE NON-PEPTIDIC NOVEL DERIVATIVES OF THE TSPO LIGAND DPA-714 FOR IN MACROCYCLES VIVO PET IMAGING OF NEUROINFLAMMATION

Björn Over (1), Pär Matsson (3), Christian Tyrchan (1), Constanze Hilgendorf (1), Stephen E. Johnston Fanny Cacheux (1), Vincent Médran-Navarrete (1), Annelaure Damont (1), Géraldine Pottier (1), (2), Richard J. Lewis (1), Matthew W.D. Perry (1), Jeremy R. Duvall (2), Jan Kihlberg (3) Bertrand Kuhnast (1), Frank Marguet (2), Frédéric Puech (2), Raphaël Boisgard (1), Frédéric Dollé (1)

1) IMED Biotech Unit, AstraZeneca Gothenburg, Sweden 1) CEA, I2BM, Service Hospitalier Frédéric Joliot, Orsay, France. 2) Broad Institute, Cambridge, Massachusetts, United States 2) Sanofi, LGCR ChemBio, Chilly-Mazarin, France. 3) BMC, Uppsala University, Sweden

Objectives: The translocator protein 18 kDa (TSPO) is today a validated target for a number of therapeutic Macrocycles, drug design, cell permeability, efflux, stereospecific permeability prediction applications, but also a well-recognized diagnostic biomarker for related-disease state and progression, prompting the development of specific and dedicated ligands worldwide. The pyrazolo[1,5-a]pyrimidine acetamide core (Alpidem (Ananxyl®) azaisoster) is of particular interest, with notably the development of alkoxy- derivatives1, including DPA-7142, a fluorine-18-labelled Positron Emission Tomography (PET) probe of microglial activation and neuroinflammation linked to neurodegenerative diseases. Herein, the synthesis and in vitro characterisation of novel DPA-714 analogues – exploring the effects of alkyl substitutions at the acetamide moiety, together with the replacement of the oxygen atom bridging the phenyl ring of the core structure and the fluorinated moiety to prevent defluorination issues encountered in vivo3 – is reported. Fluorine-18-labelling of one candidate is also described as well as its preliminary in vivo evaluation as a PET imaging agent.

Methods: Fluorinated derivatives 3a-n were prepared from the key-ester 2 (synthesized from 1) and commercially available amines. In vitro TSPO binding affinities (Ki) were determined using membrane homogenates from rat heart and screening against [3H]PK11195. TSPO versus Central Benzodiazepine Receptor

(CBR) selectivities were determined using membrane homogenates from rat cerebral cortex and screening BACKGROUND: Macrocycles are of increasing interest as chemical probes and drugs for intractable targets like against [3H]flunitrazepam. Lipophilicities (LogD7.4) were determined using a validated, standardized HPLC protein-protein-interactions, but determinants of their cell permeability and oral bioavailability are poorly method. Metabolic stabilities were evaluated using hepatic microsomal fractions from different species (human, understood.1 rat and mouse) and data expressed as intrinsic clearances (InC). The tosyloxy derivative 5 was prepared from 4 (synthesized from 1 too). No-carrier-added radiofluorination of [18F]-3b was carried on using standard OBJECTIVE: We aim to create an understanding of the properties that govern cell permeability, efflux and conditions. PET-imaging studies were conducted on an AMPA rat model of neuroinflammation. solubility of macrocycles, to incorporate our learnings into guidelines describing an orally bioavailable property space for macrocycles.2 Results: Compounds 2 and 4 were obtained in 11 % and 23 % overall yields, respectively. Amides 3a-n were obtained from 2 in moderate to good yields (23 to 81 %), and high purities (> 95 %). Compound 5 was obtained METHODS: We generated an extensive dataset by measuring cell permeability (Caco-2) and efflux ratios as well from 4 in 50 % yield. With one exception, all derivatives displayed nanomolar to subnanomolar affinity for the as aqueous solubility, LogD7.4 and pKa values under consistent experimental conditions for >200, non-peptidic, TSPO and also high selectivity versus CBR (Ki(CBR)/Ki(TSPO) > 103). Among this series, four compounds (3c de novo-designed macrocycles from the Broad Institute’s diversity-oriented screening collection, including (R1,R2 = Me,Pr), 3d (R1,R2 = Pr,Pr), 3k (R1,R2 = Me,Bn), and 3b (R1,R2 = Et,Et) showed better Ki values structurally diverse sets of matched pairs of stereo- and regioisomers. (respectively 0.25, 0.26, 0.30, 0.37 nM) than that of DPA-714 (0.91 nM), and favorable lipophilicity for brain RESULTS: This analysis revealed how specific functional groups, substituents and molecular properties impact penetration (3.5 < logD7.4 < 4.4). Compound 3b and 3c also exhibited similar metabolic stability when compared cell permeability. Analysis of energy minimized structures for stereo- and regioisomeric sets, combined with to DPA-714 in mouse, rat and human microsomes (InC value of 241 and 321 µL/min/mg protein, respectively, NMR structure verification, provided fundamental insight into how, sometimes dynamic, intramolecular versus 68). Radiochemically pure (> 95 %) [18F]-3b was obtained in 50 ± 11 % radiochemical yields and with interactions in the 3D conformations of macrocycles are linked to physicochemical properties and permeability. specific activity as high as 75 GBq/µmol. PET demonstrated a marked contrast between the lesioned area and the corresponding area in the intact contralateral hemisphere of the rat brain after 60 min with calculated IMPACT: Combined use of quantitative structure-permeability modeling and the procedure for conformational ipsilateral-to-contralateral ratio for [18F]-3b of 3.57 ± 0.48, comparable to the one reported for [18F]DPA-714 analysis now, for the first time, provides chemists with a rational approach to design cell-permeable and orally (3.71 ± 0.39), reflecting a high in vivo specific binding for the TSPO. bioavailable non-peptidic macrocycles.3 Conclusion: Fourteen new derivatives of the TSPO ligand DPA-714 have been synthesized and in vitro characterized. One selected derivative has also been labelled with fluorine-18 and stands out as a promising References candidate for use as a PET probe for in vivo neuroinflammation imaging. 1) Doak, B.C., Over, B., Giordanetto, F., Kihlberg, J. “Extent of oral druggable space beyond the rule of 5: Learnings from drugs and clinical candidates”, Chemistry & Biology, 2014, 21, p. 1115-1142 Acknowledgments: INMiND (HEALTH-F2-2011-278850) and FLI (ANR-11-INBS-0006). 2) Matsson, P., Doak, B. C., Over, B. & Kihlberg, J. Cell permeability beyond the rule of 5. Advanced Drug Delivery Reviews 101, 42-61, (2016) 3) Over, B., et al. “Understanding macrocycle cell permeability.” 2016, manuscript under review References 1) Selleri S, et al. (2001), Bioorg. Med. Chem., 9, 2661 2) Damont A, et al. (2008), J. Label. Compds Radiopharm., 51, 286 3) Peyronneau M-A, et al. (2013), Drug. Metab. Dispos., 41, 122 35 Notes

______

36 Flash Poster Presentations

37 FP01 FP02 TOWARD LIBRARIES OF ARTIFICIAL MACROCYCLES DISCOVERY, STRUCTURAL EXPLORATION AND BIOLOGICAL PROFILING OF A NOVEL CLASS OF ANTIMYCOBACTERIAL Eman Abdelraheem (1,2), George Liao (1), M. Rudrakshula (1), Arianna Rossetti (1), Constantinos Neochoritis (1), Pravin Patil (1), Samad khaksar (1), Alexander Dömling (1), DPRE1 INHIBITORS

1) Department of Drug Design, University of Groningen, The Netherlands. Olga Balabon (1,2), Eleni Pitta (1,2), Maciej K. Rogacki (1,2), Fraser Cunningham (2), Jurgen Joossens 2) Chemistry Department, Faculty of Science, Sohag University, Sohag, 82524, Egypt (1), Koen Augustyns (1), Pieter Van der Veken (1), Robert Bates (2)

1) Medicinal chemistry laboratory, Department of Pharmaceutical Sciences, University of Antwerp, Universiteitsplein 1, B-2610 Wilrijk, Belgium 2) Diseases of the Developing World (DDW), Tres Cantos Medicines Development Campus (TCMDC), GlaxoSmithKline, Synthetic macrocycles have recently seen a renaissance in medicinal chemistry due to their unusual Severo Ochoa 2, 28760 Tres Cantos, Spain conformational, binding and transport properties, beyond the Ro5. Efficient synthetic access and virtual screening of macrocylic libraries, however, is a major challenge. A synthetic platform for the fast and efficient access to hundreds and virtually millions of macrocyclic scaffolds using multicomponent reaction chemistry (MCR) is presented [1]. Our group has developed a method that can use α-isocyano-ω-carboxylic acids in Tuberculosis (TB) remains a global health threat, accounting for more than 9 million new cases per year and 1,5 macrocycle synthesis via the Ugi reaction [2]. Also, we have provide a fast and general way to artificial million deaths.1 The emergence of multi- and extensively-drug resistant Mycobacterium tuberculosis (Mtb) macrocyclic depsipeptides using the Passerini reaction [3]. α-Amino-ω-carboxylic acids were synthesized from strains as well as HIV comorbidity fuel the TB epidemic resurgence. A pressing need for the discovery and cyclic anhydrides and diamines respectively, followed by Ugi-4CR using oxo components and isocyanides to development of new antitubercular agents that target new biochemical pathways and treat drug resistant forms of give a large library of macrocycle. The synthetic chemistry will be discussed and several virtual/synthetic the disease is undeniable. examples of macrocyclic compounds targeting protein protein interactions such as PD1-PDL1 will be presented. The presented work was performed in the frame of the OpenMedChem EID-ITN project (FP7) between the University of Antwerp and GlaxoSmithKline (GSK) with focus on early hit-to-lead anti-tubercular drug development. Here, we report a novel chemical series of antimycobacterials, discovered during a compound collection screening at GSK. Compounds in the series act through non-covalent inhibition of the essential and vulnerable flavo-enzyme deca-prenylphosphoryl-beta-D-ribose 2-epimerase (DprE1). We present Structure-Activity-Relationship (SAR) data based on a set of analogues around the initial hit. Evaluation data for the obtained compounds include whole cell MIC-values for the Mtb strain H37Rv and inhibitory potencies on MtbDprE1. Moreover, physicochemical profile, cytotoxicity (HepG2) and cardiotoxicity (hERG) are reported for these molecules. Activity against DprE1 was validated by the observed increase in MIC after testing the hit against an overexpressor strain. Overall, this novel series of DprE1 inhibitors contains highly active inhibitors with very good cellular potencies and balanced physicochemical profiles. Although no cytotoxic effects were found so far, appreciable hERG affinity is present for some analogues. Finally, the primary hit was tested against a panel of Gram-negative and Gram-positive pathogens indicating very selective antimycobacterial properties.

References World Health Organization Global tuberculosis report. WHO 2015. Kolly, G. S.; Boldrin, F.; Sala, C et al. Assessing the essentiality of the decaprenyl-phospho-d-arabinofuranose pathway in Mycobacterium tuberculosis using conditional mutants. Mol. Microbiol. 2014, 92, 194–211.

References

1) Domling, A.; Ugi, I. Angew. Chem. Int. Ed. Engl. 39, 3168−3210 (2000). 2) Liao, G. P.; Abdelraheem, E. M. M.; Neochoritis, C. G.; Kurpiewska, K.; Kalinowska-Tłuścik, J.; McGowan, D. C.; Domling, A. Org. Lett. 17, 4980−4983 (2015). 3) Abdelraheem, E. M. M.; Kurpiewska, K.; Kalinowska-Tłuścik, J.; Domling, A. Org. Lett. (2016), submitted

38 FP01 FP02 TOWARD LIBRARIES OF ARTIFICIAL MACROCYCLES DISCOVERY, STRUCTURAL EXPLORATION AND BIOLOGICAL PROFILING OF A NOVEL CLASS OF ANTIMYCOBACTERIAL Eman Abdelraheem (1,2), George Liao (1), M. Rudrakshula (1), Arianna Rossetti (1), Constantinos Neochoritis (1), Pravin Patil (1), Samad khaksar (1), Alexander Dömling (1), DPRE1 INHIBITORS

1) Department of Drug Design, University of Groningen, The Netherlands. Olga Balabon (1,2), Eleni Pitta (1,2), Maciej K. Rogacki (1,2), Fraser Cunningham (2), Jurgen Joossens 2) Chemistry Department, Faculty of Science, Sohag University, Sohag, 82524, Egypt (1), Koen Augustyns (1), Pieter Van der Veken (1), Robert Bates (2)

1) Medicinal chemistry laboratory, Department of Pharmaceutical Sciences, University of Antwerp, Universiteitsplein 1, B-2610 Wilrijk, Belgium 2) Diseases of the Developing World (DDW), Tres Cantos Medicines Development Campus (TCMDC), GlaxoSmithKline, Synthetic macrocycles have recently seen a renaissance in medicinal chemistry due to their unusual Severo Ochoa 2, 28760 Tres Cantos, Spain conformational, binding and transport properties, beyond the Ro5. Efficient synthetic access and virtual screening of macrocylic libraries, however, is a major challenge. A synthetic platform for the fast and efficient access to hundreds and virtually millions of macrocyclic scaffolds using multicomponent reaction chemistry (MCR) is presented [1]. Our group has developed a method that can use α-isocyano-ω-carboxylic acids in Tuberculosis (TB) remains a global health threat, accounting for more than 9 million new cases per year and 1,5 macrocycle synthesis via the Ugi reaction [2]. Also, we have provide a fast and general way to artificial million deaths.1 The emergence of multi- and extensively-drug resistant Mycobacterium tuberculosis (Mtb) macrocyclic depsipeptides using the Passerini reaction [3]. α-Amino-ω-carboxylic acids were synthesized from strains as well as HIV comorbidity fuel the TB epidemic resurgence. A pressing need for the discovery and cyclic anhydrides and diamines respectively, followed by Ugi-4CR using oxo components and isocyanides to development of new antitubercular agents that target new biochemical pathways and treat drug resistant forms of give a large library of macrocycle. The synthetic chemistry will be discussed and several virtual/synthetic the disease is undeniable. examples of macrocyclic compounds targeting protein protein interactions such as PD1-PDL1 will be presented. The presented work was performed in the frame of the OpenMedChem EID-ITN project (FP7) between the University of Antwerp and GlaxoSmithKline (GSK) with focus on early hit-to-lead anti-tubercular drug development. Here, we report a novel chemical series of antimycobacterials, discovered during a compound collection screening at GSK. Compounds in the series act through non-covalent inhibition of the essential and vulnerable flavo-enzyme deca-prenylphosphoryl-beta-D-ribose 2-epimerase (DprE1). We present Structure-Activity-Relationship (SAR) data based on a set of analogues around the initial hit. Evaluation data for the obtained compounds include whole cell MIC-values for the Mtb strain H37Rv and inhibitory potencies on MtbDprE1. Moreover, physicochemical profile, cytotoxicity (HepG2) and cardiotoxicity (hERG) are reported for these molecules. Activity against DprE1 was validated by the observed increase in MIC after testing the hit against an overexpressor strain. Overall, this novel series of DprE1 inhibitors contains highly active inhibitors with very good cellular potencies and balanced physicochemical profiles. Although no cytotoxic effects were found so far, appreciable hERG affinity is present for some analogues. Finally, the primary hit was tested against a panel of Gram-negative and Gram-positive pathogens indicating very selective antimycobacterial properties.

References World Health Organization Global tuberculosis report. WHO 2015. Kolly, G. S.; Boldrin, F.; Sala, C et al. Assessing the essentiality of the decaprenyl-phospho-d-arabinofuranose pathway in Mycobacterium tuberculosis using conditional mutants. Mol. Microbiol. 2014, 92, 194–211.

References

1) Domling, A.; Ugi, I. Angew. Chem. Int. Ed. Engl. 39, 3168−3210 (2000). 2) Liao, G. P.; Abdelraheem, E. M. M.; Neochoritis, C. G.; Kurpiewska, K.; Kalinowska-Tłuścik, J.; McGowan, D. C.; Domling, A. Org. Lett. 17, 4980−4983 (2015). 3) Abdelraheem, E. M. M.; Kurpiewska, K.; Kalinowska-Tłuścik, J.; Domling, A. Org. Lett. (2016), submitted

39 FP03 FP04 ORALLY BIOAVAILABLE ANTIMALARIAL 4(1H)-QUINOLONE DEVELOPEMENT OF SYNTHESIS METHODOLOGY FOR PRODRUGS WITH SINGLE-DOSE CURES DNA-ENCODED SMALL MOLECULE SCREENING LIBRARIES

Fabian Brockmeyer (1), Andrii Monastyrskyi (2), Alexis LaCrue (3), Tina Mutka (3), Dennis Kyle (3), Mateja Klika Skopic, Andreas Brunschweiger Roman Manetsch (1) TU Dortmund, Department for Chemistry and Chemical Biology, 1) Northeastern University, Department of Chemistry and Chemical Biology, Otto-Hahn-Str. 6, 44227 Dortmund, Germany Boston, MA, 02115, United States 2) University of South Florida, Department of Chemistry, Tampa, Florida 33620, United States 3) University of South Florida, Department of Global Health, Tampa, Florida 33612, United States The selection of DNA-encoded small molecule libraries (DELs) is an attractive technology for target-based identification of bioactive compounds.[1]DELs are synthesized through iterative, combinatorial organic preparative chemistry and enzymatic encoding steps. Thus, chemical reactions applied to DEL synthesis strictly need to be DNA-compatible. Synthetic transformations are currently restricted to mostly carbonyl and Malaria is estimated to have caused 438,000 deaths and 214 million cases of the disease globally in 2015. Four Pd-catalyzed C-C cross-coupling reactions. This restriction defines a challenge for organic chemists: strains of Plasmodium parasite cause malaria in humans and the disease is transferred by Anopheles mosquitos. Development of synthesis methodology for DELs is desperately needed to expand chemical space covered by Though mortality rates are down 47% globally since 2000 and significant progress has been made in the quest these libraries. We developed a solid support-based synthesis strategy that broadens the range of applicable for eradication, reported occurrences of resistance against current therapeutics threaten to reverse that progress. catalytic methods. [2] Among the catalytic systems that are now available for DEL synthesis are organocatalysts, Longstanding treatment chloroquine has seen resistance since the 1950’s, with resistance becoming widespread and transition metal ions such as Au(I), Ag(I), and Cu(I). Our strategy opened access to substituted and in the 70’s and 80’s. Artemisinin, the current main line of defense against malaria, is used in artemisinin functionalized heterocyclic scaffold structures as encodable DNA-conjugates from simple, readily available combination therapies (ACTs) in order to curtail resistance, though at last count, artemisinin resistant parasites starting materials. For example, application of transition metal catalysts furnished DNA-heterocycle conjugates have been reported in 5 countries of the Greater Mekong sub region. In order to curb further resistance, it is through [3+2] cycloaddition reactions. Some of the newly synthesized DNA-conjugated heterocycles display essential that new antimalarial compounds be brought through the pipeline. structural motifs from natural products, while others represent core structures of clinical candidates or approved drugs. All heterocyclic scaffolds enable subsequent DNA-encoded combinatorial library synthesis by For approximately half a century, 4(1H)-quinolones such as endochin or ICI 56,780 were known to be causal well-described, robust reactions. prophylactic and potent erythrocytic stage agents in avian but not in mammalian malaria models. Hit-to-lead optimization of endochin lead to 4(1H)-quinolones ELQ-300 and P4Q-391, which target the liver, the blood as well as the transmitting stages of the parasite. Despite entering preclinical development, ELQ-300 did not enter phase I trials due to limited aqueous solubility and high crystallinity.1 To overcome these limitations, we designed and developed a prodrug approach containing an amino group linked to the parent 4(1H)-quinolone by an acetal carbonate group. Different reaction conditions were found to attach the prodrug moiety selectively onto the oxygen or the nitrogen of the 4(1H)-quinolone scaffold. The resulting O-alkylated prodrugs P4Q-1290 and P4Q-1291 were profiled for physicochemical properties such as chemical stability and aqueous solubility. The prodrugs are stable at low pHs and start releasing the parent drug independently of any enzyme activity at a pH level of about 7. Furthermore, prodrugs P4Q-1290 and P4Q-1291 were highly efficacious in in vivo efficacy assays displaying single-dose cures at low doses. Scheme 1. Solid phase-based transition metal ion- and organocatalyzed synthesis of DNA-heterocycle conjugates serving as starting points for encoded library synthesis.

References 1) Salamon H, Klika Škopić M, Jung K, Bugain O, Brunschweiger A. ACS Chem Biol. 2016, 11, 296-307. 2) Brunschweiger A, Krause N, Antonchick A, Klika Skopic M, Salamon H, Bugain O, Jung K, Wagner B. Eur. Pat. Appl. 15202448.5.

The new discoveries are significant as mitochondrial inhibitors have the potential to advance the malaria elimination campaign by blocking parasite development in the blood and liver, as well as preventing transmission to mosquitoes.

References 1) A. A. Nilsen, A. LaCrue, K. L. White, I. P. Forquer, R. M. Cross, J. Marfurt, M. W. Mather, M. J. Delves, D. M. Shackleford, F. E. Sáenz, J. M. Morrisey, J. Steuten, T. Mutka, Y. Li, G. Wirjanata, E. Ryan, S. Duffy, J. X. Kelly, B. F. Sebayang, A.-M. Zeeman, R. Noviyanti, R. E. Sinden, C. H. M. Kocken, R. N. Price, V. M. Avery, I. Angulo-Barturen, M. B. JiménezDíaz, S. Ferrer, E. Herreros, L. M. Sanz, F. J. G. Benito, I. Bathurst, J. Burrows, P. Siegl, R. K. Guy, R. W. Winter, A. B. Vaidya, S. A. Charman, D. E. Kyle, R. Manetsch*, M. K. Riscoe*, Sci. Transl. Med., 2013, 5, 177ra37.

40 FP03 FP04 ORALLY BIOAVAILABLE ANTIMALARIAL 4(1H)-QUINOLONE DEVELOPEMENT OF SYNTHESIS METHODOLOGY FOR PRODRUGS WITH SINGLE-DOSE CURES DNA-ENCODED SMALL MOLECULE SCREENING LIBRARIES

Fabian Brockmeyer (1), Andrii Monastyrskyi (2), Alexis LaCrue (3), Tina Mutka (3), Dennis Kyle (3), Mateja Klika Skopic, Andreas Brunschweiger Roman Manetsch (1) TU Dortmund, Department for Chemistry and Chemical Biology, 1) Northeastern University, Department of Chemistry and Chemical Biology, Otto-Hahn-Str. 6, 44227 Dortmund, Germany Boston, MA, 02115, United States 2) University of South Florida, Department of Chemistry, Tampa, Florida 33620, United States 3) University of South Florida, Department of Global Health, Tampa, Florida 33612, United States The selection of DNA-encoded small molecule libraries (DELs) is an attractive technology for target-based identification of bioactive compounds.[1]DELs are synthesized through iterative, combinatorial organic preparative chemistry and enzymatic encoding steps. Thus, chemical reactions applied to DEL synthesis strictly need to be DNA-compatible. Synthetic transformations are currently restricted to mostly carbonyl and Malaria is estimated to have caused 438,000 deaths and 214 million cases of the disease globally in 2015. Four Pd-catalyzed C-C cross-coupling reactions. This restriction defines a challenge for organic chemists: strains of Plasmodium parasite cause malaria in humans and the disease is transferred by Anopheles mosquitos. Development of synthesis methodology for DELs is desperately needed to expand chemical space covered by Though mortality rates are down 47% globally since 2000 and significant progress has been made in the quest these libraries. We developed a solid support-based synthesis strategy that broadens the range of applicable for eradication, reported occurrences of resistance against current therapeutics threaten to reverse that progress. catalytic methods. [2] Among the catalytic systems that are now available for DEL synthesis are organocatalysts, Longstanding treatment chloroquine has seen resistance since the 1950’s, with resistance becoming widespread and transition metal ions such as Au(I), Ag(I), and Cu(I). Our strategy opened access to substituted and in the 70’s and 80’s. Artemisinin, the current main line of defense against malaria, is used in artemisinin functionalized heterocyclic scaffold structures as encodable DNA-conjugates from simple, readily available combination therapies (ACTs) in order to curtail resistance, though at last count, artemisinin resistant parasites starting materials. For example, application of transition metal catalysts furnished DNA-heterocycle conjugates have been reported in 5 countries of the Greater Mekong sub region. In order to curb further resistance, it is through [3+2] cycloaddition reactions. Some of the newly synthesized DNA-conjugated heterocycles display essential that new antimalarial compounds be brought through the pipeline. structural motifs from natural products, while others represent core structures of clinical candidates or approved drugs. All heterocyclic scaffolds enable subsequent DNA-encoded combinatorial library synthesis by For approximately half a century, 4(1H)-quinolones such as endochin or ICI 56,780 were known to be causal well-described, robust reactions. prophylactic and potent erythrocytic stage agents in avian but not in mammalian malaria models. Hit-to-lead optimization of endochin lead to 4(1H)-quinolones ELQ-300 and P4Q-391, which target the liver, the blood as well as the transmitting stages of the parasite. Despite entering preclinical development, ELQ-300 did not enter phase I trials due to limited aqueous solubility and high crystallinity.1 To overcome these limitations, we designed and developed a prodrug approach containing an amino group linked to the parent 4(1H)-quinolone by an acetal carbonate group. Different reaction conditions were found to attach the prodrug moiety selectively onto the oxygen or the nitrogen of the 4(1H)-quinolone scaffold. The resulting O-alkylated prodrugs P4Q-1290 and P4Q-1291 were profiled for physicochemical properties such as chemical stability and aqueous solubility. The prodrugs are stable at low pHs and start releasing the parent drug independently of any enzyme activity at a pH level of about 7. Furthermore, prodrugs P4Q-1290 and P4Q-1291 were highly efficacious in in vivo efficacy assays displaying single-dose cures at low doses. Scheme 1. Solid phase-based transition metal ion- and organocatalyzed synthesis of DNA-heterocycle conjugates serving as starting points for encoded library synthesis.

References 1) Salamon H, Klika Škopić M, Jung K, Bugain O, Brunschweiger A. ACS Chem Biol. 2016, 11, 296-307. 2) Brunschweiger A, Krause N, Antonchick A, Klika Skopic M, Salamon H, Bugain O, Jung K, Wagner B. Eur. Pat. Appl. 15202448.5.

The new discoveries are significant as mitochondrial inhibitors have the potential to advance the malaria elimination campaign by blocking parasite development in the blood and liver, as well as preventing transmission to mosquitoes.

References 1) A. A. Nilsen, A. LaCrue, K. L. White, I. P. Forquer, R. M. Cross, J. Marfurt, M. W. Mather, M. J. Delves, D. M. Shackleford, F. E. Sáenz, J. M. Morrisey, J. Steuten, T. Mutka, Y. Li, G. Wirjanata, E. Ryan, S. Duffy, J. X. Kelly, B. F. Sebayang, A.-M. Zeeman, R. Noviyanti, R. E. Sinden, C. H. M. Kocken, R. N. Price, V. M. Avery, I. Angulo-Barturen, M. B. JiménezDíaz, S. Ferrer, E. Herreros, L. M. Sanz, F. J. G. Benito, I. Bathurst, J. Burrows, P. Siegl, R. K. Guy, R. W. Winter, A. B. Vaidya, S. A. Charman, D. E. Kyle, R. Manetsch*, M. K. Riscoe*, Sci. Transl. Med., 2013, 5, 177ra37.

41 FP05 FP06 FISHING FOR AN "OFF-TARGET": DECONVOLUTING THE DESIGN, SYNTHESIS AND PHARMACOLOGICAL EVALUATION OF MOLECULAR MECHANISM OF GPCR-INDUCED MACROPHAGE FLUORESCENT TOOLS FOR STUDYING CANNABINOID TYPE 2 CHEMOTAXIS RECEPTOR

Maria Chatzopoulou (1), Lewis Taylor (2), Ryan L Davies (1), Minjun Yang (1), Rebecca Hancock (1), Anna G. Cooper (1), Christa Macdonald (2), Michelle Glass (2), Sarah Hook (1), Joel D. A. Tyndall (1), Carole JR Bataille (1), Graham M Wynne (1), David R Greaves (2), Angela J Russell (1,3) Andrea J. Vernall (1)

1) Department of Chemistry, Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford OX1 3TA, UK 1) New Zealand’s National School of Pharmacy, University of Otago, Dunedin, New Zealand 2) Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK 2) Department of Pharmacology and Clinical Pharmacology, University of Auckland, Auckland, New Zealand 3) Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK

Cannabinoid type 2 receptor (CB2R) is a G protein-coupled receptor (GPCR) found primarily in the spleen, The cannabinoid receptors 1 and 2 (CB1 and CB2), are key components of the endocannabinoid system, and tonsils and immune cells, as well as in low levels in the central nervous system. Evidence suggests that CB2R havea significant role in modulating pain perception, the immune system, metabolism and mood. CB1 is could be a promising drug target for many diseases including osteoporosis, inflammatory bowel disease, mostlylocalized in the central nervous system (CNS) and modulation of this GPCR is thought be responsible for atherosclerosis and cancer1. Whilst some immune system functions have been identified, there is a lack of the psychotropic effects of cannabinoids. By contrast, CB2 can be mostly found on cells of the immune detailed understanding of the role of CB2R in immune regulation and the role of neuronal CB2R is largely system,and has been proposed as a therapeutic target for pain and inflammatory conditions such as unknown. Chemical tools that could reliably detect variations in CB2R expression, between different cell types atherosclerosis [1]. and in healthy and diseased tissue would be extremely useful. This would facilitate elucidation of the role CB2R In a previous study to discover selective CB2 agonists [2], whilst studying their effect on macrophage signalling pathways play in disease and how this could be regulated using CB2R targeted drugs. There are a chemotaxis, we demonstrated that chemotaxis induced by selective CB2 agonists can be uncoupled from CB2 variety of existing methods available for studying CB2R such as fluorescent immunohistochemistry, activation and be considered as an off-target effect at a non-CB1/CB2 Gi/o-coupled receptor [3]. radioligands and reporter gene assays. However each of these have limitations such as poor receptor specificity Following that, we performed a ligand-based virtual screening of in-house library, using four positive for and low reproducibility. Fluorescent ligands are excellent tools to study receptor structure and function in live macrophage chemotaxis CB2 agonists as templates. Further hit validation, revealed that several cells and have been successfully designed for other GPCRs. Fluorescent ligands can be used to investigate 2-thioxo-2,3-dihydroquinazolin-4(1H)-one derivatives share significant structural and electronic similarity to the dynamic receptor processes such as receptor activation and trafficking and can be employed in a variety of reference compounds, as well as, hold potential as mediators of macrophage chemotaxis. As a result, a fluorescence based imaging techniques2. In this study a library of aminoalkylindole derivatives has been structure-activity relationship study was undertaken in order to uncover the sites in this scaffold, where a synthesised with the aim of developing a selective, high affinity fluorescent ligand for CB2R. The indole benzophenone photo-affinity label may be installed. scaffold and attached substituents were selected on the basis of existing structure activity relationships (SAR) Gaining access to such a probe is of great importance as it will lead to the deconvolution of the CB2 role during and a linker and fluorophore were coupled at a position thought to be tolerant of steric bulk. The synthesis was inflammation, and/or possibly lead to the identification of new players in macrophage chemotaxis and carried out over nine steps, generating seven final fluorescent compounds. These fluorescent compounds along inflammation. with key synthetic intermediates underwent pharmacological evaluation. Radioligand binding assays were carried out to determine affinity and CBR subtype selectivity. Functional BRET assays measuring change in References cAMP were then carried out on lead compounds. Whilst the fluorescent compounds were not of sufficient affinity to be used as chemical tools, a number of new CB2R selective agonists were identified amongst the 1) Steffens S. & Mach F. Curr Opin Lipidology 2006, 17, 519 2) Gianella-Borradori M.; Christou I.; Bataille C.J.R., Cross R.L.; Wynne G.L.; Greaves D.R.; Russell A.J. Bioorg Med synthetic intermediates, the best of which showed a Ki = 97.8 nM (hCB2R). In addition, valuable new SAR have Chem 2015, 23, 241 been derived for this compound class, which will guide our future fluorescent ligand design. 3) Taylor L.; Christou I.; Kapellos T.S.; Buchan A.; Brodermann M.H.; Gianella-Borradori M.; Russell A.J.; Iqbal A.J.; Greaves D.R. Sci Rep 2015, 5, 10682 References 1) Pacher, P.; Batkai, S.; Kunos, G., The endocannabinoid system as an emerging target of pharmacotherapy. Pharmacological reviews 2006, 58 (3), 389-462. 2) Vernall, A. J.; Hill, S. J.; Kellam, B., The evolving small-molecule fluorescent-conjugate toolbox for Class A GPCRs. British journal of pharmacology 2014, 171 (5), 1073-84.

42 FP05 FP06 FISHING FOR AN "OFF-TARGET": DECONVOLUTING THE DESIGN, SYNTHESIS AND PHARMACOLOGICAL EVALUATION OF MOLECULAR MECHANISM OF GPCR-INDUCED MACROPHAGE FLUORESCENT TOOLS FOR STUDYING CANNABINOID TYPE 2 CHEMOTAXIS RECEPTOR

Maria Chatzopoulou (1), Lewis Taylor (2), Ryan L Davies (1), Minjun Yang (1), Rebecca Hancock (1), Anna G. Cooper (1), Christa Macdonald (2), Michelle Glass (2), Sarah Hook (1), Joel D. A. Tyndall (1), Carole JR Bataille (1), Graham M Wynne (1), David R Greaves (2), Angela J Russell (1,3) Andrea J. Vernall (1)

1) Department of Chemistry, Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford OX1 3TA, UK 1) New Zealand’s National School of Pharmacy, University of Otago, Dunedin, New Zealand 2) Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK 2) Department of Pharmacology and Clinical Pharmacology, University of Auckland, Auckland, New Zealand 3) Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK

Cannabinoid type 2 receptor (CB2R) is a G protein-coupled receptor (GPCR) found primarily in the spleen, The cannabinoid receptors 1 and 2 (CB1 and CB2), are key components of the endocannabinoid system, and tonsils and immune cells, as well as in low levels in the central nervous system. Evidence suggests that CB2R havea significant role in modulating pain perception, the immune system, metabolism and mood. CB1 is could be a promising drug target for many diseases including osteoporosis, inflammatory bowel disease, mostlylocalized in the central nervous system (CNS) and modulation of this GPCR is thought be responsible for atherosclerosis and cancer1. Whilst some immune system functions have been identified, there is a lack of the psychotropic effects of cannabinoids. By contrast, CB2 can be mostly found on cells of the immune detailed understanding of the role of CB2R in immune regulation and the role of neuronal CB2R is largely system,and has been proposed as a therapeutic target for pain and inflammatory conditions such as unknown. Chemical tools that could reliably detect variations in CB2R expression, between different cell types atherosclerosis [1]. and in healthy and diseased tissue would be extremely useful. This would facilitate elucidation of the role CB2R In a previous study to discover selective CB2 agonists [2], whilst studying their effect on macrophage signalling pathways play in disease and how this could be regulated using CB2R targeted drugs. There are a chemotaxis, we demonstrated that chemotaxis induced by selective CB2 agonists can be uncoupled from CB2 variety of existing methods available for studying CB2R such as fluorescent immunohistochemistry, activation and be considered as an off-target effect at a non-CB1/CB2 Gi/o-coupled receptor [3]. radioligands and reporter gene assays. However each of these have limitations such as poor receptor specificity Following that, we performed a ligand-based virtual screening of in-house library, using four positive for and low reproducibility. Fluorescent ligands are excellent tools to study receptor structure and function in live macrophage chemotaxis CB2 agonists as templates. Further hit validation, revealed that several cells and have been successfully designed for other GPCRs. Fluorescent ligands can be used to investigate 2-thioxo-2,3-dihydroquinazolin-4(1H)-one derivatives share significant structural and electronic similarity to the dynamic receptor processes such as receptor activation and trafficking and can be employed in a variety of reference compounds, as well as, hold potential as mediators of macrophage chemotaxis. As a result, a fluorescence based imaging techniques2. In this study a library of aminoalkylindole derivatives has been structure-activity relationship study was undertaken in order to uncover the sites in this scaffold, where a synthesised with the aim of developing a selective, high affinity fluorescent ligand for CB2R. The indole benzophenone photo-affinity label may be installed. scaffold and attached substituents were selected on the basis of existing structure activity relationships (SAR) Gaining access to such a probe is of great importance as it will lead to the deconvolution of the CB2 role during and a linker and fluorophore were coupled at a position thought to be tolerant of steric bulk. The synthesis was inflammation, and/or possibly lead to the identification of new players in macrophage chemotaxis and carried out over nine steps, generating seven final fluorescent compounds. These fluorescent compounds along inflammation. with key synthetic intermediates underwent pharmacological evaluation. Radioligand binding assays were carried out to determine affinity and CBR subtype selectivity. Functional BRET assays measuring change in References cAMP were then carried out on lead compounds. Whilst the fluorescent compounds were not of sufficient affinity to be used as chemical tools, a number of new CB2R selective agonists were identified amongst the 1) Steffens S. & Mach F. Curr Opin Lipidology 2006, 17, 519 2) Gianella-Borradori M.; Christou I.; Bataille C.J.R., Cross R.L.; Wynne G.L.; Greaves D.R.; Russell A.J. Bioorg Med synthetic intermediates, the best of which showed a Ki = 97.8 nM (hCB2R). In addition, valuable new SAR have Chem 2015, 23, 241 been derived for this compound class, which will guide our future fluorescent ligand design. 3) Taylor L.; Christou I.; Kapellos T.S.; Buchan A.; Brodermann M.H.; Gianella-Borradori M.; Russell A.J.; Iqbal A.J.; Greaves D.R. Sci Rep 2015, 5, 10682 References 1) Pacher, P.; Batkai, S.; Kunos, G., The endocannabinoid system as an emerging target of pharmacotherapy. Pharmacological reviews 2006, 58 (3), 389-462. 2) Vernall, A. J.; Hill, S. J.; Kellam, B., The evolving small-molecule fluorescent-conjugate toolbox for Class A GPCRs. British journal of pharmacology 2014, 171 (5), 1073-84.

43 FP07 FP08 DEVELOPMENT OF NOVEL, SELECTIVE AND IRREVERSIBLE IDENTIFICATION OF NOVEL NON-NATURAL SUPRAMOLECULAR PI3Kδ INHIBITORS LIGANDS AS STABILIZERS OF 14-3-3? PROTEIN-PROTEIN INTERACTIONS Samuel E. Dalton (1,2), Sébastien A. Campos (2), Jacob T. Bush (2), Daniel A. Thomas (2), Máire A. Convery (2), John A. Murphy (1) Alba Gigante, Jeroen Briels, Maria Bartel, Christian Ottmann, Carsten Schmuck 1) University of Strathclyde Pure and Applied Chemistry, Glasgow, United Kingdom 1) Department of Organic Chemistry, University of Duisburg-Essen. 2) GlaxoSmithKline, Stevenage, Hertfordshire, United Kingdom Universitätsstraße 7, 45117-Essen (Germany) 2) Department of Biomedical Engineering, University of Technology. Postbus 513, 5600-MB-Eindhoven (The Nederlands)

Phosphoinositide 3-kinase δ (PI3Kδ) is involved in T-cell receptor signalling, B-cell development and neutrophil trafficking into inflamed tissues, making it an attractive target for the treatment of inflammatory and autoimmune diseases. A number of selective reversible PI3Kδ inhibitors have been developed, most notably 14-3-3 adaptor proteins play a central role in signal transduction. The recent resolution of crystal structures of ZydeligTM, which has recently been approved by the FDA for the treatment of chronic lymphocytic leukemia. different ligands in complex with these adaptor proteins opens up a formidable opportunity of modulating their Wortmannin and its structurally related analogues (e.g. PX-866) are, to date, the only reported PI3Kδ inhibitors physiological functions, and thereby, those implicated in pathological processes.[1] Herein is reported the which covalently bind to the kinase via reaction with a conserved lysine residue situated in the ATP binding site. identification of the first synthetic supramolecular stabilizers of the interaction between the adapter protein However, these compounds show poor selectivity, especially for the various PI3K isoforms, restricting their use 14-3-3ζ and two of its effectors: c-Raf and Tau, which are involved in proliferative signal transduction and to specific indications. neurodegenerative diseases, respectively.[2],[3] These new ligands are decorated with the non-proteinogenic GCP (guanidinocarbonylpyrrole), an arginine mimetic designed by our group, which allows the By modifying selective reversible PI3Kδ inhibitors with carefully positioned electrophilic moieties, it was stabilization of these interactions in the micromolar range. proposed that selective irreversible PI3Kδ inhibition could be achieved, despite the conserved nature of the targeted lysine residue within the PI3K family. The design and development of such compounds will be discussed, with data supporting their potency and selectivity in biochemical assays. Full characterisation of the covalent reaction using mass spectrometry, time-course experiments, and X-ray crystallography will also be presented. To our knowledge, this work represents the first established evidence of selective irreversible PI3Kδ inhibitors, and constitutes a novel conceptual approach in this area with the potential to assist in the strategic design of related medicinal chemistry programmes.

References 1) M. Molzan, S. Kasper, L. Röglin, M. Skwarczynska, T. Sassa, T. Inoue, F. Breitenbuecher, J. Ohkanda, N. Kato, M. Schuler, et al., ACS Chem. Biol. 2013, 8, 1869–1875 2) E. Wilker, M. B. Yaffe, J. Mol. Cell. Cardiol. 2004, 37, 633–642. 3) M. Otto, J. Wiltfang, L. Cepek, M. Neumann, B. Mollenhauer, P. Steinacker, B. Ciesielczyk, W. Schulz-Schaeffer, H. Kretzschmar, S. Poser, Neurol. 2002, 58, 192-7.

44 FP07 FP08 DEVELOPMENT OF NOVEL, SELECTIVE AND IRREVERSIBLE IDENTIFICATION OF NOVEL NON-NATURAL SUPRAMOLECULAR PI3Kδ INHIBITORS LIGANDS AS STABILIZERS OF 14-3-3? PROTEIN-PROTEIN INTERACTIONS Samuel E. Dalton (1,2), Sébastien A. Campos (2), Jacob T. Bush (2), Daniel A. Thomas (2), Máire A. Convery (2), John A. Murphy (1) Alba Gigante, Jeroen Briels, Maria Bartel, Christian Ottmann, Carsten Schmuck 1) University of Strathclyde Pure and Applied Chemistry, Glasgow, United Kingdom 1) Department of Organic Chemistry, University of Duisburg-Essen. 2) GlaxoSmithKline, Stevenage, Hertfordshire, United Kingdom Universitätsstraße 7, 45117-Essen (Germany) 2) Department of Biomedical Engineering, University of Technology. Postbus 513, 5600-MB-Eindhoven (The Nederlands)

Phosphoinositide 3-kinase δ (PI3Kδ) is involved in T-cell receptor signalling, B-cell development and neutrophil trafficking into inflamed tissues, making it an attractive target for the treatment of inflammatory and autoimmune diseases. A number of selective reversible PI3Kδ inhibitors have been developed, most notably 14-3-3 adaptor proteins play a central role in signal transduction. The recent resolution of crystal structures of ZydeligTM, which has recently been approved by the FDA for the treatment of chronic lymphocytic leukemia. different ligands in complex with these adaptor proteins opens up a formidable opportunity of modulating their Wortmannin and its structurally related analogues (e.g. PX-866) are, to date, the only reported PI3Kδ inhibitors physiological functions, and thereby, those implicated in pathological processes.[1] Herein is reported the which covalently bind to the kinase via reaction with a conserved lysine residue situated in the ATP binding site. identification of the first synthetic supramolecular stabilizers of the interaction between the adapter protein However, these compounds show poor selectivity, especially for the various PI3K isoforms, restricting their use 14-3-3ζ and two of its effectors: c-Raf and Tau, which are involved in proliferative signal transduction and to specific indications. neurodegenerative diseases, respectively.[2],[3] These new ligands are decorated with the non-proteinogenic amino acid GCP (guanidinocarbonylpyrrole), an arginine mimetic designed by our group, which allows the By modifying selective reversible PI3Kδ inhibitors with carefully positioned electrophilic moieties, it was stabilization of these interactions in the micromolar range. proposed that selective irreversible PI3Kδ inhibition could be achieved, despite the conserved nature of the targeted lysine residue within the PI3K family. The design and development of such compounds will be discussed, with data supporting their potency and selectivity in biochemical assays. Full characterisation of the covalent reaction using mass spectrometry, time-course experiments, and X-ray crystallography will also be presented. To our knowledge, this work represents the first established evidence of selective irreversible PI3Kδ inhibitors, and constitutes a novel conceptual approach in this area with the potential to assist in the strategic design of related medicinal chemistry programmes.

References 1) M. Molzan, S. Kasper, L. Röglin, M. Skwarczynska, T. Sassa, T. Inoue, F. Breitenbuecher, J. Ohkanda, N. Kato, M. Schuler, et al., ACS Chem. Biol. 2013, 8, 1869–1875 2) E. Wilker, M. B. Yaffe, J. Mol. Cell. Cardiol. 2004, 37, 633–642. 3) M. Otto, J. Wiltfang, L. Cepek, M. Neumann, B. Mollenhauer, P. Steinacker, B. Ciesielczyk, W. Schulz-Schaeffer, H. Kretzschmar, S. Poser, Neurol. 2002, 58, 192-7.

45 FP09 FP10 TARGETS IN ORPHAN DISEASES AND THEIR REPURPOSING SELECTIVITY PROFILING OF THE HUMAN MONOAMINE CANDIDATES - ANALYSIS WITH A KNIME WORKFLOW USING TRANSPORTERS: A CASE STUDY ON CATHINONE ANALOGS OPEN PHACTS Eva Hellsberg, Barbara Zdrazil, Gerhard F. Ecker Jana Gurinova, Daniela Digles, Gerhard F. Ecker University of Vienna, Althanstraße 14, 1090 Vienna, Austria University of Vienna, Department of Pharmaceutical Chemistry, Althanstraße 14, 1090 Vienna, Austria

The human monoamine transporters including hSERT (human serotonin transporter), hDAT (human dopamine Worldwide an estimated number of 400 million people are affected by orphan diseases1. An orphan disease is transporter) and hNET (human norepinephrine transporter) belong to the solute carrier 6 (SLC6) gene family defined as affecting less than 1 in 2000 citizens2. Such a low prevalence coupled with the sheer number of (also referred to the neurotransmitter-sodium-symporter family (NSS) or Na+ /Cl- -dependent transporters). They orphan diseases, estimated to be about 5000-8000, is the main reason for the small number of marketing play an important role in the central and peripheral nervous system by regulating the signaling among neurons. approvals, amounting to treatments for roughly 200 conditions in the US and only about 45 in the European Numerous compound classes have been identified to interact with these transporters, and they are used either in Union1. Drug repurposing therefore may prove to be the future of drug discovery for orphan diseases because it atherapeutic setting or are abused as illicit drugs. is an attractive option of reaching many patients with treatments that have already been deemed safe. This work This project concentrates on the molecular basis and the chemical characteristics of ligand transporter interaction aims at providing an overview of targets linked to orphan diseases as well as relevant compounds, consisting of and selectivity at hSERT and hDAT. By exploring the chemical space of hSERT and hDAT interacting possible repurposing candidates for these targets as well as experimental compounds as a starting point for drug compounds via the OpenPHACTS Discovery Platform by using KNIME, we analyzed the scaffolds appearing discovery. selective for either hSERT or hDAT. As use case for further studies we chose the class of cathinones. They The biggest European platform for orphan diseases is Orphanet3, with comprehensive information for patients as represent a subclass of the amphetamines and are a quite prominent group of abusive drugs with a rising trend of well as for healthcare professionals, providing also identifiers (UMLS, MeSH, OMIM etc.) for 5345 of its 9235 consumption. Throughout the data extraction process, we collected 56 compounds sharing this scaffold and listed diseases, as well as for associated genes. These identifiers are perfectly suitable for data integration across being tested in the same biological assay type. The main structural variations include the substituent on the different databases and thus also for data enrichment with the aim of understanding and visualizing the role of nitrogen atom, the substituent at the aromatic ring, as well as some modifications at the Cα-atom. diverse protein classes such as ion channels or transporters in orphan diseases. The Open PHACTS Discovery Subsequent structure-activity relationship studies with hDAT pIC50 values and selectivity as dependent Platform contains the linked datasets for this kind of data integration, given that the included databases (such as variables,as well as docking and MD simulations of selected compounds into protein homology models of DisGeNET, UniProt, DrugBank, SureChEMBL and ChEMBL) enable the user to go from the disease to its hSERT and hDAT revealed first insights into the molecular basis of transporter selectivity by pointing out an targets and ultimately to approved and experimental compounds connected to these targets. The approved influence of the substituent at the Cα-atom to the carbonyl group. compounds may include candidates for drug repositioning, whereas the experimental compounds may provide a foundation for drug discovery. Acknowledgements: The workflow was established by using the KNIME Analytics Platform and starts with extracting data related to The research leading to these results has received support from the Innovative Medicines Initiative Joint orphan diseases from different sources. Primarily it extracts identifiers provided by Orphanet and links them to Undertaking under grant agreement n° 115191, resources of which are composed of financial contribution from respective targets through the use of DisGeNET and UniProt, additionally target references provided by the EuropeanUnion's Seventh Framework Programme (FP7/2007-2013) and EFPIA companies in kind Orphanet are also linked to the Open PHACTS Discovery Platform for further data integration, resulting in a contribution. We further wish to acknowledge the Open PHACTS Foundation, the charitable organization combined dataset of specific targets for orphan diseases. The workflow then links this dataset to DrugBank, responsible for the Open PHACTS Discovery Platform, for providing access to the Platform aswell as the SureChEMBL and ChEMBL, with the results consisting of approved drugs as well as experimental compounds Austrian Science Fund (FWF), grants F03502 and W1232. for the targets involved in orphan diseases. It is remarkable that solely by using the identifiers from Orphanet the workflow already results in 1269 approved compounds that may be examined for suitable drug repurposing candidates. Acknowledgements The work has received support from the Innovative Medicines Initiative Joint Undertaking under grant agreement no. [115191], resources of which are composed of financial contribution from the European Union's Seventh Framework Programme (FP7/2007-2013) and in-kind contribution of EFPIA companies. We further wish to acknowledge the Open PHACTS Foundation, the charitable organisation responsible for the Open PHACTS Discovery Platform, without which this work would not have been possible.

References 1) Kaplan W et al., Priority Medicines for Europe and the World Update 2013, World Health Organization, Geneva, 2013 2) EURORDIS, Rare Diseases: understanding this Public Health Priority, EURORDIS, Paris, 2005 3) Orphanet: an online rare disease and orphan drug data base. © INSERM 1997. Available at http://www.orpha.net (accessed December 2015)

46 FP09 FP10 TARGETS IN ORPHAN DISEASES AND THEIR REPURPOSING SELECTIVITY PROFILING OF THE HUMAN MONOAMINE CANDIDATES - ANALYSIS WITH A KNIME WORKFLOW USING TRANSPORTERS: A CASE STUDY ON CATHINONE ANALOGS OPEN PHACTS Eva Hellsberg, Barbara Zdrazil, Gerhard F. Ecker Jana Gurinova, Daniela Digles, Gerhard F. Ecker University of Vienna, Althanstraße 14, 1090 Vienna, Austria University of Vienna, Department of Pharmaceutical Chemistry, Althanstraße 14, 1090 Vienna, Austria

The human monoamine transporters including hSERT (human serotonin transporter), hDAT (human dopamine Worldwide an estimated number of 400 million people are affected by orphan diseases1. An orphan disease is transporter) and hNET (human norepinephrine transporter) belong to the solute carrier 6 (SLC6) gene family defined as affecting less than 1 in 2000 citizens2. Such a low prevalence coupled with the sheer number of (also referred to the neurotransmitter-sodium-symporter family (NSS) or Na+ /Cl- -dependent transporters). They orphan diseases, estimated to be about 5000-8000, is the main reason for the small number of marketing play an important role in the central and peripheral nervous system by regulating the signaling among neurons. approvals, amounting to treatments for roughly 200 conditions in the US and only about 45 in the European Numerous compound classes have been identified to interact with these transporters, and they are used either in Union1. Drug repurposing therefore may prove to be the future of drug discovery for orphan diseases because it atherapeutic setting or are abused as illicit drugs. is an attractive option of reaching many patients with treatments that have already been deemed safe. This work This project concentrates on the molecular basis and the chemical characteristics of ligand transporter interaction aims at providing an overview of targets linked to orphan diseases as well as relevant compounds, consisting of and selectivity at hSERT and hDAT. By exploring the chemical space of hSERT and hDAT interacting possible repurposing candidates for these targets as well as experimental compounds as a starting point for drug compounds via the OpenPHACTS Discovery Platform by using KNIME, we analyzed the scaffolds appearing discovery. selective for either hSERT or hDAT. As use case for further studies we chose the class of cathinones. They The biggest European platform for orphan diseases is Orphanet3, with comprehensive information for patients as represent a subclass of the amphetamines and are a quite prominent group of abusive drugs with a rising trend of well as for healthcare professionals, providing also identifiers (UMLS, MeSH, OMIM etc.) for 5345 of its 9235 consumption. Throughout the data extraction process, we collected 56 compounds sharing this scaffold and listed diseases, as well as for associated genes. These identifiers are perfectly suitable for data integration across being tested in the same biological assay type. The main structural variations include the substituent on the different databases and thus also for data enrichment with the aim of understanding and visualizing the role of nitrogen atom, the substituent at the aromatic ring, as well as some modifications at the Cα-atom. diverse protein classes such as ion channels or transporters in orphan diseases. The Open PHACTS Discovery Subsequent structure-activity relationship studies with hDAT pIC50 values and selectivity as dependent Platform contains the linked datasets for this kind of data integration, given that the included databases (such as variables,as well as docking and MD simulations of selected compounds into protein homology models of DisGeNET, UniProt, DrugBank, SureChEMBL and ChEMBL) enable the user to go from the disease to its hSERT and hDAT revealed first insights into the molecular basis of transporter selectivity by pointing out an targets and ultimately to approved and experimental compounds connected to these targets. The approved influence of the substituent at the Cα-atom to the carbonyl group. compounds may include candidates for drug repositioning, whereas the experimental compounds may provide a foundation for drug discovery. Acknowledgements: The workflow was established by using the KNIME Analytics Platform and starts with extracting data related to The research leading to these results has received support from the Innovative Medicines Initiative Joint orphan diseases from different sources. Primarily it extracts identifiers provided by Orphanet and links them to Undertaking under grant agreement n° 115191, resources of which are composed of financial contribution from respective targets through the use of DisGeNET and UniProt, additionally target references provided by the EuropeanUnion's Seventh Framework Programme (FP7/2007-2013) and EFPIA companies in kind Orphanet are also linked to the Open PHACTS Discovery Platform for further data integration, resulting in a contribution. We further wish to acknowledge the Open PHACTS Foundation, the charitable organization combined dataset of specific targets for orphan diseases. The workflow then links this dataset to DrugBank, responsible for the Open PHACTS Discovery Platform, for providing access to the Platform aswell as the SureChEMBL and ChEMBL, with the results consisting of approved drugs as well as experimental compounds Austrian Science Fund (FWF), grants F03502 and W1232. for the targets involved in orphan diseases. It is remarkable that solely by using the identifiers from Orphanet the workflow already results in 1269 approved compounds that may be examined for suitable drug repurposing candidates. Acknowledgements The work has received support from the Innovative Medicines Initiative Joint Undertaking under grant agreement no. [115191], resources of which are composed of financial contribution from the European Union's Seventh Framework Programme (FP7/2007-2013) and in-kind contribution of EFPIA companies. We further wish to acknowledge the Open PHACTS Foundation, the charitable organisation responsible for the Open PHACTS Discovery Platform, without which this work would not have been possible.

References 1) Kaplan W et al., Priority Medicines for Europe and the World Update 2013, World Health Organization, Geneva, 2013 2) EURORDIS, Rare Diseases: understanding this Public Health Priority, EURORDIS, Paris, 2005 3) Orphanet: an online rare disease and orphan drug data base. © INSERM 1997. Available at http://www.orpha.net (accessed December 2015)

47 FP11 FP12 PHOTOSWITCHABLE 4-(PIPERIDIN-4-YL)-1-HYDROXYPYRAZOLE EXTENDING SAR OF BILE ACIDS AS FXR/TGR5 MODULATORS: ANALOGUES FOR THE GABA TYPE A RECEPTORS SYNTHESIS AND BIOLOGICAL ACTIVITY OF RING C MODIFIED CDCA DERIVATIVES Henriette Kihl, Jacob Krall, Kenneth Thermann Kongstad, Birgitte Nielsen, Bente Frølund Serena Mostarda (1), Emiliano Rosatelli (2), Daniela Passeri (2), Francesca De Franco (2), Tiziana Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Benicchi (2), Andrea Carotti (1), Antonio Macchiarulo (1), Roberto Pellicciari (2), Antimo Gioiello (1) Universitetsparken 2, DK-2100 Copenhagen, Denmark. 1) Dipartimento di Scienze Farmaceutiche, Università degli Studi di Perugia, Via del Liceo 1, I-06123, Perugia, Italy. 2) TES-Pharma S.r.l., Via Palmiro Togliatti 20, 06073 Loc. Taverne, Corciano (Perugia), Italy.

γ-aminobutyric acid (GABA) is the major inhibitory neurotransmitter in the mammalian central nervous system (CNS) and exerts the majority of its effects through the GABAA receptors (GABAAR). The GABAARs are pentameric ligand-gated ion channels that mediate fast synaptic responses to GABA and belong to the Cys-loop During the last two decades, there have been extraordinary advances in the knowledge of the chemistry and receptor superfamily. GABAARs are widely distributed in the CNS and proposed to be involved in several biology of bile acids (BAs).1 The astonishing discovery of the hormone-like properties of BAs has led to a 1 neurodegenerative and psychiatric disorders including anxiety, schizophrenia, and Alzheimer's . complex scenario in which their action was found crucial for a network of signalling pathways mainly driven by Photopharmacology provides an opportunity to regulate biological processes by light. Light is non-toxic and the nuclear receptor Farnesoid X Receptor (FXR) and the membrane receptor TGR5, controlling triglyceride and 2 non-invasive and can be delivered with high spatiotemporal precision. Furthermore, qualitative and quantitative cholesterol levels, as well as energy and glucose homeostasis. Intense endeavors have thus been dedicated by regulation of light is possible by adjusting wavelength and intensity, respectively. Photoswitches are small medicinal chemists to design and develop semisynthetic BA derivatives, natural products and non-steroidal 3 molecules, which upon absorption of a photon undergo reversible conformational changes. The commonly used ligands as potent and selective FXR and TGR5 modulators. In particular, the work of our group in the field has photoswitch azobenzene undergo cis/trans isomerization upon photon absorption resulting in a large change in elucidated the structure-activity relationships (SAR) of BAs as FXR/TGR5 ligands, identifying functional hot geometry and polarity2. Coupling of a photoswitch to a ligand enables reversible control by light of not spots on the BA structure and disclosing a number of compounds that are on track for preclinical and clinical ordinarily light-sensitive receptors. assessments of liver and metabolic disorders. These studies have also contributed to show how apparently minor chemical modifications of the BA scaffold influence the physicochemical, pharmacokinetic and We have previously shown that 3- or 5-substituted 4-(piperidin-4-yl)-1-hydroxypyrazoles (4-PHP) bind to the pharmacodynamic profiles of the resulting analogues, thereby determining their fate for the advancement in orthosteric binding site of GABAARs and the identified cavities are capable of accommodating relatively large clinical settings. substituents on 4-PHP3,4. These cavities could enable introduction of photoswitches such as azobenzene with retained affinity of the ligands at the GABAAR. As a continuation of our work in the field, in this communication we report the synthesis and preliminary biological characterization of new BA derivatives as FXR/TGR5 ligands designed with the aim to unveil the role of unexplored positions of the biliary scaffold in the binding and activation of both receptors. Thus, the synthesis of a set of novel CDCA derivatives bearing modifications at the ring C, the evaluation of their ability to modulate the FXR and TGR5 receptors, and the results of computational analysis on the hitherto hidden SAR aspects of BAs will be presented and discussed.

References 1) Hofmann, A. F, Hagey, L. R. Key discoveries in bile acid chemistry and biology and their clinical applications: history of the last eight decades. J Lipid Res. 2014, 55, 1553-1595. 2) Schaap, F. G., Trauner, M., Jansen P. L. Bile acid receptors as targets for drug development. Nat. Rev. Gastroenterol. Hepatol. 2014, 11, 55-67. 3) a) Gioiello, A., Cerra, B., Mostarda, S., Guercini, C., Pellicciari, R., Macchiarulo, A. Bile acid derivatives as ligands of the Farnesoid X Receptor: molecular determinants for bile acid binding and receptor modulation. Curr. Top. Med. Chem. 2014, 14, 2159-2174; b) Carotti, A., Marinozzi, M., Custodi, C., Cerra, B., Pellicciari, R., Gioiello, A., Macchiarulo, A. Beyond bile acids: targeting Farnesoid X Receptor (FXR) with natural and synthetic ligands. Curr. Top. Med. Chem. 2014, 14, 2129-2142; c) Gioiello, A., Rosatelli, E., Nuti, R., Macchiarulo, A., Pellicciari, R. Patented TGR5 modulators: a review (2006 - present). Expert Opin. Ther. Pat. 2012, 22, 1399-1414. Based on the abovementioned observations we introduced azobenzene in the 3- or 5-position of 4-PHP, which led to a series of photoswitchable analogues. The analogues behave similar to azobenzene with cis/trans isomerization upon irradiation with UV-Vis light and thermal relaxation over time.

Pharmacological characterization at GABAARs was performed in a [3H] binding assay using rat brain cortical membranes. Binding affinities of all dark adapted analogues were in the low micro- to low nanomolar range at native rat GABAARs.

References 1) Foster AC, Kemp JA. Glutamate- and GABA-based CNS therapeutics. Curr. Opin. Pharmacol. 2006, 6, 7-17. 2) Beharry AA, Woolley GA. Azobenzene photoswitches for biomolecules. Chem. Soc. Rev. 2011, 40, 4422–4437. 3) Krall J, Jensen CH, Sørensen TE, Nielsen B, Jensen AA, Sander T, Balle T, Frølund B. Exploring the orthosteric binding site of the γ-aminobutyric acid type A receptor using 4-(Piperidin-4-yl)-1-hydroxypyrazoles 3- or 5-imidazolyl substituted: design, synthesis, and pharmacological evaluation. J. Med. Chem. 2013, 56, 6536-6540. 4) Møller, H. A.; Sander, T.; Kristensen, J. L.; Nielsen, B.; Krall, J.; Bergmann, M. L.; Christiansen, B.; Balle, T.; Jensen, A. A.; Frølund, B. Novel 4-(Piperidin-4-yl)-1-hydroxypyrazoles as γ-Aminobutyric AcidA Receptor Ligands: Synthesis, Pharmacology, and Structure−Activity Relationships. J. Med. Chem. 2010, 53, 3417-3421.

48 FP11 FP12 PHOTOSWITCHABLE 4-(PIPERIDIN-4-YL)-1-HYDROXYPYRAZOLE EXTENDING SAR OF BILE ACIDS AS FXR/TGR5 MODULATORS: ANALOGUES FOR THE GABA TYPE A RECEPTORS SYNTHESIS AND BIOLOGICAL ACTIVITY OF RING C MODIFIED CDCA DERIVATIVES Henriette Kihl, Jacob Krall, Kenneth Thermann Kongstad, Birgitte Nielsen, Bente Frølund Serena Mostarda (1), Emiliano Rosatelli (2), Daniela Passeri (2), Francesca De Franco (2), Tiziana Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Benicchi (2), Andrea Carotti (1), Antonio Macchiarulo (1), Roberto Pellicciari (2), Antimo Gioiello (1) Universitetsparken 2, DK-2100 Copenhagen, Denmark. 1) Dipartimento di Scienze Farmaceutiche, Università degli Studi di Perugia, Via del Liceo 1, I-06123, Perugia, Italy. 2) TES-Pharma S.r.l., Via Palmiro Togliatti 20, 06073 Loc. Taverne, Corciano (Perugia), Italy.

γ-aminobutyric acid (GABA) is the major inhibitory neurotransmitter in the mammalian central nervous system (CNS) and exerts the majority of its effects through the GABAA receptors (GABAAR). The GABAARs are pentameric ligand-gated ion channels that mediate fast synaptic responses to GABA and belong to the Cys-loop During the last two decades, there have been extraordinary advances in the knowledge of the chemistry and receptor superfamily. GABAARs are widely distributed in the CNS and proposed to be involved in several biology of bile acids (BAs).1 The astonishing discovery of the hormone-like properties of BAs has led to a 1 neurodegenerative and psychiatric disorders including anxiety, schizophrenia, and Alzheimer's . complex scenario in which their action was found crucial for a network of signalling pathways mainly driven by Photopharmacology provides an opportunity to regulate biological processes by light. Light is non-toxic and the nuclear receptor Farnesoid X Receptor (FXR) and the membrane receptor TGR5, controlling triglyceride and 2 non-invasive and can be delivered with high spatiotemporal precision. Furthermore, qualitative and quantitative cholesterol levels, as well as energy and glucose homeostasis. Intense endeavors have thus been dedicated by regulation of light is possible by adjusting wavelength and intensity, respectively. Photoswitches are small medicinal chemists to design and develop semisynthetic BA derivatives, natural products and non-steroidal 3 molecules, which upon absorption of a photon undergo reversible conformational changes. The commonly used ligands as potent and selective FXR and TGR5 modulators. In particular, the work of our group in the field has photoswitch azobenzene undergo cis/trans isomerization upon photon absorption resulting in a large change in elucidated the structure-activity relationships (SAR) of BAs as FXR/TGR5 ligands, identifying functional hot geometry and polarity2. Coupling of a photoswitch to a ligand enables reversible control by light of not spots on the BA structure and disclosing a number of compounds that are on track for preclinical and clinical ordinarily light-sensitive receptors. assessments of liver and metabolic disorders. These studies have also contributed to show how apparently minor chemical modifications of the BA scaffold influence the physicochemical, pharmacokinetic and We have previously shown that 3- or 5-substituted 4-(piperidin-4-yl)-1-hydroxypyrazoles (4-PHP) bind to the pharmacodynamic profiles of the resulting analogues, thereby determining their fate for the advancement in orthosteric binding site of GABAARs and the identified cavities are capable of accommodating relatively large clinical settings. substituents on 4-PHP3,4. These cavities could enable introduction of photoswitches such as azobenzene with retained affinity of the ligands at the GABAAR. As a continuation of our work in the field, in this communication we report the synthesis and preliminary biological characterization of new BA derivatives as FXR/TGR5 ligands designed with the aim to unveil the role of unexplored positions of the biliary scaffold in the binding and activation of both receptors. Thus, the synthesis of a set of novel CDCA derivatives bearing modifications at the ring C, the evaluation of their ability to modulate the FXR and TGR5 receptors, and the results of computational analysis on the hitherto hidden SAR aspects of BAs will be presented and discussed.

References 1) Hofmann, A. F, Hagey, L. R. Key discoveries in bile acid chemistry and biology and their clinical applications: history of the last eight decades. J Lipid Res. 2014, 55, 1553-1595. 2) Schaap, F. G., Trauner, M., Jansen P. L. Bile acid receptors as targets for drug development. Nat. Rev. Gastroenterol. Hepatol. 2014, 11, 55-67. 3) a) Gioiello, A., Cerra, B., Mostarda, S., Guercini, C., Pellicciari, R., Macchiarulo, A. Bile acid derivatives as ligands of the Farnesoid X Receptor: molecular determinants for bile acid binding and receptor modulation. Curr. Top. Med. Chem. 2014, 14, 2159-2174; b) Carotti, A., Marinozzi, M., Custodi, C., Cerra, B., Pellicciari, R., Gioiello, A., Macchiarulo, A. Beyond bile acids: targeting Farnesoid X Receptor (FXR) with natural and synthetic ligands. Curr. Top. Med. Chem. 2014, 14, 2129-2142; c) Gioiello, A., Rosatelli, E., Nuti, R., Macchiarulo, A., Pellicciari, R. Patented TGR5 modulators: a review (2006 - present). Expert Opin. Ther. Pat. 2012, 22, 1399-1414. Based on the abovementioned observations we introduced azobenzene in the 3- or 5-position of 4-PHP, which led to a series of photoswitchable analogues. The analogues behave similar to azobenzene with cis/trans isomerization upon irradiation with UV-Vis light and thermal relaxation over time.

Pharmacological characterization at GABAARs was performed in a [3H]muscimol binding assay using rat brain cortical membranes. Binding affinities of all dark adapted analogues were in the low micro- to low nanomolar range at native rat GABAARs.

References 1) Foster AC, Kemp JA. Glutamate- and GABA-based CNS therapeutics. Curr. Opin. Pharmacol. 2006, 6, 7-17. 2) Beharry AA, Woolley GA. Azobenzene photoswitches for biomolecules. Chem. Soc. Rev. 2011, 40, 4422–4437. 3) Krall J, Jensen CH, Sørensen TE, Nielsen B, Jensen AA, Sander T, Balle T, Frølund B. Exploring the orthosteric binding site of the γ-aminobutyric acid type A receptor using 4-(Piperidin-4-yl)-1-hydroxypyrazoles 3- or 5-imidazolyl substituted: design, synthesis, and pharmacological evaluation. J. Med. Chem. 2013, 56, 6536-6540. 4) Møller, H. A.; Sander, T.; Kristensen, J. L.; Nielsen, B.; Krall, J.; Bergmann, M. L.; Christiansen, B.; Balle, T.; Jensen, A. A.; Frølund, B. Novel 4-(Piperidin-4-yl)-1-hydroxypyrazoles as γ-Aminobutyric AcidA Receptor Ligands: Synthesis, Pharmacology, and Structure−Activity Relationships. J. Med. Chem. 2010, 53, 3417-3421.

49 FP13 FP14 LEAD OPTIMISATION OF A NOVEL CLASS OF SYNTHESIS AND BIOLOGICAL EVALUATION OF DIAROMATIC TRYPANOSOMACIDAL AGENTS GUANIDINIUM-LIKE DERIVATIVES AS PROTEIN KINASE INHIBITORS FOR THE TREATMENT OF CANCER Harriet Newson (1), Stephanie Russell (1), Raphaël Rahmani (2), Amy Jones (3), Kevin Neilde (2,4), Lori Ferrins (2), Susan Charman (2,5), Albane Kessler (4), Vicky Avery (3), Jonathan Baell (2), Matthew Viola Previtali, Isabel Rozas Piggott (1) School of Chemistry, 1) School of Chemistry and Biochemistry, The University of Western Australia, Perth 6009, Australia Trinity Biomedical Sciences Institute, 2) Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, 152-160, Pearse St., Trinity College Dublin, Australia Dublin 2, Ireland 3) Eskitis Institute for Cell and Molecular Therapies, Griffith University, Brisbane Innovation Park, Don Young Road, Nathan, Queensland 4111, Australia 4) GlaxoSmithKline, Tres Cantos, Spain 5) Centre for Drug Candidate Optimisation, Monash University, Parkville, Victoria 3052, Australia The Ras–Raf–MEK–ERK pathway (ERK signalling) is an evolutionary conserved kinases’ cascade that transmits signals from cell surface receptors to promote cell proliferation and survival. In physiological conditions, ERK signalling is tightly controlled by feedback loops at multiple levels, which are essential for maintaining regulated cell growth and homeostasis.1Components of the ERK signalling cascade are frequently mutated in cancer with approximately 1/3 of human tumours expressing a constitutively activated mutant form of RAS2 and approximately 8% of tumours expressing an activated form of BRAF. These findings prompted the development of small-molecule inhibitors targeting components of ERK signalling to be used as cancer therapeutics.1Previous studies in Rozas’ group identified compound 1 (Figure 1) has a kinase inhibitor capable of inhibiting MAPK/Erk pathway through a type-III allosteric mechanism. As such, compound 1 provided a new chemical entity for further refinement of kinase selectivity and potency that may not face the limitations of ATP-competitive inhibition that challenge the translation of current generation PKIs towards the clinic.2 Previous and on-going molecular-modelling studies point toward the idea that this compound forms hydrogen-bond/electrostatic interactions with one of the ATP-phosphates through the guanidinium moiety thus 3 positioning the lipophilic (4-Cl-3-CF3)-Ph group in a hydrophobic pocket of the enzyme.

Human African Trypanosomiasis (HAT) and Chagas Disease, caused by the protozoan parasites Trypanosoma brucei and T. cruzi, respectively, cause significant suffering and mortality in some of the poorest regions of the world. Current treatment regimens are not suited to third world conditions, have low efficacy and high toxicity. New, orally available and safe drugs for these parasitic infections are urgently required. In this presentation, the hit to lead optimization of a novel class of potent, selective and broad spectrum trypansomacidal agents,1 based on the high-throughput screening hit 1,2 will be reported. Pharmacokinetic characteristics and mode of action studies of the most potent compounds will also be discussed.

References 1) Heterocyclic compounds and use of same”, Jonathan Baell, Matthew Piggott, Stephanie Russell, Arthur Toynton, Raphael Rahmani, Lori Ferrins, Nghi Nguyen; PCT Int. Appl. (2015), WO 2015172196 A1 20151119 2) Sykes, M. L.; Baell, J. B.; Kaiser, M.; Chatelain, E.; Moawad, S. R.; Ganame, D.; Ioset, J.-R.; Avery, V. M. PLoS Negl Trop Dis 2012, 6, e1896

Figure 1: Optimization cycle of new guanidine-based kinases’ inhibitors with anticancer activity. New guanidinium aromatic derivatives (e.g. 2 and 3) have now been synthesized to clearly identify the structural motifs that determine the allosteric inhibition of the MAPK/Erk pathway. The cytotoxic effect of these new derivatives has already been assessed, with a 3/4-fold improvement (IC50 = 3.08 µM for 2 and 1.53 µM for 3) compared to 1 (IC50 = 9.72 µM). Moreover, their role as apoptotic inducers and inhibitors of different kinases will be evaluated by performing several biochemical assays.

References 1) Samatar, A. A.; Poulikakos, P. I. Nat. Rev. Drug Discov. 2014, 13, 928. 2) Diez-Cecilia, E.; Carson, R.; Kelly, B.; van Schaeybroeck, S.; Murray, J. T.; Rozas, I. Bioorg. Med. Chem. Lett. 2015, 25, 4287. 3) Diez-Cecilia, E.; Kelly, B.; Perez, C.; Zisterer, D. M.; Nevin, D. K.; Lloyd, D. G.; Rozas, I. Eur. J. Med. Chem. 2014, 81, 427. 50 FP13 FP14 LEAD OPTIMISATION OF A NOVEL CLASS OF SYNTHESIS AND BIOLOGICAL EVALUATION OF DIAROMATIC TRYPANOSOMACIDAL AGENTS GUANIDINIUM-LIKE DERIVATIVES AS PROTEIN KINASE INHIBITORS FOR THE TREATMENT OF CANCER Harriet Newson (1), Stephanie Russell (1), Raphaël Rahmani (2), Amy Jones (3), Kevin Neilde (2,4), Lori Ferrins (2), Susan Charman (2,5), Albane Kessler (4), Vicky Avery (3), Jonathan Baell (2), Matthew Viola Previtali, Isabel Rozas Piggott (1) School of Chemistry, 1) School of Chemistry and Biochemistry, The University of Western Australia, Perth 6009, Australia Trinity Biomedical Sciences Institute, 2) Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, 152-160, Pearse St., Trinity College Dublin, Australia Dublin 2, Ireland 3) Eskitis Institute for Cell and Molecular Therapies, Griffith University, Brisbane Innovation Park, Don Young Road, Nathan, Queensland 4111, Australia 4) GlaxoSmithKline, Tres Cantos, Spain 5) Centre for Drug Candidate Optimisation, Monash University, Parkville, Victoria 3052, Australia The Ras–Raf–MEK–ERK pathway (ERK signalling) is an evolutionary conserved kinases’ cascade that transmits signals from cell surface receptors to promote cell proliferation and survival. In physiological conditions, ERK signalling is tightly controlled by feedback loops at multiple levels, which are essential for maintaining regulated cell growth and homeostasis.1Components of the ERK signalling cascade are frequently mutated in cancer with approximately 1/3 of human tumours expressing a constitutively activated mutant form of RAS2 and approximately 8% of tumours expressing an activated form of BRAF. These findings prompted the development of small-molecule inhibitors targeting components of ERK signalling to be used as cancer therapeutics.1Previous studies in Rozas’ group identified compound 1 (Figure 1) has a kinase inhibitor capable of inhibiting MAPK/Erk pathway through a type-III allosteric mechanism. As such, compound 1 provided a new chemical entity for further refinement of kinase selectivity and potency that may not face the limitations of ATP-competitive inhibition that challenge the translation of current generation PKIs towards the clinic.2 Previous and on-going molecular-modelling studies point toward the idea that this compound forms hydrogen-bond/electrostatic interactions with one of the ATP-phosphates through the guanidinium moiety thus 3 positioning the lipophilic (4-Cl-3-CF3)-Ph group in a hydrophobic pocket of the enzyme.

Human African Trypanosomiasis (HAT) and Chagas Disease, caused by the protozoan parasites Trypanosoma brucei and T. cruzi, respectively, cause significant suffering and mortality in some of the poorest regions of the world. Current treatment regimens are not suited to third world conditions, have low efficacy and high toxicity. New, orally available and safe drugs for these parasitic infections are urgently required. In this presentation, the hit to lead optimization of a novel class of potent, selective and broad spectrum trypansomacidal agents,1 based on the high-throughput screening hit 1,2 will be reported. Pharmacokinetic characteristics and mode of action studies of the most potent compounds will also be discussed.

References 1) Heterocyclic compounds and use of same”, Jonathan Baell, Matthew Piggott, Stephanie Russell, Arthur Toynton, Raphael Rahmani, Lori Ferrins, Nghi Nguyen; PCT Int. Appl. (2015), WO 2015172196 A1 20151119 2) Sykes, M. L.; Baell, J. B.; Kaiser, M.; Chatelain, E.; Moawad, S. R.; Ganame, D.; Ioset, J.-R.; Avery, V. M. PLoS Negl Trop Dis 2012, 6, e1896

Figure 1: Optimization cycle of new guanidine-based kinases’ inhibitors with anticancer activity. New guanidinium aromatic derivatives (e.g. 2 and 3) have now been synthesized to clearly identify the structural motifs that determine the allosteric inhibition of the MAPK/Erk pathway. The cytotoxic effect of these new derivatives has already been assessed, with a 3/4-fold improvement (IC50 = 3.08 µM for 2 and 1.53 µM for 3) compared to 1 (IC50 = 9.72 µM). Moreover, their role as apoptotic inducers and inhibitors of different kinases will be evaluated by performing several biochemical assays.

References 1) Samatar, A. A.; Poulikakos, P. I. Nat. Rev. Drug Discov. 2014, 13, 928. 2) Diez-Cecilia, E.; Carson, R.; Kelly, B.; van Schaeybroeck, S.; Murray, J. T.; Rozas, I. Bioorg. Med. Chem. Lett. 2015, 25, 4287. 3) Diez-Cecilia, E.; Kelly, B.; Perez, C.; Zisterer, D. M.; Nevin, D. K.; Lloyd, D. G.; Rozas, I. Eur. J. Med. Chem. 2014, 81, 427. 51 FP15 FP16 DEVELOPMENT OF NOVEL 1,4-BENZODIAZEPINES AS DETERMINING THE BINDING MODE OF ANTITRYPANOSOMAL AGENTS 3-(1-ALKYL-1H-IMIDAZOL-5-YL)-1H-INDOLES

Santo Previti (1), Roberta Ettari (1), Santina Maiorana (1), Sandro Cosconati (2), Tanja Schirmeister (3), Adam Hogendorf (1,2), Agata Hogendorf (1), Rafał Kurczab (1), Grzegorz Satała (1), Tomasz Lenda (3), Silvana Grasso (1), Maria Zappalà (1) Gniewomir Latacz (4), Ryszard Bugno (1), Jakub Staroń (1), Andrzej J. Bojarski (1)

1) Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale 1) Department of Medicinal Chemistry, Institute of Pharmacology, Polish Academy of Sciences, Annunziata, 98168 Messina, Italy 12 Smętna Street, 31-343 Krakow, Poland 2) DiSTABiF, Second University of Naples, Via Vivaldi 43, 81100 Caserta, Italy 2) Faculty of Chemistry, Jagiellonian University, 3 Ingardena Street, 30-060 Krakow, Poland 3) Institute of Pharmacy and Biochemistry, University of Mainz, Staudinger Weg 5, 55099 Mainz, Germany 3) Department of Neuropsychopharmacology Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna Street, 31-343 Krakow, Poland 4) Jagiellonian University Medical College, Department of Technology and Biotechnology of Drugs, 9 Medyczna Street, 30-688 Kraków, Poland

Human African Trypanosomiasis (HAT) is a parasitic disease of sub-Saharan Africa, caused by two subspecies of protozoa of Trypanosoma genus: T. brucei gambiense and T. brucei rhodesiense, which cause the chronic and acute form of HAT respectively [1]. Both parasitic subspecies produce two main stages of HAT. Stage 1 (also Non-basic aminergic GPCR ligands have recently emerged as pharmacological rule breakers. Despite lacking the named hemolymphatic stage) initiates after the tsetse fly bite and can persist several weeks. During this stage, capability of forming a salt bridge (strong, charge assisted hydrogen bond) with Asp3.32 such chemical entities the parasite initially lives within the bloodstream and then migrates to the lymph nodes, spleen, and spinal fluid, have been shown to bind to 5-HT2AR1 and later to 5-HT6 and 5-HT1B receptors with nanomolar and causing symptoms like rash, fever, muscle aches, and fatigue. If left untreated, stage 1 HAT evolves into the subnanomolar affinities.2,3 Interestingly, there have been no published examples of low-basicity ligands of the neurological stage (stage 2 HAT), during which the parasite crosses the blood brain barrier, leading to serious 5-HT7 receptor.4 mental deterioration, sleep disturbances, coma and death. Unfortunately, an effective vaccine has not been We have developed a 32-member series of 5-HT7 ligands, which are notably the first examples of low-basicity developed yet, because of the high degree of antigenic variation expressed by the glycoprotein forming their 5-HT7 receptor agonists and one of the very few low-basicity agonists of an aminergic receptor. surface coat. Docking to homology models revealed a possible binding mode for the compounds, indicating an indole In this context, in the last decade our research group has been actively involved in the development of several hydrogen bond with Asp3.32 and -Arg6.58 interaction. peptidomimetic parasitic cysteine protease inhibitors as antimalarial and antitrypanosomal agents [2]. Among all Despite different binding modes (Fig. 1), the 5-HT7R SAR of tryptamines and are very similar due to the synthesized inhibitors, one of the most promising antitrypanosomal agent is represented by the Michael involvement of indole 5-substitutent interaction with Ser5.42 (or Thr5.39) in both series. Halogen bonding acceptor 1 (Fig.1), characterized by the presence of a 1,4-benzodiazepine nucleus as recognition moiety and by a explains the very high binding affinity of 5-haloindole derivatives of the title compounds. hydroxymethyl group at C3 of the scaffold, used to tie an adamantyl nucleus able to create additional interactions with the S3 pocket of the target enzyme. Ester 1 is a potent rhodesain inhibitor (Ki=2.6 nM), endowed with a good antitrypanosomal activity (IC50=4.8 µM) [3]. While previous lead optimization efforts were instrumental to optimize the activity profile of this inhibitor, however, they were not helpful in enhancing its putative pharmacokinetic properties. Most probably, its high molecular weight is one of the most evident structural feature that should hamper a possible development of ester 1 as chemotherapeutic agent, together with its high clogP that is a bigger challenge to address. Starting from these considerations, and with the aim to improve its physicochemical properties, thus obtaining new drug-like antiparasitic agents, better matching the Lipinski's Rule of 5, we designed novel simplified 1,4-benzodiazepines 2 (Fig. 1), endowed with a Michael acceptor portion. To design new analogues of compound 1 we first decided to take advantage of the computational prediction of its pharmacokinetic (PK) parameters. Therefore, we decided to start from our lead compound 1 to attain a set of structural simplification aimed at optimizing its PK parameters, while maintaining its antiparasitic activity. Among all the synthesized compounds, we identified a novel 1,4-benzodiazepine, with an IC50 value against T. b. brucei in the low micromolar range and with lack of cytotoxicity towards mammalian cells, to be considered as a promising lead compound to be further developed for HAT treatment.

The study was partially supported by the Polish-Norwegian Research Programme operated by the National Centre for Research and Development under the Norwegian Financial Mechanism 2009–2014 in the frame of the Project PLATFORMex (Pol-Nor/198887/73/2013) Figure 1. Structure of lead compound 1 and of novel 1,4-benzodiazepines 2 References References 1) Ladduwahetty, T et. al. Bioorg. Med. Chem. Lett. 2006, 16, 3201-3204 2) Harris, R. N. 3rd et. al. Bioorg. Med. Chem. Lett. 2010, 20, 3436-3440 1) Cox, F. E. G. History of sleeping sickness (African trypanosomiasis). Infect. Dis. Clin. North Am. 2004, 18, 231−245. 3) Nugiel, D. A.; et. al. J. Med. Chem. 2010, 53, 1876-1880 2) Ettari, R.; Pinto, A.; Tamborini, L.; Angelo, I. C.; Grasso, S.; Zappalà, M.; Capodicasa, N.; Yzeiraj, L.; Gruber, E.; 4) Bento, A. P. et. al. Nucleic Acids Res. 2014, 42, 1083-1090 Aminake, M. N.; Pradel, G.; Schirmeister, T.; De Micheli, C.; Conti, P. ChemMedChem 2014, 9, 1817 3) Bova, F.; Ettari, R.; Micale, N.; Carnovale, C.; Schirmeister, T.; Gelhaus, C.; Leippe, M.; Grasso, S.; Zappalà, M.; Bioorg. Med. Chem. 2010, 18, 4928. 52 FP15 FP16 DEVELOPMENT OF NOVEL 1,4-BENZODIAZEPINES AS DETERMINING THE BINDING MODE OF ANTITRYPANOSOMAL AGENTS 3-(1-ALKYL-1H-IMIDAZOL-5-YL)-1H-INDOLES

Santo Previti (1), Roberta Ettari (1), Santina Maiorana (1), Sandro Cosconati (2), Tanja Schirmeister (3), Adam Hogendorf (1,2), Agata Hogendorf (1), Rafał Kurczab (1), Grzegorz Satała (1), Tomasz Lenda (3), Silvana Grasso (1), Maria Zappalà (1) Gniewomir Latacz (4), Ryszard Bugno (1), Jakub Staroń (1), Andrzej J. Bojarski (1)

1) Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale 1) Department of Medicinal Chemistry, Institute of Pharmacology, Polish Academy of Sciences, Annunziata, 98168 Messina, Italy 12 Smętna Street, 31-343 Krakow, Poland 2) DiSTABiF, Second University of Naples, Via Vivaldi 43, 81100 Caserta, Italy 2) Faculty of Chemistry, Jagiellonian University, 3 Ingardena Street, 30-060 Krakow, Poland 3) Institute of Pharmacy and Biochemistry, University of Mainz, Staudinger Weg 5, 55099 Mainz, Germany 3) Department of Neuropsychopharmacology Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna Street, 31-343 Krakow, Poland 4) Jagiellonian University Medical College, Department of Technology and Biotechnology of Drugs, 9 Medyczna Street, 30-688 Kraków, Poland

Human African Trypanosomiasis (HAT) is a parasitic disease of sub-Saharan Africa, caused by two subspecies of protozoa of Trypanosoma genus: T. brucei gambiense and T. brucei rhodesiense, which cause the chronic and acute form of HAT respectively [1]. Both parasitic subspecies produce two main stages of HAT. Stage 1 (also Non-basic aminergic GPCR ligands have recently emerged as pharmacological rule breakers. Despite lacking the named hemolymphatic stage) initiates after the tsetse fly bite and can persist several weeks. During this stage, capability of forming a salt bridge (strong, charge assisted hydrogen bond) with Asp3.32 such chemical entities the parasite initially lives within the bloodstream and then migrates to the lymph nodes, spleen, and spinal fluid, have been shown to bind to 5-HT2AR1 and later to 5-HT6 and 5-HT1B receptors with nanomolar and causing symptoms like rash, fever, muscle aches, and fatigue. If left untreated, stage 1 HAT evolves into the subnanomolar affinities.2,3 Interestingly, there have been no published examples of low-basicity ligands of the neurological stage (stage 2 HAT), during which the parasite crosses the blood brain barrier, leading to serious 5-HT7 receptor.4 mental deterioration, sleep disturbances, coma and death. Unfortunately, an effective vaccine has not been We have developed a 32-member series of 5-HT7 ligands, which are notably the first examples of low-basicity developed yet, because of the high degree of antigenic variation expressed by the glycoprotein forming their 5-HT7 receptor agonists and one of the very few low-basicity agonists of an aminergic receptor. surface coat. Docking to homology models revealed a possible binding mode for the compounds, indicating an indole In this context, in the last decade our research group has been actively involved in the development of several hydrogen bond with Asp3.32 and imidazole-Arg6.58 interaction. peptidomimetic parasitic cysteine protease inhibitors as antimalarial and antitrypanosomal agents [2]. Among all Despite different binding modes (Fig. 1), the 5-HT7R SAR of tryptamines and imidazoles are very similar due to the synthesized inhibitors, one of the most promising antitrypanosomal agent is represented by the Michael involvement of indole 5-substitutent interaction with Ser5.42 (or Thr5.39) in both series. Halogen bonding acceptor 1 (Fig.1), characterized by the presence of a 1,4-benzodiazepine nucleus as recognition moiety and by a explains the very high binding affinity of 5-haloindole derivatives of the title compounds. hydroxymethyl group at C3 of the scaffold, used to tie an adamantyl nucleus able to create additional interactions with the S3 pocket of the target enzyme. Ester 1 is a potent rhodesain inhibitor (Ki=2.6 nM), endowed with a good antitrypanosomal activity (IC50=4.8 µM) [3]. While previous lead optimization efforts were instrumental to optimize the activity profile of this inhibitor, however, they were not helpful in enhancing its putative pharmacokinetic properties. Most probably, its high molecular weight is one of the most evident structural feature that should hamper a possible development of ester 1 as chemotherapeutic agent, together with its high clogP that is a bigger challenge to address. Starting from these considerations, and with the aim to improve its physicochemical properties, thus obtaining new drug-like antiparasitic agents, better matching the Lipinski's Rule of 5, we designed novel simplified 1,4-benzodiazepines 2 (Fig. 1), endowed with a Michael acceptor portion. To design new analogues of compound 1 we first decided to take advantage of the computational prediction of its pharmacokinetic (PK) parameters. Therefore, we decided to start from our lead compound 1 to attain a set of structural simplification aimed at optimizing its PK parameters, while maintaining its antiparasitic activity. Among all the synthesized compounds, we identified a novel 1,4-benzodiazepine, with an IC50 value against T. b. brucei in the low micromolar range and with lack of cytotoxicity towards mammalian cells, to be considered as a promising lead compound to be further developed for HAT treatment.

The study was partially supported by the Polish-Norwegian Research Programme operated by the National Centre for Research and Development under the Norwegian Financial Mechanism 2009–2014 in the frame of the Project PLATFORMex (Pol-Nor/198887/73/2013) Figure 1. Structure of lead compound 1 and of novel 1,4-benzodiazepines 2 References References 1) Ladduwahetty, T et. al. Bioorg. Med. Chem. Lett. 2006, 16, 3201-3204 2) Harris, R. N. 3rd et. al. Bioorg. Med. Chem. Lett. 2010, 20, 3436-3440 1) Cox, F. E. G. History of sleeping sickness (African trypanosomiasis). Infect. Dis. Clin. North Am. 2004, 18, 231−245. 3) Nugiel, D. A.; et. al. J. Med. Chem. 2010, 53, 1876-1880 2) Ettari, R.; Pinto, A.; Tamborini, L.; Angelo, I. C.; Grasso, S.; Zappalà, M.; Capodicasa, N.; Yzeiraj, L.; Gruber, E.; 4) Bento, A. P. et. al. Nucleic Acids Res. 2014, 42, 1083-1090 Aminake, M. N.; Pradel, G.; Schirmeister, T.; De Micheli, C.; Conti, P. ChemMedChem 2014, 9, 1817 3) Bova, F.; Ettari, R.; Micale, N.; Carnovale, C.; Schirmeister, T.; Gelhaus, C.; Leippe, M.; Grasso, S.; Zappalà, M.; Bioorg. Med. Chem. 2010, 18, 4928. 53 FP17 FP18 TARGETING THE HUMAN DIHYDROOROTATE DEHYDROGENASE TOWARDS THE ELUCIDATION OF THE MECHANISM OF ACTION (hDHODH) BY A SCAFFOLD HOPPING BIOISOSTERIC APPROACH OF SMALL MOLECULE UPREGULATORS OF UTROPHIN USING USING HYDROXYLATED PENTAATOMIC HETEROCYCLES CHEMICAL PROTEOMICS

Stefano Sainas (1), Agnese Chiara Pippione (1), Irene Maria Carnovale (1), Alessandro Giraudo (1), Aini Vuorinen (1), Noelia Araujo (1), James Donald (1), Fernando Martinez (1), Josh Milner (1), Nicky Marta Giorgis (1), Rodolpho B. Braga (4), Carolina H. Andrade (4), Noemi Villella (1), Marco Piccinini Willis (1), Ceri Cairnduff (1), Graham Wynne (1), Stephen Davies (1), Huijia Chen (2), Rebecca (2), Elisa Lupino (2), Rosmarie Friemann (5), Salam Al-Kadaraghi (3), Donatella Boschi (1), Marco L. Fairclough (2), Simon Guiraud (2), Arran Babbs (2), Ben Edwards (2), Nandini Shah (2), Sarah Squire Lolli (1) (2), Kathryn Pugh (3), Kilian Huber (3), Kay Davies (2), Angela Russell (1,4)

1) MEDSynth at Department of Science and Drug Technology, University of Torino, via Giuria 9, 10125 Torino (Italy) 1) Department of Chemistry, University of Oxford, Chemistry Research Laboratory, 12 Mansfield Road, Oxford, OX1 3TA 2) Department of Oncology, University of Torino, via Michelangelo 27/B, 10125 Torino (Italy) 2) MRC Functional Genomics Unit, University of Oxford, South Parks Road, Oxford, OX1 3PT 3) Department of Biochemistry and Structural Biology, University of Lund, (Sweden). 3) Target Discovery Institute, University of Oxford, Old Road Campus, Oxford, OX3 7FZ 4) LabMol, Faculty of Pharmacy, Federal University of Goias, 74605-170 Goiania (Brazil) 4) Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT 5) Department of Chemistry and Molecular Biology Biochemistry and Biophysics, University of Gothenburg, (Sweden).

Duchenne Muscular Dystrophy (DMD) is an X-linked recessive and progressive muscle-wasting disease caused The human isoform of the dihydroorotate dehydrogenase enzyme (hDHODH) catalyzes the fourth step of the de by lack of the cytoskeletal protein dystrophin. There is currently no cure for DMD. Although various approaches novo pyrimidine synthesis, a biosynthetic pathway enhanced in proliferating cells such as activated (e.g. exon skipping, read through of stop codons, gene therapy) are being developed none of them have yet T-lymphocytes, and cancer cells. The efficacy of hDHODH inhibitors in the treatment of rheumatoid arthritis and multiple sclerosis has been evaluated, and leflunomide (Arava®) and its active metabolite teriflunomide shown efficacy in man and gained FDA approval. Our strategy to deliver an effective therapy for DMD is to (Aubagio®) have been already approved for therapy. Brequinar is another well-studied hDHODH inhibitor, but develop an oral small molecule upregulator that replaces and compensates for the missing dystrophin with it was unsuccessfully evaluated against a number of tumor categories due to several drug-related side effects.1 utrophin, its autosomal paralogue. This will be applicable to all patients regardless of their dystrophin mutation Inside the inhibitor-binding site, conventionally divided into five subsites, two different binding-modes have and will target skeletal muscle, heart and diaphragm. In partnership with Summit Therapeutics, Ezutromid (SMT been described in literature.2 In recent years our group at DSTF has directed its efforts towards the investigation C1100), a small molecule utrophin modulator that reduces dystrophic symptoms in the mdx mouse,1,2 is in a of a general tool able to mimic the carboxyl group. We focused our attention on a pool of over ten acidic Phase 2 clinical trial.3,4 hydroxylated pentatomic heterocyclic systems. The result of such work was the development of a flexible tool capable of adapting to the requirements of different targets, both in terms of acidity as well as general Ezutromid demonstrates proof of principle for the strategy, but we still need to rapidly parallel track follow-on chemio-physical profiles. Starting from structural information from both brequinar and teriflunomide, a scaffold compounds which have better efficacy, pharmaceutical properties and/or complementary mechanisms to hopping approach was applied using our heterocyclic panel affording a new series of products able to establish maximise the success of the utrophin modulation approach. We have discovered novel utrophin modulator additional interactions with subsites 3 and 4. The general model of these compounds (1) is a hydroxylated 3 chemotypes using an improved in vitro screening assay based on immortalised myoblasts from the heterocycle linked to a biphenyl system through an amide bond. According to molecular modeling studies, the mdx deprotonated acidic moiety should interact with residues Arg136 and Gln47 of subsite 2 (Brequinar-like dystrophin-null, utrophin luciferase knock-in mouse (LU ). Multiple structural classes which significantly binding-mode2), while the biphenyl system may establish lipophilic interactions with residues of subsites 1 and 5 modulate utrophin expression in both murine and human DMD myoblasts have been identified and are now (Figure a). New compounds showed inhibitory activity on recombinant hDHODH with IC50 values in the being optimised. However the precise mechanism by which these small molecules increase levels of utrophin is nanomolar range and inhibition of human T-cell proliferation comparable to brequinar and teriflunomide. Our not understood. Importantly initial evidence suggests that some of these small molecules modulate utrophin theoretical design, modeling, synthesis, SAR and biological assays, as well as cell viability, proliferation, transcription through an alternative regulatory mechanism to Ezutromid. cytotoxicity and immunosuppression results are presented in detail. To discover the molecular mechanism of action of these utrophin modulators we have conducted structure-activity relationship studies within one of the novel compound classes and as a result pull-down chemical probes have been prepared for chemoproteomic analyses. Finding optimal conditions for the pull-down assay is a challenging process because the cellular location of the potential target(s) and binding affinity of the probe to target(s) remains unknown. To overcome these problems and poor cell-permeability of biotin-tagged probes we have prepared improved dual tagged probes which incorporate both a photoaffinity group (diazirine) and an alkyne tag which retain comparable activity to their unlabelled counterparts. By applying Cu(I)-catalysed azide-alkyne “click chemistry” the biotin tag is introduced after cell lysis. Pull-down experiments and cellular thermal shift proteomics analysis5 using the improved dual tagged probes are underway.

References 1) J. Med. Chem., 2011, 54, 3241-3250 2) PLoS One, 2011, 6, e19189 3) J Clin Pharmacol, 2015, 55, 698-707 4) PLoS One, 2016, 11(4), e0152840 5) Nat. Methods, 2015, 12, 1055-1057

Fig. 1. a) The lipophilic patch of the hDHODH binding site in complex with brequinar (green) and our best compound (yellow). b) General structure of new inhibitors (1).

References 1) Munier-Lehmann H, Vidalain PO, Tangy F, Janin YL. On dihydroorotate dehydrogenases and their inhibitors and uses. J Med Chem. 2013;56:3148-3167. doi:10.1021/jm301848w. 2) Baumgartner R, Walloschek M, Kralik M, et al. Dual binding mode of a novel series of DHODH inhibitors. J Med Chem. 2006;49:1239-1247. doi:10.1021/jm0506975. 3) Lolli ML, Giorgis M, Tosco P, Foti A, Fruttero R, Gasco A. New inhibitors of dihydroorotate dehydrogenase (DHODH) based on the 4-hydroxy-1,2,5-oxadiazol-3-yl (hydroxyfurazanyl) scaffold. Eur J Med Chem. 2012;49:102-109. doi:10.1016/j.ejmech.2011.12.038. 54 FP17 FP18 TARGETING THE HUMAN DIHYDROOROTATE DEHYDROGENASE TOWARDS THE ELUCIDATION OF THE MECHANISM OF ACTION (hDHODH) BY A SCAFFOLD HOPPING BIOISOSTERIC APPROACH OF SMALL MOLECULE UPREGULATORS OF UTROPHIN USING USING HYDROXYLATED PENTAATOMIC HETEROCYCLES CHEMICAL PROTEOMICS

Stefano Sainas (1), Agnese Chiara Pippione (1), Irene Maria Carnovale (1), Alessandro Giraudo (1), Aini Vuorinen (1), Noelia Araujo (1), James Donald (1), Fernando Martinez (1), Josh Milner (1), Nicky Marta Giorgis (1), Rodolpho B. Braga (4), Carolina H. Andrade (4), Noemi Villella (1), Marco Piccinini Willis (1), Ceri Cairnduff (1), Graham Wynne (1), Stephen Davies (1), Huijia Chen (2), Rebecca (2), Elisa Lupino (2), Rosmarie Friemann (5), Salam Al-Kadaraghi (3), Donatella Boschi (1), Marco L. Fairclough (2), Simon Guiraud (2), Arran Babbs (2), Ben Edwards (2), Nandini Shah (2), Sarah Squire Lolli (1) (2), Kathryn Pugh (3), Kilian Huber (3), Kay Davies (2), Angela Russell (1,4)

1) MEDSynth at Department of Science and Drug Technology, University of Torino, via Giuria 9, 10125 Torino (Italy) 1) Department of Chemistry, University of Oxford, Chemistry Research Laboratory, 12 Mansfield Road, Oxford, OX1 3TA 2) Department of Oncology, University of Torino, via Michelangelo 27/B, 10125 Torino (Italy) 2) MRC Functional Genomics Unit, University of Oxford, South Parks Road, Oxford, OX1 3PT 3) Department of Biochemistry and Structural Biology, University of Lund, (Sweden). 3) Target Discovery Institute, University of Oxford, Old Road Campus, Oxford, OX3 7FZ 4) LabMol, Faculty of Pharmacy, Federal University of Goias, 74605-170 Goiania (Brazil) 4) Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT 5) Department of Chemistry and Molecular Biology Biochemistry and Biophysics, University of Gothenburg, (Sweden).

Duchenne Muscular Dystrophy (DMD) is an X-linked recessive and progressive muscle-wasting disease caused The human isoform of the dihydroorotate dehydrogenase enzyme (hDHODH) catalyzes the fourth step of the de by lack of the cytoskeletal protein dystrophin. There is currently no cure for DMD. Although various approaches novo pyrimidine synthesis, a biosynthetic pathway enhanced in proliferating cells such as activated (e.g. exon skipping, read through of stop codons, gene therapy) are being developed none of them have yet T-lymphocytes, and cancer cells. The efficacy of hDHODH inhibitors in the treatment of rheumatoid arthritis and multiple sclerosis has been evaluated, and leflunomide (Arava®) and its active metabolite teriflunomide shown efficacy in man and gained FDA approval. Our strategy to deliver an effective therapy for DMD is to (Aubagio®) have been already approved for therapy. Brequinar is another well-studied hDHODH inhibitor, but develop an oral small molecule upregulator that replaces and compensates for the missing dystrophin with it was unsuccessfully evaluated against a number of tumor categories due to several drug-related side effects.1 utrophin, its autosomal paralogue. This will be applicable to all patients regardless of their dystrophin mutation Inside the inhibitor-binding site, conventionally divided into five subsites, two different binding-modes have and will target skeletal muscle, heart and diaphragm. In partnership with Summit Therapeutics, Ezutromid (SMT been described in literature.2 In recent years our group at DSTF has directed its efforts towards the investigation C1100), a small molecule utrophin modulator that reduces dystrophic symptoms in the mdx mouse,1,2 is in a of a general tool able to mimic the carboxyl group. We focused our attention on a pool of over ten acidic Phase 2 clinical trial.3,4 hydroxylated pentatomic heterocyclic systems. The result of such work was the development of a flexible tool capable of adapting to the requirements of different targets, both in terms of acidity as well as general Ezutromid demonstrates proof of principle for the strategy, but we still need to rapidly parallel track follow-on chemio-physical profiles. Starting from structural information from both brequinar and teriflunomide, a scaffold compounds which have better efficacy, pharmaceutical properties and/or complementary mechanisms to hopping approach was applied using our heterocyclic panel affording a new series of products able to establish maximise the success of the utrophin modulation approach. We have discovered novel utrophin modulator additional interactions with subsites 3 and 4. The general model of these compounds (1) is a hydroxylated 3 chemotypes using an improved in vitro screening assay based on immortalised myoblasts from the heterocycle linked to a biphenyl system through an amide bond. According to molecular modeling studies, the mdx deprotonated acidic moiety should interact with residues Arg136 and Gln47 of subsite 2 (Brequinar-like dystrophin-null, utrophin luciferase knock-in mouse (LU ). Multiple structural classes which significantly binding-mode2), while the biphenyl system may establish lipophilic interactions with residues of subsites 1 and 5 modulate utrophin expression in both murine and human DMD myoblasts have been identified and are now (Figure a). New compounds showed inhibitory activity on recombinant hDHODH with IC50 values in the being optimised. However the precise mechanism by which these small molecules increase levels of utrophin is nanomolar range and inhibition of human T-cell proliferation comparable to brequinar and teriflunomide. Our not understood. Importantly initial evidence suggests that some of these small molecules modulate utrophin theoretical design, modeling, synthesis, SAR and biological assays, as well as cell viability, proliferation, transcription through an alternative regulatory mechanism to Ezutromid. cytotoxicity and immunosuppression results are presented in detail. To discover the molecular mechanism of action of these utrophin modulators we have conducted structure-activity relationship studies within one of the novel compound classes and as a result pull-down chemical probes have been prepared for chemoproteomic analyses. Finding optimal conditions for the pull-down assay is a challenging process because the cellular location of the potential target(s) and binding affinity of the probe to target(s) remains unknown. To overcome these problems and poor cell-permeability of biotin-tagged probes we have prepared improved dual tagged probes which incorporate both a photoaffinity group (diazirine) and an alkyne tag which retain comparable activity to their unlabelled counterparts. By applying Cu(I)-catalysed azide-alkyne “click chemistry” the biotin tag is introduced after cell lysis. Pull-down experiments and cellular thermal shift proteomics analysis5 using the improved dual tagged probes are underway.

References 1) J. Med. Chem., 2011, 54, 3241-3250 2) PLoS One, 2011, 6, e19189 3) J Clin Pharmacol, 2015, 55, 698-707 4) PLoS One, 2016, 11(4), e0152840 5) Nat. Methods, 2015, 12, 1055-1057

Fig. 1. a) The lipophilic patch of the hDHODH binding site in complex with brequinar (green) and our best compound (yellow). b) General structure of new inhibitors (1).

References 1) Munier-Lehmann H, Vidalain PO, Tangy F, Janin YL. On dihydroorotate dehydrogenases and their inhibitors and uses. J Med Chem. 2013;56:3148-3167. doi:10.1021/jm301848w. 2) Baumgartner R, Walloschek M, Kralik M, et al. Dual binding mode of a novel series of DHODH inhibitors. J Med Chem. 2006;49:1239-1247. doi:10.1021/jm0506975. 3) Lolli ML, Giorgis M, Tosco P, Foti A, Fruttero R, Gasco A. New inhibitors of dihydroorotate dehydrogenase (DHODH) based on the 4-hydroxy-1,2,5-oxadiazol-3-yl (hydroxyfurazanyl) scaffold. Eur J Med Chem. 2012;49:102-109. doi:10.1016/j.ejmech.2011.12.038. 55 FP19 FP20 DEVELOPMENT OF AN ALBUMIN-BINDING LIGAND FOR NEAR-INFRARED (NIR) PHOTOTHERMAL THERAPY USING PROLONGING THE PLASMA HALF-LIFE OF PEPTIDE BIOCOMPATIBLE PALLADIUM NANOPARTICLES THERAPEUTICS Belén Rubio-Ruiz, Ana M. Pérez-López, Thomas Bray, Martin Lee, Jason T. Weiss, Alan Serrels, Asier Alessandro Zorzi, Christian Heinis Unciti-Broceta

Laboratory of Therapeutic Proteins and Peptides (LPPT) Edinburgh Cancer Research UK Centre, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Institute of Chemical Sciences and Engineering, EPFL, Avenue Forel 2, CH-1015 Lausanne, Switzerland Crewe Road South, Edinburgh EH4 2XR, UK

Peptide therapeutics applied intravenously are rapidly cleared from the blood circulation by renal filtration. The Photothermal therapy (PTT), also known as photothermal ablation or optical hyperthermia, has gained wide short half-life prevents their application to diseases that require drug exposure of several hours or days (1). An attention as a precise and minimally invasive alternative for cancer treatment. This therapeutic approach consists attractive strategy to hamper filtration of peptides in the kidneys is to tether them non-covalently to a long-lived of using near-infrared (NIR) light-absorbing agents to convert photons into heat for ablation of cancer cells. serum protein such as human albumin (2). Several albumin-binding ligands based on peptides or small molecules Although the unique light-to-heat conversion property of many types of metallic nanostructures has been 1 were developed but they suffer from relatively low affinities for human albumin as well as a poor solubility in explored for photothermal therapy application , their unclear biotoxicity limits their therapeutic usefulness. physiological buffers, reducing their potential application to peptide therapeutics. Palladium is a transition metal with remarkable catalytic, mechanic and electronic properties. Despite these 2 To overcome these limitations, a chimeric peptide-small molecule albumin ligand with low nanomolar affinity characteristics, the use of palladium nanostructures in the nanomedicine field is still widely under-explored . 3 0 for human, rat and rabbit albumin, a high solubility and a small size suitable for automated synthesis of complex Using a green method we have produced Pd nanoparticles (PdNP) with high NIR light absorbance and conjugates was successfully developed. Peptides conjugated to the tag retained their bioactivity and displayed biocompatibility in cancer cell culture. By NIR irradiation, we have mediated the in vitro thermolysis of A549 around a 30-fold increase in half-life in rats. lung carcinoma cells demonstrating its potential for cancer photothermal therapy.

Schematic structure of the albumin-binding ligand (tag) (left panel) and pharmacokinetics of a therapeutic References peptide and its conjugated format in rat plasma upon i.v. injection (right panel). 1) Cheng, L. et al. Chem. Rev. 2014, 114, 10869-10939. 2) Dumas, A., Couvreur P. Chem. Sci. 2015, 6, 2153-2157. 3) Zhang, A. et al. J. Mater. Chem. A. 2014, 2, 1369- 1374. References 1) Werle, M. and Bernkop-Schnürch, A. (2006), Amino Acids, 30, 351-67 2) Pollaro, L. and Heinis, C. (2010), Med. Chem. Commun., 1, 319–324

56 FP19 FP20 DEVELOPMENT OF AN ALBUMIN-BINDING LIGAND FOR NEAR-INFRARED (NIR) PHOTOTHERMAL THERAPY USING PROLONGING THE PLASMA HALF-LIFE OF PEPTIDE BIOCOMPATIBLE PALLADIUM NANOPARTICLES THERAPEUTICS Belén Rubio-Ruiz, Ana M. Pérez-López, Thomas Bray, Martin Lee, Jason T. Weiss, Alan Serrels, Asier Alessandro Zorzi, Christian Heinis Unciti-Broceta

Laboratory of Therapeutic Proteins and Peptides (LPPT) Edinburgh Cancer Research UK Centre, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Institute of Chemical Sciences and Engineering, EPFL, Avenue Forel 2, CH-1015 Lausanne, Switzerland Crewe Road South, Edinburgh EH4 2XR, UK

Peptide therapeutics applied intravenously are rapidly cleared from the blood circulation by renal filtration. The Photothermal therapy (PTT), also known as photothermal ablation or optical hyperthermia, has gained wide short half-life prevents their application to diseases that require drug exposure of several hours or days (1). An attention as a precise and minimally invasive alternative for cancer treatment. This therapeutic approach consists attractive strategy to hamper filtration of peptides in the kidneys is to tether them non-covalently to a long-lived of using near-infrared (NIR) light-absorbing agents to convert photons into heat for ablation of cancer cells. serum protein such as human albumin (2). Several albumin-binding ligands based on peptides or small molecules Although the unique light-to-heat conversion property of many types of metallic nanostructures has been 1 were developed but they suffer from relatively low affinities for human albumin as well as a poor solubility in explored for photothermal therapy application , their unclear biotoxicity limits their therapeutic usefulness. physiological buffers, reducing their potential application to peptide therapeutics. Palladium is a transition metal with remarkable catalytic, mechanic and electronic properties. Despite these 2 To overcome these limitations, a chimeric peptide-small molecule albumin ligand with low nanomolar affinity characteristics, the use of palladium nanostructures in the nanomedicine field is still widely under-explored . 3 0 for human, rat and rabbit albumin, a high solubility and a small size suitable for automated synthesis of complex Using a green method we have produced Pd nanoparticles (PdNP) with high NIR light absorbance and conjugates was successfully developed. Peptides conjugated to the tag retained their bioactivity and displayed biocompatibility in cancer cell culture. By NIR irradiation, we have mediated the in vitro thermolysis of A549 around a 30-fold increase in half-life in rats. lung carcinoma cells demonstrating its potential for cancer photothermal therapy.

Schematic structure of the albumin-binding ligand (tag) (left panel) and pharmacokinetics of a therapeutic References peptide and its conjugated format in rat plasma upon i.v. injection (right panel). 1) Cheng, L. et al. Chem. Rev. 2014, 114, 10869-10939. 2) Dumas, A., Couvreur P. Chem. Sci. 2015, 6, 2153-2157. 3) Zhang, A. et al. J. Mater. Chem. A. 2014, 2, 1369- 1374. References 1) Werle, M. and Bernkop-Schnürch, A. (2006), Amino Acids, 30, 351-67 2) Pollaro, L. and Heinis, C. (2010), Med. Chem. Commun., 1, 319–324

57 Notes

______

58 Posters

59 P001 P002 IDENTIFICATION OF NOVEL DESIGN AND SYNTHESIS OF DUAL INHIBITORS OF ALDOSE 5-ARYLIDENE-3-PHENYL-2-THIOXOIMIDAZOLIDIN-4-ONES AS REDUCTASE AND PROTEIN TYROSINE PHOPSHATASE 1B INHIBITORS OF THE CHYMOTRYPSIN-LIKE ACTIVITIES OF THE Ilenia Adornato (1), Francesco Balestri (2), Rosaria Ottanà (1), Paolo Paoli (3), Antonella Del Corso (2), CONSTITUTIVE AND IMMUNO-PROTEASOMES Rosanna Maccari (1), Giulia Lori (3)

Ilenia Adornato (1), Roberta Ettari (1), Rosanna Maccari (1), Julia Kaffy (2), Rosaria Ottanà (1), Silvana 1) Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale SS. Grasso (1) Annunziata, 98168 Messina, Italy 2) Department of Biology, Biochemistry Unit, University of Pisa, Via S. Zeno, 51, 56123 Pisa, Italy 1) Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale 3) Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy Annunziata, 98168 Messina, Italy 2) Molécules Fluorées et Chimie Médicinale, BioCIS, Univ. Paris-Sud, CNRS, Université Paris Saclay, 5 rue Jean-Baptiste Clément, Chaîtenay-Malabry Cedex, France

Diabetes mellitus (DM) is a widespread chronic disease, whose worldwide prevalence is predicted to rise to about 10% of total world population by 2030. It is characterized by hyperglycemia and various metabolic The ubiquitin-proteasome system (UPS) is a key pathway involved in the intracellular protein turnover in imbalances, consequent to insulin deficiency and/or insulin-resistance. DM is also associated to serious eukaryotic cells. In normal cells, the proteasome proteolytic activity is responsible for a regular cell cycle long-term complications, such as neuropathy, nephropathy, retinopathy, atherosclerosis and increased progression, immune surveillance, and homeostasis control. Defects in UPS can lead to uncontrolled cell cardiovascular risk, which originate from hyperglycemia-induced oxidative and inflammatory events and from proliferation and tumor development [1]. The 20S constitutive proteasome (c20S) is composed of four stacked the consequent tissue and vascular damage. rings: two inner β-rings, containing the catalytic subunit, and two outer α-rings, whose function is to maintain a gate through which proteins enter the barrel-like structure. The standard catalytic subunits include β1, β2 and β5, Aldose reductase (AR) is an aldo-keto reductase which is implicated in the onset and progression of diabetic normally expressed in all cells. β5 subunit displays a chymotrypsin-like activity (ChT-L), which is mainly complications, by catalyzing the reduction of various carbonyl substrates, including glucose and lipid-derived involved in protein degradation and currently considered the primary target for the development of novel aldehydes, to cytotoxic and pro-inflammatory metabolites. Recent studies have demonstrated that AR is a key anticancer agents. The other two proteolytic subunits β1 and β2 are responsible for the caspase-like (C-L) and component of oxidative stress-induced inflammatory signaling, under both hyperglycemic and normoglycemic for the trypsin-like (T-L) activities, respectively. In addition to the constitutive proteasome, vertebrates possess a conditions, and is involved in the etiology of several inflammatory diseases.1 specialized form of proteasome, named immunoproteasome (i20S), which is predominantly expressed in Moreover, in DM, particularly in type 2 diabetes (T2DM), protein tyrosine phosphatase 1B (PTP1B) plays a monocytes and lymphocytes, and is responsible for cell-mediated immunity and for the generation of major critical role in the development of insulin-resistance. It has been established that the increased activity and/or histocompatibility complex (MHC) class I ligands. While β2i and β5i subunits perform the same type of overexpression of this enzyme is implicated in alterations of post-receptor events of insulin signaling, which lead activities of the β2c and β5c subunits, on the contrary, the replacement of β1c with β1i reduces the caspase-like to attenuated cellular response to the hormone. PTP1B overexpression is also linked to the sub-clinical activity to background levels, with β1i mainly performing a ChT-L activity [2]. inflammatory condition which underlies the development of DM.2 It is worth noting that i20S is a major form of the proteasome expressed in cells of hematopoietic origin, On this basis, it has been hypothesized that dual inhibitors of both PTP1B and AR may act as multitarget agents including multiple myeloma (MM) cells. Although specific inhibition of either β5i or β5c alone is insufficient to able to counteract simultaneously different mechanisms responsible for the development of both T2DM and produce an antitumor response, inhibition of all proteasome subunits is strongly cytotoxic to both hematologic associated pathologies. tumor cells and peripheral blood mononuclear cells. On the contrary, selective inhibition of both β5i and β5c is sufficient to induce an antitumor effect in MM, non-Hodgkin lymphoma, and leukemia cells while minimizing Therefore, as a part of our search for novel antidiabetic agents, differently substituted 4-thiazolidinone the toxicity toward nontransformed cells [3]. derivatives endowed with potent AR inhibitory activity3were selected to be evaluated as PTP1B inhibitors. On the basis of preliminary results, new (5-arylidene-2-oxo/thioxo-4-oxothiazolidin-3-yl)acetic acids were designed Starting from these considerations and assuming as lead compounds the oxathiazolones HT1171 and HT2004 in order to identify dual PTP1B/AR inhibitors. (Fig.1), reported to irreversibly inhibit β5i and β5c core particles [4], with a slight preference for β5i subunit, we tested on the constitutive and immuno- core particles a panel of 5-arylidene-3-phenyl-2-thioxoimidazolidin-4-ones 1 (Fig.1) to evaluate their selectivity profile. Several inhibitors were proven to be effective to target the ChT-L activities of both i20S and c20S, with a different trend of selectivity, based on their chemical features. The results of this investigation will be reported and discussed.

Pharmacophoric elements shared by PTP1B and AR inhibitors were combined in the new molecules, which possess a polar moiety and an extended hydrophobic portion potentially able to interact with critical aminoacid residues in the catalytic site and surrounding regions of both enzymes. The results of the in vitro evaluation of the newly-synthesized compounds will be reported and discussed.

Figure 1. Structure of the lead compounds and of novel 5-arylidene-3-phenyl-2-thioxoimidazolidin-4-ones 1 References 1) R. Maccari, R. Ottanà, J. Med. Chem. 58 (2015) 2047-2067. 2) S. Koren, I.G. Fantus, Best Pract. Res. Clin. Endocrinol. Metab. 21 (2007) 621-640. References 3) (a) R. Maccari, A. Del Corso, M. Giglio, R. Moschini, U. Mura, R. Ottanà, Bioorg. Med. Chem. Lett. 21 (2011) 200-203. 1) N. Micale, K. Scarbaci, V. Troiano, R. Ettari, S. Grasso, M. Zappalà, Peptide-based Proteasome Inhibitors in Anticancer (b) R. Maccari, R.M. Vitale, R. Ottanà, M. Rocchiccioli, A. Marrazzo, V. Cardile, A.C.E. Graziano, P. Amodeo, U. Mura, A. Drug Design, Med. Res. Rev. 2014, 34, 1001-1069. Del Corso, Eur. J. Med. Chem. 81 (2014) 1-14. 2) R. Ettari, S. Previti, A. Bitto, S. Grasso, M. Zappalà, Immunoproteasome-selective inhibitors: a promising strategy to treat hematologic malignancies, autoimmune and inflammatory diseases, Curr. Med. Chem. 2016, 23, 1217-1238. 3) F. Parlati, S. J. Lee, M. Aujay, E. Suzuki, K. Levitsky, J. B. Lorens, D. R. Micklem, P. Ruurs, C. Sylvain, Y. Lu, K. D. Shenk, M. K. Bennett, Carfilzomib can induce tumor cell death through selective inhibition of the chymotrypsin-like activity of the proteasome, Blood. 2009, 114, 3439-3447. 4) H. Fan, N. G. Angelo, J. D. Warren, C. F. Nathan G. Lin, Oxathiazolones selectively inhibit the human immunoproteasome over the constitutive proteasome. Med. Chem. Lett., 2014, 5, 405-410. 60 P001 P002 IDENTIFICATION OF NOVEL DESIGN AND SYNTHESIS OF DUAL INHIBITORS OF ALDOSE 5-ARYLIDENE-3-PHENYL-2-THIOXOIMIDAZOLIDIN-4-ONES AS REDUCTASE AND PROTEIN TYROSINE PHOPSHATASE 1B INHIBITORS OF THE CHYMOTRYPSIN-LIKE ACTIVITIES OF THE Ilenia Adornato (1), Francesco Balestri (2), Rosaria Ottanà (1), Paolo Paoli (3), Antonella Del Corso (2), CONSTITUTIVE AND IMMUNO-PROTEASOMES Rosanna Maccari (1), Giulia Lori (3)

Ilenia Adornato (1), Roberta Ettari (1), Rosanna Maccari (1), Julia Kaffy (2), Rosaria Ottanà (1), Silvana 1) Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale SS. Grasso (1) Annunziata, 98168 Messina, Italy 2) Department of Biology, Biochemistry Unit, University of Pisa, Via S. Zeno, 51, 56123 Pisa, Italy 1) Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale 3) Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy Annunziata, 98168 Messina, Italy 2) Molécules Fluorées et Chimie Médicinale, BioCIS, Univ. Paris-Sud, CNRS, Université Paris Saclay, 5 rue Jean-Baptiste Clément, Chaîtenay-Malabry Cedex, France

Diabetes mellitus (DM) is a widespread chronic disease, whose worldwide prevalence is predicted to rise to about 10% of total world population by 2030. It is characterized by hyperglycemia and various metabolic The ubiquitin-proteasome system (UPS) is a key pathway involved in the intracellular protein turnover in imbalances, consequent to insulin deficiency and/or insulin-resistance. DM is also associated to serious eukaryotic cells. In normal cells, the proteasome proteolytic activity is responsible for a regular cell cycle long-term complications, such as neuropathy, nephropathy, retinopathy, atherosclerosis and increased progression, immune surveillance, and homeostasis control. Defects in UPS can lead to uncontrolled cell cardiovascular risk, which originate from hyperglycemia-induced oxidative and inflammatory events and from proliferation and tumor development [1]. The 20S constitutive proteasome (c20S) is composed of four stacked the consequent tissue and vascular damage. rings: two inner β-rings, containing the catalytic subunit, and two outer α-rings, whose function is to maintain a gate through which proteins enter the barrel-like structure. The standard catalytic subunits include β1, β2 and β5, Aldose reductase (AR) is an aldo-keto reductase which is implicated in the onset and progression of diabetic normally expressed in all cells. β5 subunit displays a chymotrypsin-like activity (ChT-L), which is mainly complications, by catalyzing the reduction of various carbonyl substrates, including glucose and lipid-derived involved in protein degradation and currently considered the primary target for the development of novel aldehydes, to cytotoxic and pro-inflammatory metabolites. Recent studies have demonstrated that AR is a key anticancer agents. The other two proteolytic subunits β1 and β2 are responsible for the caspase-like (C-L) and component of oxidative stress-induced inflammatory signaling, under both hyperglycemic and normoglycemic for the trypsin-like (T-L) activities, respectively. In addition to the constitutive proteasome, vertebrates possess a conditions, and is involved in the etiology of several inflammatory diseases.1 specialized form of proteasome, named immunoproteasome (i20S), which is predominantly expressed in Moreover, in DM, particularly in type 2 diabetes (T2DM), protein tyrosine phosphatase 1B (PTP1B) plays a monocytes and lymphocytes, and is responsible for cell-mediated immunity and for the generation of major critical role in the development of insulin-resistance. It has been established that the increased activity and/or histocompatibility complex (MHC) class I ligands. While β2i and β5i subunits perform the same type of overexpression of this enzyme is implicated in alterations of post-receptor events of insulin signaling, which lead activities of the β2c and β5c subunits, on the contrary, the replacement of β1c with β1i reduces the caspase-like to attenuated cellular response to the hormone. PTP1B overexpression is also linked to the sub-clinical activity to background levels, with β1i mainly performing a ChT-L activity [2]. inflammatory condition which underlies the development of DM.2 It is worth noting that i20S is a major form of the proteasome expressed in cells of hematopoietic origin, On this basis, it has been hypothesized that dual inhibitors of both PTP1B and AR may act as multitarget agents including multiple myeloma (MM) cells. Although specific inhibition of either β5i or β5c alone is insufficient to able to counteract simultaneously different mechanisms responsible for the development of both T2DM and produce an antitumor response, inhibition of all proteasome subunits is strongly cytotoxic to both hematologic associated pathologies. tumor cells and peripheral blood mononuclear cells. On the contrary, selective inhibition of both β5i and β5c is sufficient to induce an antitumor effect in MM, non-Hodgkin lymphoma, and leukemia cells while minimizing Therefore, as a part of our search for novel antidiabetic agents, differently substituted 4-thiazolidinone the toxicity toward nontransformed cells [3]. derivatives endowed with potent AR inhibitory activity3were selected to be evaluated as PTP1B inhibitors. On the basis of preliminary results, new (5-arylidene-2-oxo/thioxo-4-oxothiazolidin-3-yl)acetic acids were designed Starting from these considerations and assuming as lead compounds the oxathiazolones HT1171 and HT2004 in order to identify dual PTP1B/AR inhibitors. (Fig.1), reported to irreversibly inhibit β5i and β5c core particles [4], with a slight preference for β5i subunit, we tested on the constitutive and immuno- core particles a panel of 5-arylidene-3-phenyl-2-thioxoimidazolidin-4-ones 1 (Fig.1) to evaluate their selectivity profile. Several inhibitors were proven to be effective to target the ChT-L activities of both i20S and c20S, with a different trend of selectivity, based on their chemical features. The results of this investigation will be reported and discussed.

Pharmacophoric elements shared by PTP1B and AR inhibitors were combined in the new molecules, which possess a polar moiety and an extended hydrophobic portion potentially able to interact with critical aminoacid residues in the catalytic site and surrounding regions of both enzymes. The results of the in vitro evaluation of the newly-synthesized compounds will be reported and discussed.

Figure 1. Structure of the lead compounds and of novel 5-arylidene-3-phenyl-2-thioxoimidazolidin-4-ones 1 References 1) R. Maccari, R. Ottanà, J. Med. Chem. 58 (2015) 2047-2067. 2) S. Koren, I.G. Fantus, Best Pract. Res. Clin. Endocrinol. Metab. 21 (2007) 621-640. References 3) (a) R. Maccari, A. Del Corso, M. Giglio, R. Moschini, U. Mura, R. Ottanà, Bioorg. Med. Chem. Lett. 21 (2011) 200-203. 1) N. Micale, K. Scarbaci, V. Troiano, R. Ettari, S. Grasso, M. Zappalà, Peptide-based Proteasome Inhibitors in Anticancer (b) R. Maccari, R.M. Vitale, R. Ottanà, M. Rocchiccioli, A. Marrazzo, V. Cardile, A.C.E. Graziano, P. Amodeo, U. Mura, A. Drug Design, Med. Res. Rev. 2014, 34, 1001-1069. Del Corso, Eur. J. Med. Chem. 81 (2014) 1-14. 2) R. Ettari, S. Previti, A. Bitto, S. Grasso, M. Zappalà, Immunoproteasome-selective inhibitors: a promising strategy to treat hematologic malignancies, autoimmune and inflammatory diseases, Curr. Med. Chem. 2016, 23, 1217-1238. 3) F. Parlati, S. J. Lee, M. Aujay, E. Suzuki, K. Levitsky, J. B. Lorens, D. R. Micklem, P. Ruurs, C. Sylvain, Y. Lu, K. D. Shenk, M. K. Bennett, Carfilzomib can induce tumor cell death through selective inhibition of the chymotrypsin-like activity of the proteasome, Blood. 2009, 114, 3439-3447. 4) H. Fan, N. G. Angelo, J. D. Warren, C. F. Nathan G. Lin, Oxathiazolones selectively inhibit the human immunoproteasome over the constitutive proteasome. Med. Chem. Lett., 2014, 5, 405-410. 61 P003 P004 DESIGN AND SYNTHESIS OF SOME CERTAIN COMMON REGULARITIES IN THE CHEMICAL 2-ARYL-5-FLUORO-6-(4-METHYL-1-PIPERAZINYL) STRUCTURE OF ANTIHISTAMINES, AND THEIR INFLUENCE ON BENZOTHIAZOLES OF POTENTIAL ANTITUMOR ACTIVITIES PROPERTIES

Thuraya Al-Harthy, Raid Abdel-Jalil, Wajdi Zoghaib Eldar Movsumzade (1), Galina Kolchina (2), Oksana Kirillova (1), Anastasya Bakhtina (1), Il'nara Safiullina (1) Chemistry Department, College of Science, Sultan Qaboos University, Muscat, Oman; [email protected] 1) Ufa State Petroleum Technological University 2) Sterlitamak branch of Bashkir State University

Heterocyclic compounds are vastly distributed in nature and commonly found in most of available drugs in the market. Benzothiazole, for instance, is one of the most important dug-based constitute in medicinal chemistry. In particular, 2-arylbenzothiazle has been investigated extensively in the last decade due to its biological activities, Allergic diseases are a serious social, economic and medical problem. For effective treatment of these diseases specifically anticancer activity1. using H1-receptor blockers. ® (1) is a potent antibiotic drug commercially available with a wide spectrum of biological activity Basic structural requirements for H1-receptor antagonists have been identified, as shown below: and studies revealed that the potent activity is attributed to the presence of both fluorine and piperazine appendages2. Previously, we have successfully synthesized several heterocycles incorporating fluorine and piperazine which also demonstrated promising biological activities3.

Diaryl structure is present both in the generation of antihistamines and is essential for high affinity for the H1 receptor. Several SAR studies indicate that two aryl moiety capable of taking the conformation non-coplanar with respect to each other for optimal interaction with H1-receptors. The amino group is basic, pKa ranges from 8.5 to 10, and thus, it is expected that it will be protonated upon binding to the receptor. X - linking atom O, C or N. This group serves as a dividing band for key pharmacophore fragments. Many In pursuit to develop novel potential bioactive heterocyclic systems, herein we report a synthetic methodology antihistamines contain a chiral carbon atom in the connecting passage, and therefore there is a stereoselective for the synthesis of novel series of 2-aryl-5-fluoro-6-(4-methyl-piperazin-1-yl)-benzothiazoles (2) expecting to binding to receptors. deliver promising anticancer activity. In many formulations branching (CH2)n- band reduces antihistaminic activity. The compounds which are unsymmetrically substituted unsaturated carbon chain, typically one geometric isomer shows a higher affinity for References the receptor than the other. 1) (a)I. Hutchinson,M.-S. Chua,H. L. Browne,V. Trapani,T. D. Bradshaw,A. D. Westwell,M. F. G. Stevens, J. Med. Chem.,2001, 44, 1446; (b)K. Sztanke,A. Maziarka,A. Osinka,M. Sztanke, Bioorg. Med. Chem.,2013, 21 3648; (c)V. S. Typically, antihistamines are significantly more lipophilic than the endogenous agonist histamine, and Patil,K. P. Nandre,S. Ghosh,V. J. Rao,B. A. Chopade,B. Sridhar,S. V. Bhosale,S. V. Bhosale, Eur. J. Med. Chem.,2013, 59, antihistamines are more competitive in relation to H1 receptors. 304; (d)P. K. Deb,R. Kaur,B. Chandrasekaran,M. Bala,D. Gill,V. R. Kaki,R. R. Akkinepalli,R. Mailavaram, Med Chem Res,2014, 23, 2780; (e)M. K. Singh,R. Tilak,G. Nath,S. K. Awasthi,A. Agarwal, Eur. J. Med. Chem., 2013, 63, 635. Variations represented groups, are the basis for the differences in pharmacological action and side effects. These 2) J. P. Sanchez,J. M. Domagala,S. E. Hagen,C. L. Heifetz,M. P. Hutt,J. B. Nichols,A. K. Trehan, J. Med. Chem.,1988, 31, preparations contain pharmacophores which are necessary for binding to muscarinic, adrenergic, serotonergic, 983. 3) (a)R. J. Abdel-jalil,R. A. Al-Qawasemeh,W. Voleter,P. Heeg,M. M. El-Abadelah,S. S. Sabri, J. Heterocycl. Chem. ,2009, and dopamine receptors. 37, 1273; (b)K. H. Abu-Elteen,R. J. Abdel-Jalil,M. A. Hamad,M. Ghaleb,K. M. Khan,W. Voelter, J. Med. Sci.,2008, 8, 673; Sedative effect of first generation drugs arises due to their penetration across blood-brain barrier and blockade (c)R. J. Abdel-Jalil,H. M. Aldoqum,M. T. Ayoub,W. Voelter, Heterocycles,2005, 65, 2061 the central H1-receptors. They quickly and reversibly bind to the receptors. Representatives of the second generation have antihistaminic effect lasting due to the slow dissociation of the receptor - ligand complex. Typically, alkyl groups or polar groups bonded to the piperidine/piperazine these compounds reduce their affinity for muscarinic and adrenergic receptors.All these compounds are devoid of sedative effects.

62 P003 P004 DESIGN AND SYNTHESIS OF SOME CERTAIN COMMON REGULARITIES IN THE CHEMICAL 2-ARYL-5-FLUORO-6-(4-METHYL-1-PIPERAZINYL) STRUCTURE OF ANTIHISTAMINES, AND THEIR INFLUENCE ON BENZOTHIAZOLES OF POTENTIAL ANTITUMOR ACTIVITIES PROPERTIES

Thuraya Al-Harthy, Raid Abdel-Jalil, Wajdi Zoghaib Eldar Movsumzade (1), Galina Kolchina (2), Oksana Kirillova (1), Anastasya Bakhtina (1), Il'nara Safiullina (1) Chemistry Department, College of Science, Sultan Qaboos University, Muscat, Oman; [email protected] 1) Ufa State Petroleum Technological University 2) Sterlitamak branch of Bashkir State University

Heterocyclic compounds are vastly distributed in nature and commonly found in most of available drugs in the market. Benzothiazole, for instance, is one of the most important dug-based constitute in medicinal chemistry. In particular, 2-arylbenzothiazle has been investigated extensively in the last decade due to its biological activities, Allergic diseases are a serious social, economic and medical problem. For effective treatment of these diseases specifically anticancer activity1. using H1-receptor blockers. Ciprofloxacin® (1) is a potent antibiotic drug commercially available with a wide spectrum of biological activity Basic structural requirements for H1-receptor antagonists have been identified, as shown below: and studies revealed that the potent activity is attributed to the presence of both fluorine and piperazine appendages2. Previously, we have successfully synthesized several heterocycles incorporating fluorine and piperazine which also demonstrated promising biological activities3.

Diaryl structure is present both in the generation of antihistamines and is essential for high affinity for the H1 receptor. Several SAR studies indicate that two aryl moiety capable of taking the conformation non-coplanar with respect to each other for optimal interaction with H1-receptors. The amino group is basic, pKa ranges from 8.5 to 10, and thus, it is expected that it will be protonated upon binding to the receptor. X - linking atom O, C or N. This group serves as a dividing band for key pharmacophore fragments. Many In pursuit to develop novel potential bioactive heterocyclic systems, herein we report a synthetic methodology antihistamines contain a chiral carbon atom in the connecting passage, and therefore there is a stereoselective for the synthesis of novel series of 2-aryl-5-fluoro-6-(4-methyl-piperazin-1-yl)-benzothiazoles (2) expecting to binding to receptors. deliver promising anticancer activity. In many formulations branching (CH2)n- band reduces antihistaminic activity. The compounds which are unsymmetrically substituted unsaturated carbon chain, typically one geometric isomer shows a higher affinity for References the receptor than the other. 1) (a)I. Hutchinson,M.-S. Chua,H. L. Browne,V. Trapani,T. D. Bradshaw,A. D. Westwell,M. F. G. Stevens, J. Med. Chem.,2001, 44, 1446; (b)K. Sztanke,A. Maziarka,A. Osinka,M. Sztanke, Bioorg. Med. Chem.,2013, 21 3648; (c)V. S. Typically, antihistamines are significantly more lipophilic than the endogenous agonist histamine, and Patil,K. P. Nandre,S. Ghosh,V. J. Rao,B. A. Chopade,B. Sridhar,S. V. Bhosale,S. V. Bhosale, Eur. J. Med. Chem.,2013, 59, antihistamines are more competitive in relation to H1 receptors. 304; (d)P. K. Deb,R. Kaur,B. Chandrasekaran,M. Bala,D. Gill,V. R. Kaki,R. R. Akkinepalli,R. Mailavaram, Med Chem Res,2014, 23, 2780; (e)M. K. Singh,R. Tilak,G. Nath,S. K. Awasthi,A. Agarwal, Eur. J. Med. Chem., 2013, 63, 635. Variations represented groups, are the basis for the differences in pharmacological action and side effects. These 2) J. P. Sanchez,J. M. Domagala,S. E. Hagen,C. L. Heifetz,M. P. Hutt,J. B. Nichols,A. K. Trehan, J. Med. Chem.,1988, 31, preparations contain pharmacophores which are necessary for binding to muscarinic, adrenergic, serotonergic, 983. 3) (a)R. J. Abdel-jalil,R. A. Al-Qawasemeh,W. Voleter,P. Heeg,M. M. El-Abadelah,S. S. Sabri, J. Heterocycl. Chem. ,2009, and dopamine receptors. 37, 1273; (b)K. H. Abu-Elteen,R. J. Abdel-Jalil,M. A. Hamad,M. Ghaleb,K. M. Khan,W. Voelter, J. Med. Sci.,2008, 8, 673; Sedative effect of first generation drugs arises due to their penetration across blood-brain barrier and blockade (c)R. J. Abdel-Jalil,H. M. Aldoqum,M. T. Ayoub,W. Voelter, Heterocycles,2005, 65, 2061 the central H1-receptors. They quickly and reversibly bind to the receptors. Representatives of the second generation have antihistaminic effect lasting due to the slow dissociation of the receptor - ligand complex. Typically, alkyl groups or polar groups bonded to the piperidine/piperazine these compounds reduce their affinity for muscarinic and adrenergic receptors.All these compounds are devoid of sedative effects.

63 P005 P006 MODIFICATION OF STRUCTURE OF A DEVELOPMENT OF STEREOSELECTIVE SYNTHSES AND HYDROXYTRIFLUOROMETHYLINDOLE, FOR THE PURPOSE OF CHEMICAL BIOLOGY STUDIES FOR BROMODOMAIN INHIBITORS INCREASE IN ITS WATER SOLUBILITY AS ANTI CANCER DRUG

Eldar Movsumzade (1), Galina Kolchina (2), David Aleksanyan (3), Karina Aleksanyan (3) Mohammad Amin, Philip Thompson

1) Ufa State Petroleum Technological University Monash Institute Of Pharmaceutical Sciences, Monash University, Melbourne, VIC 3052 2) Sterlitamak branch of Bashkir State University 3) Gubkin Russian State University of Oil and Gas

“Epigenetics” refers to heritable changes in gene transcription that occur without changing the underlying DNA sequence. Post translational modifications (PTMs) of histones such as acetylation of histone lysine are central to Studies show that compounds 1a,b possess high fungicidal activity. To increase biological activity and solubility epigenetic modulations of gene transcriptions. Bromodomain containing proteins (BCPs), co-ordinate of acquired compounds, methyldimethylaminogroup was added. transcription factor assembly by recognizing the lysine acetylation state and act as “readers” of histone. On the stage of development of anti-viral arbidol, which is based on indole, there was the same Bromodomains are ~ 110 amino acid 4-helix modules that exist as part of larger protein architectures. Inhibition problem that was solved by introduction of methyldimethylamin residue in the final stage of the synthesis. of BCPs has potential therapeutic applications in a range of diseases including cancer. Inhibition of BET bromodomains also allows potent anti-proliferative activity in a number of hematological cancer models partly Since the molecule of indole 1b has multiple active sites for introduction of substituents, the reaction is firstly through suppression of Myc oncogene and downstream Myc-driven pathways. High throughput screening studied on the example of 4-hydroxy-3-methyl-6-trifluoromethyl-2-phenylindole 1a. identified (S)-N-acetyl tetrahydroquinaldine ((S)-N-Ac-THQ) as a fragment-style inhibitor of the BET bromodomain and led to one clinical candidate I-BET726 developed at GSK for cancer treatment. (S)-N -Ac-THQ, the core moiety of the I-BET726, inhibits bromodomain by mimicking acetyl lysine. Importance of the enantiomeric configuration of this moiety to activity has encouraged us to examine enantiocontrolled syntheses of (S)-N-Ac-THQ analogues and to explore the structure activity relationships of these compounds. Our initial approach involved kinetic resolution (KR) using phthaloyl-leucine derivatisation to give optically pure (S)-N-Ac-THQ in >98% ee. In a second approach we used fluorenylethyl chloroformate (FLEC) as a chiral resolving agent for diastereomeric resolution. This involved the coupling of (S)-FLEC to a racemic mixture of While carrying out the reaction in a condition of introducing methyldimethylaminogroup during the synthesis of THQ. The resulting diastereomers were resolved by silica gel chromatography, deprotected and acetylated to Arbidol, decomposition of indole molecules took place 1a. As a result, selection of optimal conditions for the provide pure samples of both enantiomers in >98% ee. An improved cheap, high yielding and reproducible reaction of dimetilaminometilation. The structure of the compound was proved by means of a two-dimensional enantioselective synthesis of FLEC was also developed and carried out in multi grams. 2 spectroscopy, NMR NOESI, which showed the product - indole . A final approach, allowing access to 6-substituted derivatives, involved chiral reduction of 6-bromoquinoline to While carrying out the reaction of dimetilaminometilation indole 1b, under the conditions found during 6-bromoquinaldine. This bromodomain inhibitor core was then derivatised to create a biotinylated probe. dimethylaminomethylation of indole 1a, 1b the degradation of the molecule takes place. This reaction is Application of this probe in a pull down assay allowed the isolation of recombinant bromodomain targets and conducted in water. will, in our future work, allow the analysis of cellular extracts. The synthetic methods developed in this investigation provide a platform for the provision of further derivatives and the investigation of them as potential inhibitors of BCPs.

Thus, the optimum conditions were developed for the reaction if dimethylaminomethylation of indoles and increasing their water solubility.

64 P005 P006 MODIFICATION OF STRUCTURE OF A DEVELOPMENT OF STEREOSELECTIVE SYNTHSES AND HYDROXYTRIFLUOROMETHYLINDOLE, FOR THE PURPOSE OF CHEMICAL BIOLOGY STUDIES FOR BROMODOMAIN INHIBITORS INCREASE IN ITS WATER SOLUBILITY AS ANTI CANCER DRUG

Eldar Movsumzade (1), Galina Kolchina (2), David Aleksanyan (3), Karina Aleksanyan (3) Mohammad Amin, Philip Thompson

1) Ufa State Petroleum Technological University Monash Institute Of Pharmaceutical Sciences, Monash University, Melbourne, VIC 3052 2) Sterlitamak branch of Bashkir State University 3) Gubkin Russian State University of Oil and Gas

“Epigenetics” refers to heritable changes in gene transcription that occur without changing the underlying DNA sequence. Post translational modifications (PTMs) of histones such as acetylation of histone lysine are central to Studies show that compounds 1a,b possess high fungicidal activity. To increase biological activity and solubility epigenetic modulations of gene transcriptions. Bromodomain containing proteins (BCPs), co-ordinate of acquired compounds, methyldimethylaminogroup was added. transcription factor assembly by recognizing the lysine acetylation state and act as “readers” of histone. On the stage of development of anti-viral medication arbidol, which is based on indole, there was the same Bromodomains are ~ 110 amino acid 4-helix modules that exist as part of larger protein architectures. Inhibition problem that was solved by introduction of methyldimethylamin residue in the final stage of the synthesis. of BCPs has potential therapeutic applications in a range of diseases including cancer. Inhibition of BET bromodomains also allows potent anti-proliferative activity in a number of hematological cancer models partly Since the molecule of indole 1b has multiple active sites for introduction of substituents, the reaction is firstly through suppression of Myc oncogene and downstream Myc-driven pathways. High throughput screening studied on the example of 4-hydroxy-3-methyl-6-trifluoromethyl-2-phenylindole 1a. identified (S)-N-acetyl tetrahydroquinaldine ((S)-N-Ac-THQ) as a fragment-style inhibitor of the BET bromodomain and led to one clinical candidate I-BET726 developed at GSK for cancer treatment. (S)-N -Ac-THQ, the core moiety of the I-BET726, inhibits bromodomain by mimicking acetyl lysine. Importance of the enantiomeric configuration of this moiety to activity has encouraged us to examine enantiocontrolled syntheses of (S)-N-Ac-THQ analogues and to explore the structure activity relationships of these compounds. Our initial approach involved kinetic resolution (KR) using phthaloyl-leucine derivatisation to give optically pure (S)-N-Ac-THQ in >98% ee. In a second approach we used fluorenylethyl chloroformate (FLEC) as a chiral resolving agent for diastereomeric resolution. This involved the coupling of (S)-FLEC to a racemic mixture of While carrying out the reaction in a condition of introducing methyldimethylaminogroup during the synthesis of THQ. The resulting diastereomers were resolved by silica gel chromatography, deprotected and acetylated to Arbidol, decomposition of indole molecules took place 1a. As a result, selection of optimal conditions for the provide pure samples of both enantiomers in >98% ee. An improved cheap, high yielding and reproducible reaction of dimetilaminometilation. The structure of the compound was proved by means of a two-dimensional enantioselective synthesis of FLEC was also developed and carried out in multi grams. 2 spectroscopy, NMR NOESI, which showed the product - indole . A final approach, allowing access to 6-substituted derivatives, involved chiral reduction of 6-bromoquinoline to While carrying out the reaction of dimetilaminometilation indole 1b, under the conditions found during 6-bromoquinaldine. This bromodomain inhibitor core was then derivatised to create a biotinylated probe. dimethylaminomethylation of indole 1a, 1b the degradation of the molecule takes place. This reaction is Application of this probe in a pull down assay allowed the isolation of recombinant bromodomain targets and conducted in water. will, in our future work, allow the analysis of cellular extracts. The synthetic methods developed in this investigation provide a platform for the provision of further derivatives and the investigation of them as potential inhibitors of BCPs.

Thus, the optimum conditions were developed for the reaction if dimethylaminomethylation of indoles and increasing their water solubility.

65 P007 P008 DESIGN OF PORPHYRIN-LOADED NANOBUBBLES FOR DESIGN, SYNTHESIS AND STRUCTURE-ACTIVITY THERANOSTIC APPLICATIONS RELATIONSHIPS OF 1-(ARYLSULFONYL)-1H-INDOLE DERIVATIVES AS NON-BASIC 5-HT6 RECEPTOR LIGANDS Federica Bosca (1), Peter Bielecki (2), Christopher Hermandez (2), Alessandro Barge (1), Agata Exner (2) Ryszard Bugno, Jakub Staroń, Adam Hogendorf, Grzegorz Satała, Dawid Warszycki, Stefan Mordalski, 1) Dipartimento di Scienza e Tecnologia del Farmaco, Università di Torino, Torino, Italy Andrzej J. Bojarski 2) Departments of Biomedical Engineering and Radiology, Case Western Reserve University, Cleveland, OH, USA Department of Medicinal Chemistry, Institute of Pharmacology Polish Academy of Sciences, 12 Smętna Street, 31-343 Kraków, Poland

Nanoparticle formulations as theranostic agents for cancer therapy have gained a lot of attention for their ability to passively accumulate inside tumors through the enhance permeation and retention effect (1). A particular class of nanoparticles are nanobubbles (NBs), which are nanostructures constituted by perfluorocarbon gas stabilized Until recently, all generations of 5-HT6R ligands contained basic nitrogen atom, which was considered to be by a lipid, surfactant and/or polymer shell (2,3). Because of their gas-core structure, NBs operate as an crucial for binding to the receptor. However, in last years, inadvertently, several high-affinity ligands without a ultrasound (US) contrast agents without any further modification. Moreover, the robust and easily modifiable protonable nitrogen atom were identified.1-3 The phenomenon has been studied and some hypotheses were shell can be exploited to load drug, prodrug and/or vector in order to build the theranostic agent (4). formulated,1-3 but the mechanism of non-basic ligand-receptor interaction is still unclear. All the tested 5-HT6R Recently, sonodynamic therapy has become a promising noninvasive approach for cancer therapy (5). The ligands with reduced basicity developed so far revealed advantageous selectivity over other monoaminergic treatment exploits the ability of peculiar molecules (i.e. porphyrins) to be excited by US and to produce oxygen GPCRs and low hERG affinity. radical species during their decay process. Radical species, in turn, result in cell death. The advantage of using As a part of our study on the non-basic 5-HT6R ligands, a consistent series of indole derivatives has been US is to induce the cytotoxic effect only in a definite region at a precise time, thereby decreasing side effects. In designed in an attempt to describe their specific interactions in the binding pocket. Following the examples of order to capitalize on the real-time visualization and on-demand delivery of ultrasound contrast agents, this literature ligands with the 1-(phenylsulfonyl)-1H-indole fragment and the basic nitrogen atom, their counterparts project strives to combine porphyrins with the nanobubbles in order to obtain an US-activated theranostic agent. with reduced and/or removed basicity were synthesized. The ligands with the highest affinity have been further Here, porphyrins with different degrees of lipophilicity, molecular weight, shape and rigidity were synthesized, developed. and were encapsulated in the NBs. Resulting porphyrin-NBs were characterized by ultrasound, microscopy imaging, dynamic light scattering (DLS), and extent of porphyrin loading. The 5-HT6, 5-HT1A, 5-HT2A, 5-HT7 and D2 receptor affinities for all the synthesized compounds were assessed in radioligand displacement experiments. The structure-affinity relationships and the results of molecular Porphyrin-nanobubbles were found to be in the range of 200-300 nm in diameter, clearly visible under modelling experiments are discussed. ultrasound (Contrast harmonic Imaging, frequency 8.0 MHz, mechanical index of 0.08), and have a loading of 25% with porphyrin A (more amphiphilic and more flexible) and 0.5% with porphyrin B (more rigid and more Acknowledgements lipophylic). In vitro cell viability tests are currently ongoing. Overall, because of their nanoparticle footprint, The study was partly supported by the grant OPUS 2014/13/B/NZ7/02210 financed by the Polish National ultrasound visibility combined with the switchable cytotoxicity of porphyrin, Porphyrin-NBs could make an Science Centre exciting new class of theranostic agents References 1) Harris R.N., Stabler R.S., Repke D.B., Kress J.M., Walker K.A., Martin R.S., Brothers J.M., Ilnicka M., Lee S.W., References Mirzadegan T. Bioorg. Med. Chem. Lett. 2010, 20(11), 3436-40. 2) Ivachtchenko A.V., Golovina E.S., Kadieva M.G., Kysil V.M., Mitkin O.D., Tkachenko S.E., Okun .I.M. J. Med. Chem. 1) Maeda H., Wu J., Sawa T., Matsumura Y., Hori. K. Tumor vascular permeability and the EPR effect in macromolecular 2011, 54(23), 8161-73. therapeutics: a review. Journal of Controlled Release 2000; 65: 271–284. 3) Van Loevezijn A., Venhorst J., Iwema Bakker W.I., Lange J.H., de Looff W., Henzen R., de Vries J., van de Woestijne 2) Perera, R. H., Hernandez, C., Zhou, H., Kota, P., Burke, A. and Exner, A. A. Ultrasound imaging beyond the vasculature R.P., den Hartog A.P., Verhoog S., van der Neut M.A., de Bruin N.M., Kruse C.G. Bioorg. Med. Chem. Lett. 2016, 26(6), with new generation contrast agents. WIREs Nanomed Nanobiotechnol, 2015; 7: 593–608. 1605-11. 3) Wu H, Rognin NG, Krupka TM, Solorio L, Yoshiara H, Guenette G, Sanders C, Kamiyama N, Exner AA. Acoustic characterization and pharmacokinetic analyses of new nanobubble ultrasound contrast agents. Ultrasound Med Biol, 2013; 39(11):2137-46 4) Janib SM, Moses AS, MacKay JA, Imaging and drug delivery using theranostic nanoparticles. Adv Drug Deliv Rev. 2010; 62(11):1052-63 5) Xiong, W., Wang, P., Hu, J., Jia, Y., Wu, L., Chen, X., Liu, Q., Wang, X. A new sensitizer DVDMS combined with multiple focused ultrasound treatments: an effective antitumor strategy. Scientific report, 2015; DOI: 10.1038/srep17485

66 P007 P008 DESIGN OF PORPHYRIN-LOADED NANOBUBBLES FOR DESIGN, SYNTHESIS AND STRUCTURE-ACTIVITY THERANOSTIC APPLICATIONS RELATIONSHIPS OF 1-(ARYLSULFONYL)-1H-INDOLE DERIVATIVES AS NON-BASIC 5-HT6 RECEPTOR LIGANDS Federica Bosca (1), Peter Bielecki (2), Christopher Hermandez (2), Alessandro Barge (1), Agata Exner (2) Ryszard Bugno, Jakub Staroń, Adam Hogendorf, Grzegorz Satała, Dawid Warszycki, Stefan Mordalski, 1) Dipartimento di Scienza e Tecnologia del Farmaco, Università di Torino, Torino, Italy Andrzej J. Bojarski 2) Departments of Biomedical Engineering and Radiology, Case Western Reserve University, Cleveland, OH, USA Department of Medicinal Chemistry, Institute of Pharmacology Polish Academy of Sciences, 12 Smętna Street, 31-343 Kraków, Poland

Nanoparticle formulations as theranostic agents for cancer therapy have gained a lot of attention for their ability to passively accumulate inside tumors through the enhance permeation and retention effect (1). A particular class of nanoparticles are nanobubbles (NBs), which are nanostructures constituted by perfluorocarbon gas stabilized Until recently, all generations of 5-HT6R ligands contained basic nitrogen atom, which was considered to be by a lipid, surfactant and/or polymer shell (2,3). Because of their gas-core structure, NBs operate as an crucial for binding to the receptor. However, in last years, inadvertently, several high-affinity ligands without a ultrasound (US) contrast agents without any further modification. Moreover, the robust and easily modifiable protonable nitrogen atom were identified.1-3 The phenomenon has been studied and some hypotheses were shell can be exploited to load drug, prodrug and/or vector in order to build the theranostic agent (4). formulated,1-3 but the mechanism of non-basic ligand-receptor interaction is still unclear. All the tested 5-HT6R Recently, sonodynamic therapy has become a promising noninvasive approach for cancer therapy (5). The ligands with reduced basicity developed so far revealed advantageous selectivity over other monoaminergic treatment exploits the ability of peculiar molecules (i.e. porphyrins) to be excited by US and to produce oxygen GPCRs and low hERG affinity. radical species during their decay process. Radical species, in turn, result in cell death. The advantage of using As a part of our study on the non-basic 5-HT6R ligands, a consistent series of indole derivatives has been US is to induce the cytotoxic effect only in a definite region at a precise time, thereby decreasing side effects. In designed in an attempt to describe their specific interactions in the binding pocket. Following the examples of order to capitalize on the real-time visualization and on-demand delivery of ultrasound contrast agents, this literature ligands with the 1-(phenylsulfonyl)-1H-indole fragment and the basic nitrogen atom, their counterparts project strives to combine porphyrins with the nanobubbles in order to obtain an US-activated theranostic agent. with reduced and/or removed basicity were synthesized. The ligands with the highest affinity have been further Here, porphyrins with different degrees of lipophilicity, molecular weight, shape and rigidity were synthesized, developed. and were encapsulated in the NBs. Resulting porphyrin-NBs were characterized by ultrasound, microscopy imaging, dynamic light scattering (DLS), and extent of porphyrin loading. The 5-HT6, 5-HT1A, 5-HT2A, 5-HT7 and D2 receptor affinities for all the synthesized compounds were assessed in radioligand displacement experiments. The structure-affinity relationships and the results of molecular Porphyrin-nanobubbles were found to be in the range of 200-300 nm in diameter, clearly visible under modelling experiments are discussed. ultrasound (Contrast harmonic Imaging, frequency 8.0 MHz, mechanical index of 0.08), and have a loading of 25% with porphyrin A (more amphiphilic and more flexible) and 0.5% with porphyrin B (more rigid and more Acknowledgements lipophylic). In vitro cell viability tests are currently ongoing. Overall, because of their nanoparticle footprint, The study was partly supported by the grant OPUS 2014/13/B/NZ7/02210 financed by the Polish National ultrasound visibility combined with the switchable cytotoxicity of porphyrin, Porphyrin-NBs could make an Science Centre exciting new class of theranostic agents References 1) Harris R.N., Stabler R.S., Repke D.B., Kress J.M., Walker K.A., Martin R.S., Brothers J.M., Ilnicka M., Lee S.W., References Mirzadegan T. Bioorg. Med. Chem. Lett. 2010, 20(11), 3436-40. 2) Ivachtchenko A.V., Golovina E.S., Kadieva M.G., Kysil V.M., Mitkin O.D., Tkachenko S.E., Okun .I.M. J. Med. Chem. 1) Maeda H., Wu J., Sawa T., Matsumura Y., Hori. K. Tumor vascular permeability and the EPR effect in macromolecular 2011, 54(23), 8161-73. therapeutics: a review. Journal of Controlled Release 2000; 65: 271–284. 3) Van Loevezijn A., Venhorst J., Iwema Bakker W.I., Lange J.H., de Looff W., Henzen R., de Vries J., van de Woestijne 2) Perera, R. H., Hernandez, C., Zhou, H., Kota, P., Burke, A. and Exner, A. A. Ultrasound imaging beyond the vasculature R.P., den Hartog A.P., Verhoog S., van der Neut M.A., de Bruin N.M., Kruse C.G. Bioorg. Med. Chem. Lett. 2016, 26(6), with new generation contrast agents. WIREs Nanomed Nanobiotechnol, 2015; 7: 593–608. 1605-11. 3) Wu H, Rognin NG, Krupka TM, Solorio L, Yoshiara H, Guenette G, Sanders C, Kamiyama N, Exner AA. Acoustic characterization and pharmacokinetic analyses of new nanobubble ultrasound contrast agents. Ultrasound Med Biol, 2013; 39(11):2137-46 4) Janib SM, Moses AS, MacKay JA, Imaging and drug delivery using theranostic nanoparticles. Adv Drug Deliv Rev. 2010; 62(11):1052-63 5) Xiong, W., Wang, P., Hu, J., Jia, Y., Wu, L., Chen, X., Liu, Q., Wang, X. A new sensitizer DVDMS combined with multiple focused ultrasound treatments: an effective antitumor strategy. Scientific report, 2015; DOI: 10.1038/srep17485

67 P009 P010 IMPROVED ANALGESICS: BU08028 A NOVEL, BIFUNCTIONAL OXO-β-SULTAMS AS ACTIVITY-BASED PROBES NOP/MOP LIGAND Luís Carvalho, Lídia Gonçalves, Rui Moreira, Susana Lucas Gerta Cami-Kobeci (1), Mei-Chuan Ko (2), Lawrence Toll (3), Stephen M. Husbands (1) Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal 1) Department of Pharmacy and Pharmacology, University of Bath, Bath, BA2 7AY, UK 2) Wake Forest University, Winston-Salem, North Carolina, USA 3) Torrey Pines Institute for Molecular Studies, Port St. Lucie, Fl, USA Chronic Obstructive Pulmonary Disease (COPD) is a condition characterized by lung inflammation and progressive weakening of its structure leading to an irreversible narrowing of the airways. It is reported that it will become the third leading cause of death worldwide in 2030 by the World Health Organization1. New Although mu opioid (MOP) analgesics, such as , are the preferred analgesics, they possess well biomarkers to diagnose and predict the activity and progression of COPD are urgently needed. The central characterised, unwanted effects such as respiratory depression and abuse potential. hypothesis of this project is that COPD induced inflammation causes modulation of proteolytic enzymes in the When a NOP (nociceptin receptor) agonist is co-administered with a MOP agonist, the combination produces a lungs, like Human Neutrophil Elastase (HNE), which are clinically relevant for the diagnosis and prognosis of synergistic effect, resulting in enhanced analgesia.[1] This suggests the possibility of obtaining strong analgesia COPD. Activity-based probes (ABPs) targeting HNE can be designed to quantify the active catalytic state of 2 3 with only low efficacy partial agonism at both MOP and NOP receptors. Buprenorphine possesses partial MOP these enzymes . Following the extensive work of our group with HNE inhibitor synthesis a library of HNE 4 agonist activity and, importantly, low efficacy, modest potency NOP partial agonism. However, one study has inhibitors and ABPs based on the 3-oxo-β-sultam warhead was developed using the copper-assisted 5 Scheme 1 detected no NOP receptor involvement in buprenorphine-induced physiological responses in primates[2] and so azide-alkyne Huisgen cycloaddition ( ). The library was tested for inhibitory activity against HNE, an improved analgesic might have a buprenorphine-like profile but with enhanced NOP activity. To this end we showing inhibition values in the nanomolar region. The ABPs will target only the active form of HNE and will have synthesized novel buprenorphine-like analogs with increased NOP affinity and efficacy. be used to covalently tether a reporter tag to the enzyme, providing protein activity quantification on patient derived biospecimens in a MALDI-TOF-focused proteomics-based analysis. We envisage the study and validation of HNE as a potential biomarker in COPD. The outcome of this project will be a breakthrough in the field that ultimately will lead to important advances for COPD biomarker discovery and diagnosis.

Scheme 1: Copper-assisted azide-alkyne Huisgen cycloaddition in the synthesis of a 3-oxo-β-sultam based library of HNE inhibitors.

References 1) Lucas S. D.; Costa E.; Guedes R. C.; Moreira R. Med. Res. Rev. 2013, 33 Suppl 1, E73-e101. Fig 1. Structures of Buprenorphine and BU08028 2) Carvalho L. A. R.; Ruivo E. F. P.; Lucas S. D.; Moreira R. MedChemComm 2015, 6, 536. 3) Mulchande J.; Guedes R. C.; Tsang W.; Page M. I.; Moreira R.; Iley J. J. Med. Chem. 2008, 51, 1783. Structure-activity relationship studies suggested that the region of space occupied by the tert-butyl group in 4) Tsang W.Y.; Ahmed N.; Hemming K.; Page M. I. Org. Biomol. Chem. 2007, 5, 3993. buprenorphine was key to good NOP receptor activity.[3] We have discovered BU08028, and other compounds, 5) Speers A. E.; Adam G. C.; Cravatt, B. F.; J. Am. Chem. Soc. 2007, 125, 4686. having binding affinity at MOP receptors similar to that of buprenorphine and, as desired, higher affinity and considerably higher efficacy than buprenorphine at NOP receptors. BU08028 has been evaluated in vivo for attenuation of acute pain, using the tail flick assay and these results will be presented. In measures of hyperalgesia, both in rodents (rats) and non-human primates (rhesus monkey) BU08028 was a potent, long-acting anti-hyperalgesic with both MOP and NOP components to its activity. When administered alone in doses up to 0.01 mg/kg, BU08028 did not induce itch scratching, a standard side-effect of opioids such as buprenorphine. More importantly, BU08028 at antinociceptive doses, did not compromise physiological functions including respiration and cardiovascular activities measured using radio-telemetric probes. Compared to MOP agonists, buprenorphine and remifentanil, BU08028 did not produce reinforcing effects in monkeys under the progressive-ratio schedule of drug self-administration. This study strongly supports the therapeutic potential of ligands with mixed MOP/NOP actions as innovative analgesics in humans. BU08028 therefore represents a potential analgesic agent, with low side effect profile. This work was supported by NIDA grants DA020469 (SMH), DOD/Army W81XWH-13-2-0045 (M-ChKo) and DA023281 (LToll).

68 P009 P010 IMPROVED ANALGESICS: BU08028 A NOVEL, BIFUNCTIONAL OXO-β-SULTAMS AS ACTIVITY-BASED PROBES NOP/MOP LIGAND Luís Carvalho, Lídia Gonçalves, Rui Moreira, Susana Lucas Gerta Cami-Kobeci (1), Mei-Chuan Ko (2), Lawrence Toll (3), Stephen M. Husbands (1) Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal 1) Department of Pharmacy and Pharmacology, University of Bath, Bath, BA2 7AY, UK 2) Wake Forest University, Winston-Salem, North Carolina, USA 3) Torrey Pines Institute for Molecular Studies, Port St. Lucie, Fl, USA Chronic Obstructive Pulmonary Disease (COPD) is a condition characterized by lung inflammation and progressive weakening of its structure leading to an irreversible narrowing of the airways. It is reported that it will become the third leading cause of death worldwide in 2030 by the World Health Organization1. New Although mu opioid (MOP) analgesics, such as morphine, are the preferred analgesics, they possess well biomarkers to diagnose and predict the activity and progression of COPD are urgently needed. The central characterised, unwanted effects such as respiratory depression and abuse potential. hypothesis of this project is that COPD induced inflammation causes modulation of proteolytic enzymes in the When a NOP (nociceptin receptor) agonist is co-administered with a MOP agonist, the combination produces a lungs, like Human Neutrophil Elastase (HNE), which are clinically relevant for the diagnosis and prognosis of synergistic effect, resulting in enhanced analgesia.[1] This suggests the possibility of obtaining strong analgesia COPD. Activity-based probes (ABPs) targeting HNE can be designed to quantify the active catalytic state of 2 3 with only low efficacy partial agonism at both MOP and NOP receptors. Buprenorphine possesses partial MOP these enzymes . Following the extensive work of our group with HNE inhibitor synthesis a library of HNE 4 agonist activity and, importantly, low efficacy, modest potency NOP partial agonism. However, one study has inhibitors and ABPs based on the 3-oxo-β-sultam warhead was developed using the copper-assisted 5 Scheme 1 detected no NOP receptor involvement in buprenorphine-induced physiological responses in primates[2] and so azide-alkyne Huisgen cycloaddition ( ). The library was tested for inhibitory activity against HNE, an improved analgesic might have a buprenorphine-like profile but with enhanced NOP activity. To this end we showing inhibition values in the nanomolar region. The ABPs will target only the active form of HNE and will have synthesized novel buprenorphine-like analogs with increased NOP affinity and efficacy. be used to covalently tether a reporter tag to the enzyme, providing protein activity quantification on patient derived biospecimens in a MALDI-TOF-focused proteomics-based analysis. We envisage the study and validation of HNE as a potential biomarker in COPD. The outcome of this project will be a breakthrough in the field that ultimately will lead to important advances for COPD biomarker discovery and diagnosis.

Scheme 1: Copper-assisted azide-alkyne Huisgen cycloaddition in the synthesis of a 3-oxo-β-sultam based library of HNE inhibitors.

References 1) Lucas S. D.; Costa E.; Guedes R. C.; Moreira R. Med. Res. Rev. 2013, 33 Suppl 1, E73-e101. Fig 1. Structures of Buprenorphine and BU08028 2) Carvalho L. A. R.; Ruivo E. F. P.; Lucas S. D.; Moreira R. MedChemComm 2015, 6, 536. 3) Mulchande J.; Guedes R. C.; Tsang W.; Page M. I.; Moreira R.; Iley J. J. Med. Chem. 2008, 51, 1783. Structure-activity relationship studies suggested that the region of space occupied by the tert-butyl group in 4) Tsang W.Y.; Ahmed N.; Hemming K.; Page M. I. Org. Biomol. Chem. 2007, 5, 3993. buprenorphine was key to good NOP receptor activity.[3] We have discovered BU08028, and other compounds, 5) Speers A. E.; Adam G. C.; Cravatt, B. F.; J. Am. Chem. Soc. 2007, 125, 4686. having binding affinity at MOP receptors similar to that of buprenorphine and, as desired, higher affinity and considerably higher efficacy than buprenorphine at NOP receptors. BU08028 has been evaluated in vivo for attenuation of acute pain, using the tail flick assay and these results will be presented. In measures of hyperalgesia, both in rodents (rats) and non-human primates (rhesus monkey) BU08028 was a potent, long-acting anti-hyperalgesic with both MOP and NOP components to its activity. When administered alone in doses up to 0.01 mg/kg, BU08028 did not induce itch scratching, a standard side-effect of opioids such as buprenorphine. More importantly, BU08028 at antinociceptive doses, did not compromise physiological functions including respiration and cardiovascular activities measured using radio-telemetric probes. Compared to MOP agonists, buprenorphine and remifentanil, BU08028 did not produce reinforcing effects in monkeys under the progressive-ratio schedule of drug self-administration. This study strongly supports the therapeutic potential of ligands with mixed MOP/NOP actions as innovative analgesics in humans. BU08028 therefore represents a potential analgesic agent, with low side effect profile. This work was supported by NIDA grants DA020469 (SMH), DOD/Army W81XWH-13-2-0045 (M-ChKo) and DA023281 (LToll).

69 P011 P012 HUMAN VITAMIN K EPOXIDE REDUCTASE (HVKORC1): SEARCH DEVELOPMENT OF NEW SYNTHETIC INHIBITORS OF FOR THE FUNCTIONAL CONFORMATIONAL STATES MYCOBACTERIUM TUBERCULOSIS DprE2

Nolan Chatron (1), Bernard Chalmond (1), Alain Trouvé (1), Etienne Benoît (2), Virginie Lattard (2), Giacomo Chiodarelli (1,2), Szilvia Toth (3), Gurdyal S. Besra (3), Liam R. Cox (1), Joel Lelievre (2), Luba Tchertanov (1) Monica Cacho Izquierdo (2)

1) Centre de Mathématiques et de Leurs Applications (CMLA-UMR 8536 CNRS) 1) School of Chemistry, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK École Normale Supérieure de Cachan 2) Diseases of the Developing World, GlaxoSmithKline, Severo Ochoa 2, 28760 Tres Cantos, Madrid, Spain 61 avenue du Président Wilson 3) School of Biosciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK 94235 Cachan, France 2) INRA-VetAgro Sup, USC 1233 École Nationale Vétérinaire de Lyon 69280 Marcy l’Étoile, France Mycobacterium tuberculosis is the pathogen responsible for human tuberculosis (TB), which remains a leading killer, accounting for approximately 9 million cases and 1.7 million deaths every year.[1] The current TB treatment requires a six-month regimen involving four drugs, namely rifampicin, isoniazid, ethambutol and Human vitamin K epoxide reductase (hVKORC1), an endoplasmic reticulum-resident transmembrane protein, pyrazinamide. The length of the treatment can cause poor patient compliance, which has led to the emergence of transforms vitamin K epoxide into vitamin K quinone and hydroquinone. Through γ-carboxylation of glutamate resistant strains of the bacterium, leading to multi-drug resistant TB (MDR-TB) and extensively-drug resistant residues, vitamin K hydroquinone assists in activating vitamin K-dependent proteins (VKDPs), including blood TB (XDR-TB).[2] It is therefore important to develop more potent drugs, which allow shorter treatment times. clotting factors. hVKORC1 is thus a main target in anticoagulant treatments, usually prescribed to prevent DprE1 and DprE2 are two enzymes involved in the biosynthesis of decaprenyl thrombosis. However, several naturally occurring mutations lead to the hVKORC1 catalytic deregulation, monophosphoryl-D-arabinofuranose (DPA). Together, these two enzymes bring about the epimerisation of the promoting physiological troubles such as hemorrhaging disorders or anticoagulant resistance. of C(2) in the ribose unit of decaprenyl monophosphoryl-D-ribofuranose (DPR) to provide the arabinose To the best of our knowledge, crystallographic data is not available for hVKORC1 and its 3D structure remains sugar found in DPA. DPA is the only sugar donor used by the bacterium to synthesise arabinogalactan (AG) and experimentally uncharacterized. Moreover, biological data describing the hVKORC1 membrane topology and its lipoarabinomannan (LAM), two of the major constituents of the bacterial cell wall. Both enzymes are essential. catalytic mechanism are contradictory, leading to two topology models - 3 transmembrane (TM) helices and 4 [3] In contrast to DprE1, DprE2 has received far less attention.[4] The goal of this project is to develop a lead TM helices - and consequently, to two different visions onto the role of the two highly conserved cysteine compound from the DprE2 hit, GSK426032A, which was discovered by high-throughput screening by GSK in residues C43 and C51. Only the catalytic site, composed of a CxxC motif specific for oxidoreductases family Tres Cantos (Spain).[5] A detailed SAR investigation around this hit has been hindered by the lack of isolated proteins, is well defined. and purified DprE2, which has precluded access to crystal structures of the protein and hindered the development of enzymatic assays. Our approach to investigating the SAR around the hit has therefore involved synthesising In this work, we report a 3D model of hVKORC1, designed on the atomic scale, followed by molecular analogues and testing these against M. tuberculosis whole cells. The information derived from these SAR studies dynamics (MD) simulations (multiple 100-ns and one 500-ns trajectories). We aimed to describe the hVKORC1 can help us to obtain derivatives with improved physicochemical properties and a better safety profile. conformational space, and so to unravel the hVKORC1 catalytic mechanism. Classical analysis of MD data provides qualitative and limited insights into the simulated protein, so we completed the hVKORC1 characterization with data obtained by a novel mathematical method, ConfiScan. This new approach, based on spectral analysis, scans slow variables providing the energy profile description as well as a depiction the References configuration state(s) and their stability. 1) WHO, T.: Global tuberculosis report 2014. World Health Organization, Geneva Using classical methods and the newly developed approach, we (i) characterized the conformational space 2) Zumla, A.; Nahid, P.; Cole, S.T. Nature Rev. Drug Discov., 2013, 12, 388-404 explored by hVKORC1 over MD trajectories, (ii) attributed the catalytically significant conformations and (iii) 3) Mikušova, K.; Huang, H; Yagi, T.; Holsters, M.; Vereecke, D; D'Haeze, W.; Scherman, M.S.; Brennan, P.J.; McNeil, in silico M.R.; Crick, D.C. J Bacteriol, 2005, 187, 8020-8025 defined the protonation states of the cysteine residues in these conformations. The conformation of 4) Kolly, G.S.; Boldrin, F.; Sala, C.; Dhar, N.; Hartkoorn, R.C.; Ventura, M.; Serafini, A.; McKinney, J.D.; Manganelli, R.; hVKORC1 identified as the most probable corresponds perfectly to the active form of enzyme suggested by Cole, S.T. Mol Microbiol, 2014, 92, 194-211 experimental data. Molecular docking of vitamin K into this model produced results matching well with 5) Ballell, L., Bates, R.H., Young, R., Alvarez-Gomez, D., Alvarez-Ruiz, E., Barroso, V., Blanco, D., Crespo, B., Escribano, experimental data, confirming correctness of our structural model and the choice of the catalytically important J., González, R., Lozano, S, Huss, S., Santos‐Villarejo, A., Martín‐Plaza, J.J., Mendoza, A., Rebollo‐Lopez, M.J., conformation. Remuiñan‐Blanco, M., Lavandera, J.L., Pérez‐Herran, E., Gamo‐Benito, F.J., García‐Bustos, J.F., Barros, D., Castro, J., Cammack, N. ChemMedChem, 2013, 8, 313-321

References 1) Da-Yun Jin; Jian-Ke Tie; Darrel W.Stafford; 2007; The Conversion of Vitamin K Epoxide to Vitamin K Quinone and Vitamin K Quinone to Vitamin K Hydroquinone Uses the Same Active Site Cysteines; Biochemistry 2) Jian-Ke Tie; Da-Yun Jin; Darrel W.Stafford; 2012; Human Vitamin K Epoxide Reductase and Its Bacterial Homologue Have Different Membrane Topologies and Reaction Mechanisms; The Journal of Biological Chemistry 3) Johan Stenflo; Per Fernlund; William Egan; Peter Roepstorff; 1974; Vitamin K dependent modifications of glutamic acid residues in prothrombin; Proceeding of the National Academy of Sciences (PNAS) 4) S.Wu; Jian-Ke Tie; Darrel W.Stafford; L.G.Pedersen; 2013; Membrane topology for human vitamin K epoxide reductase; Journal of Thrombosis and Haemostasis 5) Shixuan Liu; Wei Chen; Ronald Fowle Grider; Guomin Shen; Weikai Li; 2014; Structures of an intramembrane vitamin K epoxide reductase homolog reveal control mechanisms for electron transfer; Nature Communications 6) Weikai Li; Sol Schulman; Rachel J.Dutton; Dana Boyd; Jon Beckwith; Tom A.Rapoport; 2010; Structure of a bacterial homologue of vitamin K epoxide reductase; Nature 7) Zhenbo Cao; Marcel van Lith; Lorna J.Mitchell; Marie Anne Pringle; Kenji Inaba; Neil J.Bulleid; 2016; The membrane topology of vitamin K epoxide reductase is conserved between human isoforms and the bacterial enzyme; Biochemical Journal

70 P011 P012 HUMAN VITAMIN K EPOXIDE REDUCTASE (HVKORC1): SEARCH DEVELOPMENT OF NEW SYNTHETIC INHIBITORS OF FOR THE FUNCTIONAL CONFORMATIONAL STATES MYCOBACTERIUM TUBERCULOSIS DprE2

Nolan Chatron (1), Bernard Chalmond (1), Alain Trouvé (1), Etienne Benoît (2), Virginie Lattard (2), Giacomo Chiodarelli (1,2), Szilvia Toth (3), Gurdyal S. Besra (3), Liam R. Cox (1), Joel Lelievre (2), Luba Tchertanov (1) Monica Cacho Izquierdo (2)

1) Centre de Mathématiques et de Leurs Applications (CMLA-UMR 8536 CNRS) 1) School of Chemistry, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK École Normale Supérieure de Cachan 2) Diseases of the Developing World, GlaxoSmithKline, Severo Ochoa 2, 28760 Tres Cantos, Madrid, Spain 61 avenue du Président Wilson 3) School of Biosciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK 94235 Cachan, France 2) INRA-VetAgro Sup, USC 1233 École Nationale Vétérinaire de Lyon 69280 Marcy l’Étoile, France Mycobacterium tuberculosis is the pathogen responsible for human tuberculosis (TB), which remains a leading killer, accounting for approximately 9 million cases and 1.7 million deaths every year.[1] The current TB treatment requires a six-month regimen involving four drugs, namely rifampicin, isoniazid, ethambutol and Human vitamin K epoxide reductase (hVKORC1), an endoplasmic reticulum-resident transmembrane protein, pyrazinamide. The length of the treatment can cause poor patient compliance, which has led to the emergence of transforms vitamin K epoxide into vitamin K quinone and hydroquinone. Through γ-carboxylation of glutamate resistant strains of the bacterium, leading to multi-drug resistant TB (MDR-TB) and extensively-drug resistant residues, vitamin K hydroquinone assists in activating vitamin K-dependent proteins (VKDPs), including blood TB (XDR-TB).[2] It is therefore important to develop more potent drugs, which allow shorter treatment times. clotting factors. hVKORC1 is thus a main target in anticoagulant treatments, usually prescribed to prevent DprE1 and DprE2 are two enzymes involved in the biosynthesis of decaprenyl thrombosis. However, several naturally occurring mutations lead to the hVKORC1 catalytic deregulation, monophosphoryl-D-arabinofuranose (DPA). Together, these two enzymes bring about the epimerisation of the promoting physiological troubles such as hemorrhaging disorders or anticoagulant resistance. alcohol of C(2) in the ribose unit of decaprenyl monophosphoryl-D-ribofuranose (DPR) to provide the arabinose To the best of our knowledge, crystallographic data is not available for hVKORC1 and its 3D structure remains sugar found in DPA. DPA is the only sugar donor used by the bacterium to synthesise arabinogalactan (AG) and experimentally uncharacterized. Moreover, biological data describing the hVKORC1 membrane topology and its lipoarabinomannan (LAM), two of the major constituents of the bacterial cell wall. Both enzymes are essential. catalytic mechanism are contradictory, leading to two topology models - 3 transmembrane (TM) helices and 4 [3] In contrast to DprE1, DprE2 has received far less attention.[4] The goal of this project is to develop a lead TM helices - and consequently, to two different visions onto the role of the two highly conserved cysteine compound from the DprE2 hit, GSK426032A, which was discovered by high-throughput screening by GSK in residues C43 and C51. Only the catalytic site, composed of a CxxC motif specific for oxidoreductases family Tres Cantos (Spain).[5] A detailed SAR investigation around this hit has been hindered by the lack of isolated proteins, is well defined. and purified DprE2, which has precluded access to crystal structures of the protein and hindered the development of enzymatic assays. Our approach to investigating the SAR around the hit has therefore involved synthesising In this work, we report a 3D model of hVKORC1, designed on the atomic scale, followed by molecular analogues and testing these against M. tuberculosis whole cells. The information derived from these SAR studies dynamics (MD) simulations (multiple 100-ns and one 500-ns trajectories). We aimed to describe the hVKORC1 can help us to obtain derivatives with improved physicochemical properties and a better safety profile. conformational space, and so to unravel the hVKORC1 catalytic mechanism. Classical analysis of MD data provides qualitative and limited insights into the simulated protein, so we completed the hVKORC1 characterization with data obtained by a novel mathematical method, ConfiScan. This new approach, based on spectral analysis, scans slow variables providing the energy profile description as well as a depiction the References configuration state(s) and their stability. 1) WHO, T.: Global tuberculosis report 2014. World Health Organization, Geneva Using classical methods and the newly developed approach, we (i) characterized the conformational space 2) Zumla, A.; Nahid, P.; Cole, S.T. Nature Rev. Drug Discov., 2013, 12, 388-404 explored by hVKORC1 over MD trajectories, (ii) attributed the catalytically significant conformations and (iii) 3) Mikušova, K.; Huang, H; Yagi, T.; Holsters, M.; Vereecke, D; D'Haeze, W.; Scherman, M.S.; Brennan, P.J.; McNeil, in silico M.R.; Crick, D.C. J Bacteriol, 2005, 187, 8020-8025 defined the protonation states of the cysteine residues in these conformations. The conformation of 4) Kolly, G.S.; Boldrin, F.; Sala, C.; Dhar, N.; Hartkoorn, R.C.; Ventura, M.; Serafini, A.; McKinney, J.D.; Manganelli, R.; hVKORC1 identified as the most probable corresponds perfectly to the active form of enzyme suggested by Cole, S.T. Mol Microbiol, 2014, 92, 194-211 experimental data. Molecular docking of vitamin K into this model produced results matching well with 5) Ballell, L., Bates, R.H., Young, R., Alvarez-Gomez, D., Alvarez-Ruiz, E., Barroso, V., Blanco, D., Crespo, B., Escribano, experimental data, confirming correctness of our structural model and the choice of the catalytically important J., González, R., Lozano, S, Huss, S., Santos‐Villarejo, A., Martín‐Plaza, J.J., Mendoza, A., Rebollo‐Lopez, M.J., conformation. Remuiñan‐Blanco, M., Lavandera, J.L., Pérez‐Herran, E., Gamo‐Benito, F.J., García‐Bustos, J.F., Barros, D., Castro, J., Cammack, N. ChemMedChem, 2013, 8, 313-321

References 1) Da-Yun Jin; Jian-Ke Tie; Darrel W.Stafford; 2007; The Conversion of Vitamin K Epoxide to Vitamin K Quinone and Vitamin K Quinone to Vitamin K Hydroquinone Uses the Same Active Site Cysteines; Biochemistry 2) Jian-Ke Tie; Da-Yun Jin; Darrel W.Stafford; 2012; Human Vitamin K Epoxide Reductase and Its Bacterial Homologue Have Different Membrane Topologies and Reaction Mechanisms; The Journal of Biological Chemistry 3) Johan Stenflo; Per Fernlund; William Egan; Peter Roepstorff; 1974; Vitamin K dependent modifications of glutamic acid residues in prothrombin; Proceeding of the National Academy of Sciences (PNAS) 4) S.Wu; Jian-Ke Tie; Darrel W.Stafford; L.G.Pedersen; 2013; Membrane topology for human vitamin K epoxide reductase; Journal of Thrombosis and Haemostasis 5) Shixuan Liu; Wei Chen; Ronald Fowle Grider; Guomin Shen; Weikai Li; 2014; Structures of an intramembrane vitamin K epoxide reductase homolog reveal control mechanisms for electron transfer; Nature Communications 6) Weikai Li; Sol Schulman; Rachel J.Dutton; Dana Boyd; Jon Beckwith; Tom A.Rapoport; 2010; Structure of a bacterial homologue of vitamin K epoxide reductase; Nature 7) Zhenbo Cao; Marcel van Lith; Lorna J.Mitchell; Marie Anne Pringle; Kenji Inaba; Neil J.Bulleid; 2016; The membrane topology of vitamin K epoxide reductase is conserved between human isoforms and the bacterial enzyme; Biochemical Journal

71 P013 P014 DEVELOPING HIGH THROUGHPUT SCREENS TO IDENTIFY ACRYLAMIDES AND INFLAMMASOMES: ANOTHER STEP SMALL MOLECULE ACTIVATORS OF THE ADULT EPICARDIUM TOWARDS DIRECT NLRP3 INHIBITION

Caitlin Clunie-O'Connor (1,4), Alison Howarth (2), Anke Smits (3), Angela Russell (4,5), Paul Riley (1) Mattia Cocco (1), Marta Giorgis (1), Gianluca Miglio (1), Elisa Bonino (1), Elisabetta Marini (1), Giulio Vistoli (2), Bénédicte F. Py (3), Massimo Bertinaria (1) 1) University of Oxford, Department of Physiology, Anatomy and Genetics, South Parks Road, Oxford, OX1 3PT, UK. 2) Nuffield Department of Medicine, University of Oxford Henry Wellcome Building for Molecular Physiology, Old Road 1) Dipartimento di Scienza e Tecnologia del Farmaco, Università degli Studi di Torino, Via P. Giuria 9, 10125 Torino (Italy) Campus, Oxford, OX3 7BN, UK. 2) Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, Via Mangiagalli 25, 20133 Milano (Italy) 3) Department of Molecular Cell Biology, Leiden University Medical Centre, Leiden, the Netherlands. 3) CIRI, International Center for Infectiology Research, Inserm U1111, CNRS UMR 5308, Ecole Normale Supérieure de 4) Department of Chemistry, Chemistry Research Laboratory, 12 Mansfield Road, Oxford OX1 3TA, United Kingdom. Lyon, Université Lyon 1, 21 avenue Tony Garnier, 69007 Lyon (France) 5) Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, United Kingdom.

NLRP3 inflammasome is a multi-proteic complex that plays a crucial role in activating caspase-1, processing the The epicardium plays an essential role during heart development. Cells derived from the epicardium (EPDCs) pro-inflammatory interleukin-1β (IL-1β), and triggering pyroptotic cell death cascade.1 Gain of function contribute essential cardiovascular cell types including vascular smooth muscle, interstitial fibroblasts, mutations in NLRP3 determine its abnormal activation which is a key factor in the pathogenesis of 1 endothelium and cardiomyocytes via the process of epithelial to mesenchymal transition (EMT). In addition to a autoinflammatory diseases known as cryopyrin-associated periodic syndromes (CAPS). The progression of other physical contribution, EPDC paracrine signalling is critical for cardiomyocyte differentiation, myocardial diseases, such as atherosclerosis, type-2 diabetes, Alzheimer’s disease, and gout is also dependent on NLRP3 2 compaction and proper organ formation. inflammasome activation.2 Several strategies have been proposed to dampen NLRP3 activity, and among them Whilst dormant in the adult heart, the epicardium becomes reactivated in response to injury in both mouse and covalent drugs seem to be the most promising ones.3 zebrafish. Activation is characterised by epicardial expansion, EMT and re-expression of embryonic In this study, a series of newly-synthesized acrylamide compounds is evaluated as potential NLRP3 network Tbx18 Wt1 3 transcription factors including and . Moreover priming the mouse heart with thymosin β4 (Tβ4) inhibitors by studying their pharmaco-toxicological profile. Low toxicity profile was obtained by tuning down Wt1+ in vivo increases the number of EPDCs following myocardial infarction. Subsequently small numbers of Michael acceptor reactivity which translated into cell viability enhancement and serum albumin binding Wt1+ 4 Tβ4 activated cells migrate into the wound forming functional cardiomyocytes. The number of decrease. These Michael acceptors were also able to inhibit IL-1β release from different macrophage subtypes, functional EPDC-derived cardiomyocytes is suboptimal to restore the lost heart muscle, therefore, we seek to including CAPS mutant macrophages. Mechanism of action of these acrylamides involves direct interaction with augment the process using chemical or genetic approaches to fully exploit the epicardium as a source of resident NLRP3, confirmed by NLRP3 ATPase activity inhibition on isolated protein. NLRP3 ATPase was adult stem cells for endogenous regenerative therapy. concentration-dependently inhibited by 2-(2-Chlorobenzyl)-N-(4-sulfamoylphenethyl)acrylamide (INF58) with Using both primary human patient-derived epicardial cells5 and an immortalised murine epicardial cell line6 we an IC50 of 74 µM: in silico prediction of its binding mode in the catalytic pocket indicates that a putative have established in vitro models of epicardial EMT. Subsequently we have developed two high throughput interaction with Cys419 residue might account for this activity. This study shows that these acrylamides could be screening assays for the identification of small molecules that stimulate epicardial activation and studies are good candidates to develop safe covalent inhibitors of NLRP3 inflammasome.4 ongoing to validate primary hits that promote EMT and subsequent EPDC differentiation.

References 1) Wessels, A. & Pérez-Pomares, J. M. The epicardium and epicardially derived cells (EPDCs) as cardiac stem cells. Anat. Rec. A. Discov. Mol. Cell. Evol. Biol. 276, 43–57 (2004). 2) Kang, J.-O. & Sucov, H. M. Convergent proliferative response and divergent morphogenic pathways induced by epicardial and endocardial signaling in fetal heart development. Mech. Dev. 122, 57–65 (2005). 3) González-Rosa, J. M., Martín, V., Peralta, M., Torres, M. & Mercader, N. Extensive scar formation and regression during heart regeneration after cryoinjury in zebrafish. Dev. Camb. Engl. 138, 1663–1674 (2011). 4) Smart, N. et al. De novo cardiomyocytes from within the activated adult heart after injury. Nature 474, 640–644 (2011). 5) Clunie-O’Connor, C. et al. The Derivation of Primary Human Epicardium-Derived Cells. Curr. Protoc. Stem Cell Biol. 35, 2C.5.1–2C.5.12 (2015). 6) Austin, A. F., Compton, L. A., Love, J. D., Brown, C. B. & Barnett, J. V. Primary and immortalized mouse epicardial cells undergo differentiation in response to TGFβ. Dev. Dyn. 237, 366–376 (2008).

References 1) V. Compan, F. Martın-Sanchez, A. Baroja-Mazo, G. Lopez-Castejon, A. I. Gomez, A. Verkhratsky, D. Brough, P. Pelegrın, J. Immunol. 2015, 194, 1261-1273 2) A. G. Baldwin, D. Brough, S. Freeman, J. Med. Chem. 2016, 59, 1691-1710 3) M. Cocco, D. Garella, A. Di Stilo, E. Borretto, L. Stevanato, M. Giorgis, E. Marini, R. Fantozzi, G. Miglio, M. Bertinaria, J. Med. Chem. 2014, 57, 10366-10382 4) M. Cocco, G. Miglio, M. Giorgis, D. Garella, E. Marini, A. Costale, L. Regazzoni, G. Vistoli, M. Orioli, R. Massulaha-Ahmed, I. Détraz-Durieux, M. Groslambert, B. F. Py, M. Bertinaria ChemMedChem DOI: 10.1002/cmdc.201600055

72 P013 P014 DEVELOPING HIGH THROUGHPUT SCREENS TO IDENTIFY ACRYLAMIDES AND INFLAMMASOMES: ANOTHER STEP SMALL MOLECULE ACTIVATORS OF THE ADULT EPICARDIUM TOWARDS DIRECT NLRP3 INHIBITION

Caitlin Clunie-O'Connor (1,4), Alison Howarth (2), Anke Smits (3), Angela Russell (4,5), Paul Riley (1) Mattia Cocco (1), Marta Giorgis (1), Gianluca Miglio (1), Elisa Bonino (1), Elisabetta Marini (1), Giulio Vistoli (2), Bénédicte F. Py (3), Massimo Bertinaria (1) 1) University of Oxford, Department of Physiology, Anatomy and Genetics, South Parks Road, Oxford, OX1 3PT, UK. 2) Nuffield Department of Medicine, University of Oxford Henry Wellcome Building for Molecular Physiology, Old Road 1) Dipartimento di Scienza e Tecnologia del Farmaco, Università degli Studi di Torino, Via P. Giuria 9, 10125 Torino (Italy) Campus, Oxford, OX3 7BN, UK. 2) Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, Via Mangiagalli 25, 20133 Milano (Italy) 3) Department of Molecular Cell Biology, Leiden University Medical Centre, Leiden, the Netherlands. 3) CIRI, International Center for Infectiology Research, Inserm U1111, CNRS UMR 5308, Ecole Normale Supérieure de 4) Department of Chemistry, Chemistry Research Laboratory, 12 Mansfield Road, Oxford OX1 3TA, United Kingdom. Lyon, Université Lyon 1, 21 avenue Tony Garnier, 69007 Lyon (France) 5) Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, United Kingdom.

NLRP3 inflammasome is a multi-proteic complex that plays a crucial role in activating caspase-1, processing the The epicardium plays an essential role during heart development. Cells derived from the epicardium (EPDCs) pro-inflammatory interleukin-1β (IL-1β), and triggering pyroptotic cell death cascade.1 Gain of function contribute essential cardiovascular cell types including vascular smooth muscle, interstitial fibroblasts, mutations in NLRP3 determine its abnormal activation which is a key factor in the pathogenesis of 1 endothelium and cardiomyocytes via the process of epithelial to mesenchymal transition (EMT). In addition to a autoinflammatory diseases known as cryopyrin-associated periodic syndromes (CAPS). The progression of other physical contribution, EPDC paracrine signalling is critical for cardiomyocyte differentiation, myocardial diseases, such as atherosclerosis, type-2 diabetes, Alzheimer’s disease, and gout is also dependent on NLRP3 2 compaction and proper organ formation. inflammasome activation.2 Several strategies have been proposed to dampen NLRP3 activity, and among them Whilst dormant in the adult heart, the epicardium becomes reactivated in response to injury in both mouse and covalent drugs seem to be the most promising ones.3 zebrafish. Activation is characterised by epicardial expansion, EMT and re-expression of embryonic In this study, a series of newly-synthesized acrylamide compounds is evaluated as potential NLRP3 network Tbx18 Wt1 3 transcription factors including and . Moreover priming the mouse heart with thymosin β4 (Tβ4) inhibitors by studying their pharmaco-toxicological profile. Low toxicity profile was obtained by tuning down Wt1+ in vivo increases the number of EPDCs following myocardial infarction. Subsequently small numbers of Michael acceptor reactivity which translated into cell viability enhancement and serum albumin binding Wt1+ 4 Tβ4 activated cells migrate into the wound forming functional cardiomyocytes. The number of decrease. These Michael acceptors were also able to inhibit IL-1β release from different macrophage subtypes, functional EPDC-derived cardiomyocytes is suboptimal to restore the lost heart muscle, therefore, we seek to including CAPS mutant macrophages. Mechanism of action of these acrylamides involves direct interaction with augment the process using chemical or genetic approaches to fully exploit the epicardium as a source of resident NLRP3, confirmed by NLRP3 ATPase activity inhibition on isolated protein. NLRP3 ATPase was adult stem cells for endogenous regenerative therapy. concentration-dependently inhibited by 2-(2-Chlorobenzyl)-N-(4-sulfamoylphenethyl)acrylamide (INF58) with Using both primary human patient-derived epicardial cells5 and an immortalised murine epicardial cell line6 we an IC50 of 74 µM: in silico prediction of its binding mode in the catalytic pocket indicates that a putative have established in vitro models of epicardial EMT. Subsequently we have developed two high throughput interaction with Cys419 residue might account for this activity. This study shows that these acrylamides could be screening assays for the identification of small molecules that stimulate epicardial activation and studies are good candidates to develop safe covalent inhibitors of NLRP3 inflammasome.4 ongoing to validate primary hits that promote EMT and subsequent EPDC differentiation.

References 1) Wessels, A. & Pérez-Pomares, J. M. The epicardium and epicardially derived cells (EPDCs) as cardiac stem cells. Anat. Rec. A. Discov. Mol. Cell. Evol. Biol. 276, 43–57 (2004). 2) Kang, J.-O. & Sucov, H. M. Convergent proliferative response and divergent morphogenic pathways induced by epicardial and endocardial signaling in fetal heart development. Mech. Dev. 122, 57–65 (2005). 3) González-Rosa, J. M., Martín, V., Peralta, M., Torres, M. & Mercader, N. Extensive scar formation and regression during heart regeneration after cryoinjury in zebrafish. Dev. Camb. Engl. 138, 1663–1674 (2011). 4) Smart, N. et al. De novo cardiomyocytes from within the activated adult heart after injury. Nature 474, 640–644 (2011). 5) Clunie-O’Connor, C. et al. The Derivation of Primary Human Epicardium-Derived Cells. Curr. Protoc. Stem Cell Biol. 35, 2C.5.1–2C.5.12 (2015). 6) Austin, A. F., Compton, L. A., Love, J. D., Brown, C. B. & Barnett, J. V. Primary and immortalized mouse epicardial cells undergo differentiation in response to TGFβ. Dev. Dyn. 237, 366–376 (2008).

References 1) V. Compan, F. Martın-Sanchez, A. Baroja-Mazo, G. Lopez-Castejon, A. I. Gomez, A. Verkhratsky, D. Brough, P. Pelegrın, J. Immunol. 2015, 194, 1261-1273 2) A. G. Baldwin, D. Brough, S. Freeman, J. Med. Chem. 2016, 59, 1691-1710 3) M. Cocco, D. Garella, A. Di Stilo, E. Borretto, L. Stevanato, M. Giorgis, E. Marini, R. Fantozzi, G. Miglio, M. Bertinaria, J. Med. Chem. 2014, 57, 10366-10382 4) M. Cocco, G. Miglio, M. Giorgis, D. Garella, E. Marini, A. Costale, L. Regazzoni, G. Vistoli, M. Orioli, R. Massulaha-Ahmed, I. Détraz-Durieux, M. Groslambert, B. F. Py, M. Bertinaria ChemMedChem DOI: 10.1002/cmdc.201600055

73 P015 P016 SYNTHESIS OF NR ANALOGUES TO INHIBIT GLUCOSE TARGETING ASPRS IN THE FIGHT AGAINST M. TUBERCULOSIS METABOLISM IN CANCER CELLS Bogdan M. Duma (1,3), Ramon Soto (2,3), Liam R. Cox (1), Gurdyal S. Besra (2), Monica Edgar de las Heras Ruiz (1), Sam Butterworth (1), James Tucker (1), Dan Tennant (2) Cacho-Izquierdo (3), Joel Lelievre (3)

1) School of Chemistry, University of Birmingham 1) School of Chemistry, University of Birmingham, Edgbaston, B15 2TT, United Kingdom 2) Medical School, University of Birmingham 2) School of Biosciences, University of Birmingham, Edgbaston, B15 2TT, United Kingdom 3) Tuberculosis DPU, Tres Cantos Medicines Development Campus-DDW, GlaxoSmithKline (GSK), Tres Cantos, Spain

Cancer cells depend on the glycolysis pathway to produce energy (ATP), in contrast with normal cells which use Tuberculosis (TB) is an infectious bacterial disease caused by Mycobacterium tuberculosis. Commonly affecting mainly the Krebs cycle. Thus, glycolysis inhibition may allow to selectively kill cancer cells over normal cells. the lungs, TB has been ranked by the World Health Organization (WHO), as a leading cause of death worldwide, A potential enzymatic target in the glycolysis of cancer cells is the upregulated lactate dehydrogenase A causing 1.5 million fatalities in 2014 alone. Moreover, a daunting 9.6 million people are estimated to have been (LDHA), which is NAD-dependent. The aim of this project consists of designing and synthesizing redox inactive infected with TB in the same year.[1] NAD analogues, which would compete with NAD to bind and inhibit LDHA.1 With current treatment programs under significant threat from multi-drug and extensively-drug resistant strains NAD is biosynthesized from riboside (NR), which is phosphorylated by NRK, and then recognized of M. tuberculosis,[2] new cellular targets are necessary to effectively fight these bacteria. To overcome this we by NMNAT to add the adenine mononucleotide (part in red) of NAD. Thus, redox inactive NAD analogues decided to target aspartyl-tRNA synthetase (AspRS), an enzyme that to the best of our knowledge has not been should only resemble NR, but where the C-N bond is replaced by a C-C bond at the anomeric centre, like in targeted so far against TB. Using the AspRS enzymatic assay reported by Gurcha et al.[2] a screening was Benzamide Riboside (BR). BR is recognized by NRK, and NMNAT to produce a redox inactive NAD analogue performed using the known TB inhibitors published by Ballel et al. [3] as part of the GSK TB set. (BAD).1 Herein, we would like to report the preliminary results obtained from the screening process and the synthetic methodology we followed to develop the SAR for those results deemed interesting.

References 1) WHO Global Tuberculosis Report 2015 2) Gurcha, S. S. et al. PLoS ONE 2014, 9, e113568 3) Ballell, L. et al. ChemMedChem, 2013, 8, 313-321

Previous synthetic approaches to form the key C-C bond showed low yield, and lack of diastereoselectivity, however, alkylation of tribenzylated ribonolactone followed by diastereoselective deoxygenation provided the desired β-anomer in good yields.2 The high price of the tribenzylated ribonolactone (130 pounds, 5 g, 12 mmol) led to its synthesis from a cheaper commercial source, D-ribose (15 pounds, 5 g, 33 mmol). We managed to validate the synthesis of tribenzylated ribonolactone, and optimize it to make it more reproducible, with higher yielding, and scalable (16 g). The diastereoselectivity of the BR synthesis led us to its generalization towards any NR analogue (ongoing work).

References 1) J. R. Revollo, A. A. Grimm, and S. I. Imai, J. Biol. Chem. 2014, 279, 50754-50763 2) K. W. Pankiewicz et al, Nucleosides. Nucleotides Nucleic Acids, 2007, 26, 1249

74 P015 P016 SYNTHESIS OF NR ANALOGUES TO INHIBIT GLUCOSE TARGETING ASPRS IN THE FIGHT AGAINST M. TUBERCULOSIS METABOLISM IN CANCER CELLS Bogdan M. Duma (1,3), Ramon Soto (2,3), Liam R. Cox (1), Gurdyal S. Besra (2), Monica Edgar de las Heras Ruiz (1), Sam Butterworth (1), James Tucker (1), Dan Tennant (2) Cacho-Izquierdo (3), Joel Lelievre (3)

1) School of Chemistry, University of Birmingham 1) School of Chemistry, University of Birmingham, Edgbaston, B15 2TT, United Kingdom 2) Medical School, University of Birmingham 2) School of Biosciences, University of Birmingham, Edgbaston, B15 2TT, United Kingdom 3) Tuberculosis DPU, Tres Cantos Medicines Development Campus-DDW, GlaxoSmithKline (GSK), Tres Cantos, Spain

Cancer cells depend on the glycolysis pathway to produce energy (ATP), in contrast with normal cells which use Tuberculosis (TB) is an infectious bacterial disease caused by Mycobacterium tuberculosis. Commonly affecting mainly the Krebs cycle. Thus, glycolysis inhibition may allow to selectively kill cancer cells over normal cells. the lungs, TB has been ranked by the World Health Organization (WHO), as a leading cause of death worldwide, A potential enzymatic target in the glycolysis of cancer cells is the upregulated lactate dehydrogenase A causing 1.5 million fatalities in 2014 alone. Moreover, a daunting 9.6 million people are estimated to have been (LDHA), which is NAD-dependent. The aim of this project consists of designing and synthesizing redox inactive infected with TB in the same year.[1] NAD analogues, which would compete with NAD to bind and inhibit LDHA.1 With current treatment programs under significant threat from multi-drug and extensively-drug resistant strains NAD is biosynthesized from nicotinamide riboside (NR), which is phosphorylated by NRK, and then recognized of M. tuberculosis,[2] new cellular targets are necessary to effectively fight these bacteria. To overcome this we by NMNAT to add the adenine mononucleotide (part in red) of NAD. Thus, redox inactive NAD analogues decided to target aspartyl-tRNA synthetase (AspRS), an enzyme that to the best of our knowledge has not been should only resemble NR, but where the C-N bond is replaced by a C-C bond at the anomeric centre, like in targeted so far against TB. Using the AspRS enzymatic assay reported by Gurcha et al.[2] a screening was Benzamide Riboside (BR). BR is recognized by NRK, and NMNAT to produce a redox inactive NAD analogue performed using the known TB inhibitors published by Ballel et al. [3] as part of the GSK TB set. (BAD).1 Herein, we would like to report the preliminary results obtained from the screening process and the synthetic methodology we followed to develop the SAR for those results deemed interesting.

References 1) WHO Global Tuberculosis Report 2015 2) Gurcha, S. S. et al. PLoS ONE 2014, 9, e113568 3) Ballell, L. et al. ChemMedChem, 2013, 8, 313-321

Previous synthetic approaches to form the key C-C bond showed low yield, and lack of diastereoselectivity, however, alkylation of tribenzylated ribonolactone followed by diastereoselective deoxygenation provided the desired β-anomer in good yields.2 The high price of the tribenzylated ribonolactone (130 pounds, 5 g, 12 mmol) led to its synthesis from a cheaper commercial source, D-ribose (15 pounds, 5 g, 33 mmol). We managed to validate the synthesis of tribenzylated ribonolactone, and optimize it to make it more reproducible, with higher yielding, and scalable (16 g). The diastereoselectivity of the BR synthesis led us to its generalization towards any NR analogue (ongoing work).

References 1) J. R. Revollo, A. A. Grimm, and S. I. Imai, J. Biol. Chem. 2014, 279, 50754-50763 2) K. W. Pankiewicz et al, Nucleosides. Nucleotides Nucleic Acids, 2007, 26, 1249

75 P017 P018 SYNTHESIS AND ANTIBACTERIAL ACTIVITY OF FLUORESCENT RECENT ADVANCES IN ANTITUMOUR BERBERINES QUINOLONES Gaetano Fiorillo, Tanjia Monir Syeda, Paolo Lombardi Joanna Fedorowicz (1), Jarosław Sączewski (1), Agnieszka Konopacka (2), Krzysztof Waleron (2) Naxospharma srl, via Giuseppe Di Vittorio, 70, 20026, Novate Milanese, Italy [email protected] 1) Medical University of Gdańsk, Faculty of Pharmacy with Subfaculty of Laboratory Medicine, Department of Organic [email protected] Chemistry 2) Medical University of Gdańsk, Faculty of Pharmacy with Subfaculty of Laboratory Medicine, Department of Pharmaceutical Microbiology

Berberine (1), an isoquinoline quaternary alkaloid isolated from many kinds of medicinal plants, has drawn extensive attention as a therapeutic against diseases including hyperlipidemia, diabetes, metabolic syndrome, Antimicrobial resistance (AMR) threatens the effective treatment of an ever-increasing range of infections polycystic ovary syndrome, obesity, fatty liver disease, coronary artery disease.[1] The diverse pharmacological caused by bacteria, parasites, viruses and fungi. Thus very important is development of new compounds with properties exhibited by berberine indicate that the alkaloid has a definite potential as drug in a wide spectrum of high biological activity [1]. clinical applications. Recent studies have also shown that berberine inhibits cancer cell proliferation and migration, and induces apoptosis in a variety of cancer cell lines, stimulating further development of derivatives Quinolones and fluoroquinolons exhibit antibacterial effect through inhibition of gyrase activity (preventing for cancer prevention and treatment.[2] Therefore, the structure of berberine represents a biologically interesting bacterial DNA from unwinding and duplicating). Furthermore quaternary ammonium compounds increase the skeleton by providing an attractive natural lead for the introduction of various chemical modifications in permeability of cell membrane, cause dissociation of lipid bilayers and induces leakage of cellular contents. Our appropriate positions, in search for more selective medical indications. In this respect we discovered novel aim was to synthetize a new, fluorescent, antibacterial drugs containing quinolone or fluoroquinolone core 13-(di)(hetero)arylalkyl berberine derivatives (2) with improved anticancer properties.[3] The derivatives bear exhibiting synergistic enhancement of antimicrobial activity and specific optical properties allowing fluorescent (hetero)aromatic groups bonded to position 13 of the parent alkaloid skeleton through a linker of variable length imaging. and functionality, in a fashion to possibly create a geometric propensity for additional stacking-type, The synthesis fluorescently labeled antibacterial agents employed tandem Mannich – electrophilic amination non-covalent aromatic interactions with cellular targets. It is well known that aromatic interactions are reaction of profluorophoric isoxazolo[3,4-b]quinolin-3(1H)-one (Safirinium Q derivatives) or ubiquitous in nature and their geometry plays a central role in the molecular interaction with biological 4,6-dimethylisoxazolo[3,4-b]pyridin-3(1H)-one (Safirinium P derivatives) with formaldehyde and N macromolecules. [4] The derivatives can be prepared starting from 7,8-dihydroberberine (3) by using various -unsubstituted piperazynyl groups of corresponding quinolones or fluoroquinolones [2]. The obtained synthetic methodologies, including an uncommon aldehyde-enamine condensation. Although the precise compounds were isolated by non-chromatographic methods, the zwitterions were converted quantitatively into molecular basis of the many pharmacological activities of berberine is still debated, the anticancer activity of the the hydrochlorides with methanolic solution of HCl and in this form antibacterial activity and MICs were alkaloid appears to derive from its ability to form strong complexes with nucleic acids, induce DNA damage, and determined. exert related effects such as telomerase inhibition, topoisomerase poisoning, and inhibition of gene transcription. In comparison to berberine, several of the new derivatives were found to form stronger complexes with DNA We hypothesize that the use the fluorescent techniques and analysis of the intensity and distribution of [5a-h] and showed remarkable antiproliferative effects on a variety of human cancer cell lines which either fluorescence in vivo, would make it possible to visualize the distribution and interaction site of the drugs of acquired resistance or are normally refractory to chemotherapy. [5f, 6a-e] interest.

Aknowledgements: Financial support was provided by Ministero dello Sviluppo Economico (Grant 01705) under the 6th call of the EuroTransBio initiative, transnational project BERTA (BERberine as antiTumour Agents).

References 1) M. Tillhon, L.M. Guamàn Ortiz, P. Lombardi, A.I. Scovassi, Biochem Pharmacol, 2012, 84, 1260 2) L.M. Guaman-Ortiz, P. Lombardi, Tillhon, M. Scovassi, A.I. Molecules, 2014, 19, 12349 3) US8188109B2 to Naxospharma srl 4) Waters M.L.. Curr Opin Chem Biol 2002, 6: 736. 5) (a) Bhowmik, D.; Hossain, M.; Buzzetti, F.; D’Auria, R.; Lombardi, P.; Kumar, G.S. J. Phys. Chem. B, 2012, 116, 2314. (b) Bhowmik, D.; Buzzetti, F.; Fiorillo, G.; Lombardi, P.; Kumar, G.S. Spectrochimica Acta Part A, 2014, 120, 257. (c) Bhowmik, D.; Buzzetti, F.; Fiorillo, G.; Orzi, F.; Syeda Monir, T.; Lombardi, P.; Kumar, G.S. Med Chem Comm, 2014, 5, 226-. (d) Bhowmik, D.; Buzzetti, F.; Fiorillo, G.; Franchini, L.; Syeda Monir, T.; Lombardi, P.; Kumar, G.S. J Therm Anal Calorim, 2014, 118, 461. (e) Vieira, S.; Castelli, S.; Falconi, M.; Takarada, J.; Fiorillo, G.; Buzzetti, F.; Lombardi, P.; Desideri, A.. Int J Biological Macromolecules, 2015, 77, 68. (f) Bhowmik, D.; Fiorillo, G.; Lombardi, P.; Kumar, G.S. J.Mol. Recognit. 2015; 28: 722. (g) Chatterjee, S.; Mallick, S.; Das Saha, K.; Buzzetti, F.; Fiorillo, G.; Syeda Monir, T.; Lombardi, P.; Kumar, G.S. RSC Advances 2015, 5, 90632. (h) Ferraroni,M.; Bazzicalupi,C.; Papi, F.; Fiorillo, G.; Guaman-Ortiz, L.M.; References Nocentini, A.; Scovassi, A.I.; Lombardi, P.; Gratteri, P. Chem. Asian J. 2016, 11, 1107 6) (a) Albring, K.F.; Weidemueller, J.; Mittag S.; Weiske, J.; Friedrich, K.; Geroni, C.; Lombardi, P.; Huber, O. BioFactors, 1) Antimicrobial resistance: global report on surveillance 2014, WHO, April 2014 2013, 39, 652. (b) Pierpaoli, E.; Arcamone, A.G.; Buzzetti, F.; Lombardi, P.; Salvatore, C.; Provinciali, M. BioFactors, 2013, 2) Jarosław Sączewski, Krzysztof Hinc, Michał Obuchowski Maria Gdaniec; The Tandem Mannich-Electrophilic Amination 39, 672. (c) Guaman-Ortiz, L.M.; Tillhon, M.; Parks, M.; Dutto, I.; Prosperi, E.; Savio, M.; Arcamone, A.; Buzzetti, F.; Reaction: a Versatile Platform for Fluorescent Probing and Labeling; Chem. Eur. J. 2013, 19, 11531-11535 Lombardi, P.; Scovassi, A.I. BioMed Res Int, 2014, Article ID 924585. (d) Guaman-Ortiz, L.M.; Croce, A.; Aredia, F.; Fiorillo, G.; Syeda Monir, T.; Buzzetti, F.; Lombardi, P.; Scovassi, A.I.. Acta Biochim Biophys Sinica. 2015, 47, 824. (e) Pierpaoli, E.; Damiani, E.; Orlando, F.; Lucarini, G.; Bartozzi, B.; Lombardi, P.; Salvatore, C.; Geroni, C.; Donati, A.; 76 Provinciali, M. Carcinogenesis, 2015, 10, 1169 P017 P018 SYNTHESIS AND ANTIBACTERIAL ACTIVITY OF FLUORESCENT RECENT ADVANCES IN ANTITUMOUR BERBERINES QUINOLONES Gaetano Fiorillo, Tanjia Monir Syeda, Paolo Lombardi Joanna Fedorowicz (1), Jarosław Sączewski (1), Agnieszka Konopacka (2), Krzysztof Waleron (2) Naxospharma srl, via Giuseppe Di Vittorio, 70, 20026, Novate Milanese, Italy [email protected] 1) Medical University of Gdańsk, Faculty of Pharmacy with Subfaculty of Laboratory Medicine, Department of Organic [email protected] Chemistry 2) Medical University of Gdańsk, Faculty of Pharmacy with Subfaculty of Laboratory Medicine, Department of Pharmaceutical Microbiology

Berberine (1), an isoquinoline quaternary alkaloid isolated from many kinds of medicinal plants, has drawn extensive attention as a therapeutic against diseases including hyperlipidemia, diabetes, metabolic syndrome, Antimicrobial resistance (AMR) threatens the effective treatment of an ever-increasing range of infections polycystic ovary syndrome, obesity, fatty liver disease, coronary artery disease.[1] The diverse pharmacological caused by bacteria, parasites, viruses and fungi. Thus very important is development of new compounds with properties exhibited by berberine indicate that the alkaloid has a definite potential as drug in a wide spectrum of high biological activity [1]. clinical applications. Recent studies have also shown that berberine inhibits cancer cell proliferation and migration, and induces apoptosis in a variety of cancer cell lines, stimulating further development of derivatives Quinolones and fluoroquinolons exhibit antibacterial effect through inhibition of gyrase activity (preventing for cancer prevention and treatment.[2] Therefore, the structure of berberine represents a biologically interesting bacterial DNA from unwinding and duplicating). Furthermore quaternary ammonium compounds increase the skeleton by providing an attractive natural lead for the introduction of various chemical modifications in permeability of cell membrane, cause dissociation of lipid bilayers and induces leakage of cellular contents. Our appropriate positions, in search for more selective medical indications. In this respect we discovered novel aim was to synthetize a new, fluorescent, antibacterial drugs containing quinolone or fluoroquinolone core 13-(di)(hetero)arylalkyl berberine derivatives (2) with improved anticancer properties.[3] The derivatives bear exhibiting synergistic enhancement of antimicrobial activity and specific optical properties allowing fluorescent (hetero)aromatic groups bonded to position 13 of the parent alkaloid skeleton through a linker of variable length imaging. and functionality, in a fashion to possibly create a geometric propensity for additional stacking-type, The synthesis fluorescently labeled antibacterial agents employed tandem Mannich – electrophilic amination non-covalent aromatic interactions with cellular targets. It is well known that aromatic interactions are reaction of profluorophoric isoxazolo[3,4-b]quinolin-3(1H)-one (Safirinium Q derivatives) or ubiquitous in nature and their geometry plays a central role in the molecular interaction with biological 4,6-dimethylisoxazolo[3,4-b]pyridin-3(1H)-one (Safirinium P derivatives) with formaldehyde and N macromolecules. [4] The derivatives can be prepared starting from 7,8-dihydroberberine (3) by using various -unsubstituted piperazynyl groups of corresponding quinolones or fluoroquinolones [2]. The obtained synthetic methodologies, including an uncommon aldehyde-enamine condensation. Although the precise compounds were isolated by non-chromatographic methods, the zwitterions were converted quantitatively into molecular basis of the many pharmacological activities of berberine is still debated, the anticancer activity of the the hydrochlorides with methanolic solution of HCl and in this form antibacterial activity and MICs were alkaloid appears to derive from its ability to form strong complexes with nucleic acids, induce DNA damage, and determined. exert related effects such as telomerase inhibition, topoisomerase poisoning, and inhibition of gene transcription. In comparison to berberine, several of the new derivatives were found to form stronger complexes with DNA We hypothesize that the use the fluorescent techniques and analysis of the intensity and distribution of [5a-h] and showed remarkable antiproliferative effects on a variety of human cancer cell lines which either fluorescence in vivo, would make it possible to visualize the distribution and interaction site of the drugs of acquired resistance or are normally refractory to chemotherapy. [5f, 6a-e] interest.

Aknowledgements: Financial support was provided by Ministero dello Sviluppo Economico (Grant 01705) under the 6th call of the EuroTransBio initiative, transnational project BERTA (BERberine as antiTumour Agents).

References 1) M. Tillhon, L.M. Guamàn Ortiz, P. Lombardi, A.I. Scovassi, Biochem Pharmacol, 2012, 84, 1260 2) L.M. Guaman-Ortiz, P. Lombardi, Tillhon, M. Scovassi, A.I. Molecules, 2014, 19, 12349 3) US8188109B2 to Naxospharma srl 4) Waters M.L.. Curr Opin Chem Biol 2002, 6: 736. 5) (a) Bhowmik, D.; Hossain, M.; Buzzetti, F.; D’Auria, R.; Lombardi, P.; Kumar, G.S. J. Phys. Chem. B, 2012, 116, 2314. (b) Bhowmik, D.; Buzzetti, F.; Fiorillo, G.; Lombardi, P.; Kumar, G.S. Spectrochimica Acta Part A, 2014, 120, 257. (c) Bhowmik, D.; Buzzetti, F.; Fiorillo, G.; Orzi, F.; Syeda Monir, T.; Lombardi, P.; Kumar, G.S. Med Chem Comm, 2014, 5, 226-. (d) Bhowmik, D.; Buzzetti, F.; Fiorillo, G.; Franchini, L.; Syeda Monir, T.; Lombardi, P.; Kumar, G.S. J Therm Anal Calorim, 2014, 118, 461. (e) Vieira, S.; Castelli, S.; Falconi, M.; Takarada, J.; Fiorillo, G.; Buzzetti, F.; Lombardi, P.; Desideri, A.. Int J Biological Macromolecules, 2015, 77, 68. (f) Bhowmik, D.; Fiorillo, G.; Lombardi, P.; Kumar, G.S. J.Mol. Recognit. 2015; 28: 722. (g) Chatterjee, S.; Mallick, S.; Das Saha, K.; Buzzetti, F.; Fiorillo, G.; Syeda Monir, T.; Lombardi, P.; Kumar, G.S. RSC Advances 2015, 5, 90632. (h) Ferraroni,M.; Bazzicalupi,C.; Papi, F.; Fiorillo, G.; Guaman-Ortiz, L.M.; References Nocentini, A.; Scovassi, A.I.; Lombardi, P.; Gratteri, P. Chem. Asian J. 2016, 11, 1107 6) (a) Albring, K.F.; Weidemueller, J.; Mittag S.; Weiske, J.; Friedrich, K.; Geroni, C.; Lombardi, P.; Huber, O. BioFactors, 1) Antimicrobial resistance: global report on surveillance 2014, WHO, April 2014 2013, 39, 652. (b) Pierpaoli, E.; Arcamone, A.G.; Buzzetti, F.; Lombardi, P.; Salvatore, C.; Provinciali, M. BioFactors, 2013, 2) Jarosław Sączewski, Krzysztof Hinc, Michał Obuchowski Maria Gdaniec; The Tandem Mannich-Electrophilic Amination 39, 672. (c) Guaman-Ortiz, L.M.; Tillhon, M.; Parks, M.; Dutto, I.; Prosperi, E.; Savio, M.; Arcamone, A.; Buzzetti, F.; Reaction: a Versatile Platform for Fluorescent Probing and Labeling; Chem. Eur. J. 2013, 19, 11531-11535 Lombardi, P.; Scovassi, A.I. BioMed Res Int, 2014, Article ID 924585. (d) Guaman-Ortiz, L.M.; Croce, A.; Aredia, F.; Fiorillo, G.; Syeda Monir, T.; Buzzetti, F.; Lombardi, P.; Scovassi, A.I.. Acta Biochim Biophys Sinica. 2015, 47, 824. (e) Pierpaoli, E.; Damiani, E.; Orlando, F.; Lucarini, G.; Bartozzi, B.; Lombardi, P.; Salvatore, C.; Geroni, C.; Donati, A.; Provinciali, M. Carcinogenesis, 2015, 10, 1169 77 P019 P020 SYNTHESIS AND PHARMACOLOGICAL EVALUATION OF NOVEL LIGAND-BASED DRUG DESIGN STRATEGY IN SEARCHING FOR AMINO BIOISOSTERES FOR THE GABAa RECEPTOR NOVEL ALDOSE REDUCTASE INHIBITORS IN DATABASES OF INDOLE-1-ACETIC ACIDS Alessandro Giraudo (1,2), Anders A. Jensen (1), Birgitte Nielsen (1), Donatella Boschi (2), Jacob Krall (1), Rossella De Blasio (2), Marco L. Lolli (2), Bente Frølund (1) Jana Ballekova, Marta Soltesova Prnova, Milan Stefek, Magdalena Majekova 1) Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Institute of Experimental Pharmacology and Toxicology, Slovak Academy of Sciences, Dubravska cesta 9, 841 04 Bratislava, Universitetsparken 2, DK-2100 Copenhagen, Denmark. Slovakia 2) Department of Science and Drug Technology, University of Torino (UniTO), via Pietro Giuria 9, 10125 Torino, Italy. E-mail: [email protected]

Indole-1-acetic acid moiety is known as a promising starting fragment for design of efficient aldose reductase Several neurotransmitters are known in the mammalian central nervous system (CNS) where γ-aminobutyric inhibitors (ARIs)1. Ligand-based strategy was used in searching for novel ARIs in databases of purchasable acid (GABA) exerts the main inhibitory function. GABA is known to activate ionotropic GABAA receptors compounds. Compounds of a general formula shown in Fig. 1 were selected for an experimental assessment. (GABAARs) and metabotropic GABAB receptors. GABAARs are member of the Cys-loop receptor superfamily Among the compounds studied, 2-(2-(ethoxycarbonyl)-8-methoxy-3,4-dihydro-1H-pyrido[4,3-b]indol-5(2H)-yl) of ligand-gated ion channels and composed by five subunits assembled around the chloride ion conducting pore. acetic acid (compound 1) was identified as the most promising inhibitor of aldose reductase (ALR2), with IC50 Several subunits have been identified so far (α1-6, β1-3, γ1-3, δ, ε, θ, π, ρ1-3) and they build at least 26 native and in nanomolar region and selectivity factor relative to aldehyde reductase (ALR1) around 750. Docking to the 1 mainly heteromeric GABAAR subtypes. GABAARs composed of ρ1-3 subunits assemble as homo- or active site of ALR2 performed for revealed an interaction network responsible for the high affinity and 1 pseudohomomers and are also known as the GABAC receptors. Different subtypes have different selectivity. In ex vivo experiment, sorbitol accumulation in isolated rat eye lenses was significantly inhibited by pharmacodynamic properties and they are located in disparate brain regions. Consequently, they probably exert in the presence of high glucose, starting at a concentration as low as 0.1 μM. This finding indicates the ready heterogeneous functions throughout the central nervous system, which have to be revealed in details.1 uptake of 1 by the eye lens tissue followed by inhibition of the cytosolic ALR2. In streptozotocin-induced diabetic rats, compound 1 administered intragastrically (i.g., 50 mg/kg/day) for five consecutive days Taking this knowledge, it would be important to disclose the function of each GABAAR subtype not only in significantly inhibited sorbitol accumulation in red blood cells and the sciatic nerve. This result points to a ready physiological condition but also in certain pathologies, and investigate the possibility of using orthosteric ligands uptake of 1 after its i.g. administration into the central compartment, its supply to the peripheral nerves and as therapeutic agents. However, there is still a lack of GABAAR subtype selective compounds targeting the inhibition of aldose reductase-mediated sorbitol accumulation. Molecular obesity indices, ADMET parameters orthosteric binding site. To meet the need, a large number of analogues in the GABAAR setting has been predicted along with water solubility point to an excellent „lead-likeness“ of compound 1, with prospects of synthesized aiming for subtype selectivity. A bioisosteric approach has been used extensively and most efforts further structure optimizations. have been directed to the replacement of the acidic moiety of GABA.2 On the other hand, few examples of bioisosteric replacement of the amino moiety of GABA are reported, which include imidazole-4-acetic acid Figure 1 3 (IAA, depicted in ) described as a GABAAR partial agonist. In the present study, a bioisosteric replacement of the amino moiety of IAA using a variety of five membered non-aromatic heterocycles is presented. Dihydroimidazole and 2-amino analogues of dihydrothiazole, dihydrooxazole, and dihydroimidazole (Figure 1) were chosen and shown to translate into valid novel amino bioisosteres in the GABAAR area. The synthesis, the pharmacological and physicochemical properties of these novel heterocycles are reported and discussed.

Fig. 1 Figure 1. Left: structure of lead compound imidazole-4-acetic acid (IAA). Right: structures of novel amino bioisosteres in the GABAA-R setting. References References 1. Jusková M., Májeková M., Demopoulos V.J., Štefek M.: Substituted derivatives of indole acetic acid as aldose reductase inhibitors with antioxidant activity: structure-activity relationship. General Physiology and Biophysics 30 (4), 2011, 342-349 1) Olsen, R. W.; Sieghart, W. International union of pharmacology. LXX. Subtypes of γ-aminobutyric acidA receptors: classification on the basis of subunit composition, pharmacology, and function. Update. Pharmacol. Rev. 2008, 60, 243-260. 2) Petersen, J. G.; Bergmann, R.; Krogsgaard-Larsen, P.; Balle, T.; Frolund, B. Probing the Orthosteric Binding Site of GABAA Receptors with Heterocyclic GABA Carboxylic Acid Bioisosteres. Neurochem. Res. 2014, 39, 1005-1015. 3) Madsen, C.; Jensen, A. A.; Liljefors, T.; Kristiansen, U.; Nielsen, B.; Hansen, C. P.; Larsen, M.; Ebert, B.; Bang-Andersen, B.; Krogsgaard-Larsen, P.; Frolund, B. 5-Substituted Imidazole-4-acetic Acid Analogues: Synthesis, Modeling, and Pharmacological Characterization of a Series of Novel γ-Aminobutyric AcidC Receptor Agonists. J. Med. Chem. 2007, 50, 4147-4161.

78 P019 P020 SYNTHESIS AND PHARMACOLOGICAL EVALUATION OF NOVEL LIGAND-BASED DRUG DESIGN STRATEGY IN SEARCHING FOR AMINO BIOISOSTERES FOR THE GABAa RECEPTOR NOVEL ALDOSE REDUCTASE INHIBITORS IN DATABASES OF INDOLE-1-ACETIC ACIDS Alessandro Giraudo (1,2), Anders A. Jensen (1), Birgitte Nielsen (1), Donatella Boschi (2), Jacob Krall (1), Rossella De Blasio (2), Marco L. Lolli (2), Bente Frølund (1) Jana Ballekova, Marta Soltesova Prnova, Milan Stefek, Magdalena Majekova 1) Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Institute of Experimental Pharmacology and Toxicology, Slovak Academy of Sciences, Dubravska cesta 9, 841 04 Bratislava, Universitetsparken 2, DK-2100 Copenhagen, Denmark. Slovakia 2) Department of Science and Drug Technology, University of Torino (UniTO), via Pietro Giuria 9, 10125 Torino, Italy. E-mail: [email protected]

Indole-1-acetic acid moiety is known as a promising starting fragment for design of efficient aldose reductase Several neurotransmitters are known in the mammalian central nervous system (CNS) where γ-aminobutyric inhibitors (ARIs)1. Ligand-based strategy was used in searching for novel ARIs in databases of purchasable acid (GABA) exerts the main inhibitory function. GABA is known to activate ionotropic GABAA receptors compounds. Compounds of a general formula shown in Fig. 1 were selected for an experimental assessment. (GABAARs) and metabotropic GABAB receptors. GABAARs are member of the Cys-loop receptor superfamily Among the compounds studied, 2-(2-(ethoxycarbonyl)-8-methoxy-3,4-dihydro-1H-pyrido[4,3-b]indol-5(2H)-yl) of ligand-gated ion channels and composed by five subunits assembled around the chloride ion conducting pore. acetic acid (compound 1) was identified as the most promising inhibitor of aldose reductase (ALR2), with IC50 Several subunits have been identified so far (α1-6, β1-3, γ1-3, δ, ε, θ, π, ρ1-3) and they build at least 26 native and in nanomolar region and selectivity factor relative to aldehyde reductase (ALR1) around 750. Docking to the 1 mainly heteromeric GABAAR subtypes. GABAARs composed of ρ1-3 subunits assemble as homo- or active site of ALR2 performed for revealed an interaction network responsible for the high affinity and 1 pseudohomomers and are also known as the GABAC receptors. Different subtypes have different selectivity. In ex vivo experiment, sorbitol accumulation in isolated rat eye lenses was significantly inhibited by pharmacodynamic properties and they are located in disparate brain regions. Consequently, they probably exert in the presence of high glucose, starting at a concentration as low as 0.1 μM. This finding indicates the ready heterogeneous functions throughout the central nervous system, which have to be revealed in details.1 uptake of 1 by the eye lens tissue followed by inhibition of the cytosolic ALR2. In streptozotocin-induced diabetic rats, compound 1 administered intragastrically (i.g., 50 mg/kg/day) for five consecutive days Taking this knowledge, it would be important to disclose the function of each GABAAR subtype not only in significantly inhibited sorbitol accumulation in red blood cells and the sciatic nerve. This result points to a ready physiological condition but also in certain pathologies, and investigate the possibility of using orthosteric ligands uptake of 1 after its i.g. administration into the central compartment, its supply to the peripheral nerves and as therapeutic agents. However, there is still a lack of GABAAR subtype selective compounds targeting the inhibition of aldose reductase-mediated sorbitol accumulation. Molecular obesity indices, ADMET parameters orthosteric binding site. To meet the need, a large number of analogues in the GABAAR setting has been predicted along with water solubility point to an excellent „lead-likeness“ of compound 1, with prospects of synthesized aiming for subtype selectivity. A bioisosteric approach has been used extensively and most efforts further structure optimizations. have been directed to the replacement of the acidic moiety of GABA.2 On the other hand, few examples of bioisosteric replacement of the amino moiety of GABA are reported, which include imidazole-4-acetic acid Figure 1 3 (IAA, depicted in ) described as a GABAAR partial agonist. In the present study, a bioisosteric replacement of the amino moiety of IAA using a variety of five membered non-aromatic heterocycles is presented. Dihydroimidazole and 2-amino analogues of dihydrothiazole, dihydrooxazole, and dihydroimidazole (Figure 1) were chosen and shown to translate into valid novel amino bioisosteres in the GABAAR area. The synthesis, the pharmacological and physicochemical properties of these novel heterocycles are reported and discussed.

Fig. 1 Figure 1. Left: structure of lead compound imidazole-4-acetic acid (IAA). Right: structures of novel amino bioisosteres in the GABAA-R setting. References References 1. Jusková M., Májeková M., Demopoulos V.J., Štefek M.: Substituted derivatives of indole acetic acid as aldose reductase inhibitors with antioxidant activity: structure-activity relationship. General Physiology and Biophysics 30 (4), 2011, 342-349 1) Olsen, R. W.; Sieghart, W. International union of pharmacology. LXX. Subtypes of γ-aminobutyric acidA receptors: classification on the basis of subunit composition, pharmacology, and function. Update. Pharmacol. Rev. 2008, 60, 243-260. 2) Petersen, J. G.; Bergmann, R.; Krogsgaard-Larsen, P.; Balle, T.; Frolund, B. Probing the Orthosteric Binding Site of GABAA Receptors with Heterocyclic GABA Carboxylic Acid Bioisosteres. Neurochem. Res. 2014, 39, 1005-1015. 3) Madsen, C.; Jensen, A. A.; Liljefors, T.; Kristiansen, U.; Nielsen, B.; Hansen, C. P.; Larsen, M.; Ebert, B.; Bang-Andersen, B.; Krogsgaard-Larsen, P.; Frolund, B. 5-Substituted Imidazole-4-acetic Acid Analogues: Synthesis, Modeling, and Pharmacological Characterization of a Series of Novel γ-Aminobutyric AcidC Receptor Agonists. J. Med. Chem. 2007, 50, 4147-4161.

79 P021 P022 SYNTHESIS AND PHARMACOLOGICAL EVALUATION OF NOVEL BRP-7 DERIVATIVES TARGETING FLAP: POTENT AMIDINE CONTAINING GABAA RECEPTOR AGONISTS INHIBITORS OF LEUKOTRIENE BIOSYNTHESIS

Signe Grinberga, Maria Damgaard, Vibe Andersen, Anders A. Jensen, Povl Krogsgaard-Larsen, Birgitte Zehra Tuğçe Gür (1), Burcu Çalışkan (1), Jana Gerstmeier (2), Ulrike Garscha (2), Abdurrahman Olğaç Nielsen, Helle S. Waagepetersen, Arne Schousboe, Bente Frølund (1), Oliver Werz (2), Erden Banoğlu (1)

Department of Drug Design and Pharmacology, Faculty of Health and Medicinal Sciences, University of Copenhagen, 1) Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Gazi University, Etiler, 06330 Yenimahalle, Ankara, Universitetsparken 2, DK-2100, Copenhagen, Denmark Turkey 2) Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena, Philosophenweg 14, D-07743 Jena, Germany

Synaptic inhibition in the brain is largely a result of γ-aminobutyric acid (GABA) signaling, where the fast inhibitory actions of GABA are mediated by activation of GABAA receptors (GABAARs). The GABAARs belong to the Cys-loop receptor family of ligand-gated ion channels and play essential roles in numerous Leukotrienes (LT) are important lipid mediators, which play important roles in the pathophysiology of chronic 2 physiological and pathological processes. Consequently, the GABAARs mediate the effect of a large number of inflammatory diseases. LT biosynthesis starts by the action of phospholipase A which helps the release of AA clinical administered drugs and are putative drug targets in a wide range of neurodegenerative and psychiatric (arachidonic acid) from phospholipid membranes. Then, with the aid of 5-lipoxygenase (5-LO) activating protein disorders, for example depression, schizophrenia, autism, anxiety and panic disorders. (FLAP), AA is transferred to 5-LO for efficient biosynthesis of LTs. Anti-LT molecules are proved to be efficient in treatment of inflammatory pathologies such as respiratory diseases, arthritis, atherosclerosis and even Besides the potential as drug candidates, the development of compounds capable of activating the GABAARs some cancer types like prostate cancer [1]. BRP-7 (IC50=0,31 µM) was previously discovered as a FLAP has contributed to the knowledge of the function and localization of the GABAARs as well as the architecture of inhibitor by our group bearing benzimidazole (BI) nucleus [2]. To identify the pharmacophore groups which are the orthosteric binding site. Conformational restrictions and bioisosteric replacement in the molecule of GABA responsible for FLAP inhibitor activity, and to improve drug like properties of BRP-7, structure activity have afforded a range of agonists with different pharmacological profiles. Classical agonists include muscimol, a relationship studies were performed at C(5)-BI core. Eventually, oxadiazolthione substituted BRP-7 at C(5) of naturally occurring compound in the mushroom Amanita muscaria, and thiomuscimol, the synthetic sulphur BI ring resulted in six-times more potent analogue (IC50=0,05 µM). In contrast to highly lipophilic nature of analogue of muscimol. BRP-7, recently published FLAP inhibitors such as AZD6642, BI665915 are remarkably less lipophilic and bear Bioisosteric replacement of the carboxylic group in GABA has been widely explored and include the heteroaryl amine substituents in their structures as common pharmacophores [3, 4] Thus, to improve drug like 3-hydroxyisoxazole ring in muscimol, the 3-hydroxyisothiazole ring of thiomuscimol, and the properties of BRP-7, it was substituted with 2-aminoheteroaryls as polar arms at C(2) of BI instead of 1-hydroxypyrazole ring of 4-AHP1. In contrast, the amino group in GABA has received much less attention isobutylphenyl, an ibuprofen fingerprint of BRP-7. Eventually, we show the newly introduced substituents despite the fact that transamination has been reported as a limitation for in-vivo studies of e.g. muscimol2. improved drug like properties of BRP-7 while maintaining potent FLAP inhibitor activity. (This study was However, we have recently reported on a series of 2-aminotetrahydropyridine analogues of GABA and identified supported by TUBITAK Research Grant 112S596). the amidine moiety as a valid bioisostere for the amino group3. Inspired by the above mentioned results we have explored amidine moity as an amino group bioisostere for the References GABAAR agonists: 4-AHP, thiomuscimol, muscimol, and the low efficacy partial agonist 4-PHP in terms of 1) Pettersen, D., et al., Recent advances for FLAP inhibitors. Bioorganic and Medicinal Chemistry Letters, 2015. 25(13): p. pharmacological profile and metabolic stability (compounds 1-4). We here report on the synthesis and 2607-12. 2) Banoglu, E., et al., Identification of novel benzimidazole derivatives as inhibitors of leukotriene biosynthesis by virtual pharmacological characterization at the GABAAR of a series of amidine GABA analogues as novel GABAAR screening targeting 5-lipoxygenase-activating protein (FLAP). Bioorganic Medicinal Chemistry, 2012. 20(12): p. 3728-41. agonists. 3) Lemurell, M., et al., Discovery of AZD6642, an inhibitor of 5-lipoxygenase activating protein (FLAP) for the treatment of inflammatory diseases. J Med Chem, 2015. 58(2): p. 897-911. 4) Takahashi, H., et al., Synthesis, SAR, and series evolution of novel oxadiazole-containing 5-lipoxygenase activating protein inhibitors: discovery of 2-[4-(3-{(r)-1-[4-(2-amino-pyrimidin-5-yl)-phenyl]-1-cyclopropyl-ethyl}-[1,2,4]ox adiazol-5-yl)-pyrazol-1-yl]-N,N-dimethyl-acetamide (BI 665915). Journal of Medicinal Chemistry, 2015. 58(4): p. 1669-90.

The binding affinities of the target compounds at native GABAARs were measured by displacement of [3 H]muscimol in rat membrane preparations. Functional characterization was carried out at the human α1β2γ2S and ρ1 GABAARs using the FLIPRTM Membrane Potential Blue Assay. The compounds were shown to be moderate to highly potent GABAAR agonists, some with low-nanomolar affinity and equipotency as agonist to the amino-containing analogues. Furthermore, the most potent compounds were examined as substrates for the GABA-metabolizing enzyme GABA transaminase.

References 1) Petersen, J. G.; Bergamnn, R.; Krogsgaard-Larsen, P.; Balle, T.; Frølund, B.. Neurochem. Res., 2014, 39, 1005-1015 2) Maggi, A.; Enna, A.. Neuropharmacology, 1979, 18, 361-166 3) Petersen, J. G. et al. E. J. Med. Chem., 2014, 84, 404-416 80 P021 P022 SYNTHESIS AND PHARMACOLOGICAL EVALUATION OF NOVEL BRP-7 DERIVATIVES TARGETING FLAP: POTENT AMIDINE CONTAINING GABAA RECEPTOR AGONISTS INHIBITORS OF LEUKOTRIENE BIOSYNTHESIS

Signe Grinberga, Maria Damgaard, Vibe Andersen, Anders A. Jensen, Povl Krogsgaard-Larsen, Birgitte Zehra Tuğçe Gür (1), Burcu Çalışkan (1), Jana Gerstmeier (2), Ulrike Garscha (2), Abdurrahman Olğaç Nielsen, Helle S. Waagepetersen, Arne Schousboe, Bente Frølund (1), Oliver Werz (2), Erden Banoğlu (1)

Department of Drug Design and Pharmacology, Faculty of Health and Medicinal Sciences, University of Copenhagen, 1) Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Gazi University, Etiler, 06330 Yenimahalle, Ankara, Universitetsparken 2, DK-2100, Copenhagen, Denmark Turkey 2) Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena, Philosophenweg 14, D-07743 Jena, Germany

Synaptic inhibition in the brain is largely a result of γ-aminobutyric acid (GABA) signaling, where the fast inhibitory actions of GABA are mediated by activation of GABAA receptors (GABAARs). The GABAARs belong to the Cys-loop receptor family of ligand-gated ion channels and play essential roles in numerous Leukotrienes (LT) are important lipid mediators, which play important roles in the pathophysiology of chronic 2 physiological and pathological processes. Consequently, the GABAARs mediate the effect of a large number of inflammatory diseases. LT biosynthesis starts by the action of phospholipase A which helps the release of AA clinical administered drugs and are putative drug targets in a wide range of neurodegenerative and psychiatric (arachidonic acid) from phospholipid membranes. Then, with the aid of 5-lipoxygenase (5-LO) activating protein disorders, for example depression, schizophrenia, autism, anxiety and panic disorders. (FLAP), AA is transferred to 5-LO for efficient biosynthesis of LTs. Anti-LT molecules are proved to be efficient in treatment of inflammatory pathologies such as respiratory diseases, arthritis, atherosclerosis and even Besides the potential as drug candidates, the development of compounds capable of activating the GABAARs some cancer types like prostate cancer [1]. BRP-7 (IC50=0,31 µM) was previously discovered as a FLAP has contributed to the knowledge of the function and localization of the GABAARs as well as the architecture of inhibitor by our group bearing benzimidazole (BI) nucleus [2]. To identify the pharmacophore groups which are the orthosteric binding site. Conformational restrictions and bioisosteric replacement in the molecule of GABA responsible for FLAP inhibitor activity, and to improve drug like properties of BRP-7, structure activity have afforded a range of agonists with different pharmacological profiles. Classical agonists include muscimol, a relationship studies were performed at C(5)-BI core. Eventually, oxadiazolthione substituted BRP-7 at C(5) of naturally occurring compound in the mushroom Amanita muscaria, and thiomuscimol, the synthetic sulphur BI ring resulted in six-times more potent analogue (IC50=0,05 µM). In contrast to highly lipophilic nature of analogue of muscimol. BRP-7, recently published FLAP inhibitors such as AZD6642, BI665915 are remarkably less lipophilic and bear Bioisosteric replacement of the carboxylic group in GABA has been widely explored and include the heteroaryl amine substituents in their structures as common pharmacophores [3, 4] Thus, to improve drug like 3-hydroxyisoxazole ring in muscimol, the 3-hydroxyisothiazole ring of thiomuscimol, and the properties of BRP-7, it was substituted with 2-aminoheteroaryls as polar arms at C(2) of BI instead of 1-hydroxypyrazole ring of 4-AHP1. In contrast, the amino group in GABA has received much less attention isobutylphenyl, an ibuprofen fingerprint of BRP-7. Eventually, we show the newly introduced substituents despite the fact that transamination has been reported as a limitation for in-vivo studies of e.g. muscimol2. improved drug like properties of BRP-7 while maintaining potent FLAP inhibitor activity. (This study was However, we have recently reported on a series of 2-aminotetrahydropyridine analogues of GABA and identified supported by TUBITAK Research Grant 112S596). the amidine moiety as a valid bioisostere for the amino group3. Inspired by the above mentioned results we have explored amidine moity as an amino group bioisostere for the References GABAAR agonists: 4-AHP, thiomuscimol, muscimol, and the low efficacy partial agonist 4-PHP in terms of 1) Pettersen, D., et al., Recent advances for FLAP inhibitors. Bioorganic and Medicinal Chemistry Letters, 2015. 25(13): p. pharmacological profile and metabolic stability (compounds 1-4). We here report on the synthesis and 2607-12. 2) Banoglu, E., et al., Identification of novel benzimidazole derivatives as inhibitors of leukotriene biosynthesis by virtual pharmacological characterization at the GABAAR of a series of amidine GABA analogues as novel GABAAR screening targeting 5-lipoxygenase-activating protein (FLAP). Bioorganic Medicinal Chemistry, 2012. 20(12): p. 3728-41. agonists. 3) Lemurell, M., et al., Discovery of AZD6642, an inhibitor of 5-lipoxygenase activating protein (FLAP) for the treatment of inflammatory diseases. J Med Chem, 2015. 58(2): p. 897-911. 4) Takahashi, H., et al., Synthesis, SAR, and series evolution of novel oxadiazole-containing 5-lipoxygenase activating protein inhibitors: discovery of 2-[4-(3-{(r)-1-[4-(2-amino-pyrimidin-5-yl)-phenyl]-1-cyclopropyl-ethyl}-[1,2,4]ox adiazol-5-yl)-pyrazol-1-yl]-N,N-dimethyl-acetamide (BI 665915). Journal of Medicinal Chemistry, 2015. 58(4): p. 1669-90.

The binding affinities of the target compounds at native GABAARs were measured by displacement of [3 H]muscimol in rat membrane preparations. Functional characterization was carried out at the human α1β2γ2S and ρ1 GABAARs using the FLIPRTM Membrane Potential Blue Assay. The compounds were shown to be moderate to highly potent GABAAR agonists, some with low-nanomolar affinity and equipotency as agonist to the amino-containing analogues. Furthermore, the most potent compounds were examined as substrates for the GABA-metabolizing enzyme GABA transaminase.

References 1) Petersen, J. G.; Bergamnn, R.; Krogsgaard-Larsen, P.; Balle, T.; Frølund, B.. Neurochem. Res., 2014, 39, 1005-1015 2) Maggi, A.; Enna, A.. Neuropharmacology, 1979, 18, 361-166 3) Petersen, J. G. et al. E. J. Med. Chem., 2014, 84, 404-416 81 P023 P024 BACTERIAL GROWTH INHIBITION BY TARGETING DAHP INNOVATIVE MEDICINES INITIATIVE CHEM21: SYNTHASE WITH A RATIONALLY DESIGNED OXIME-BASED IMPLEMENTATION OF KEY TECHNOLOGIES AT ORION PHARMA INHIBITOR MEDICINAL CHEMISTRY

Maren Heimhalt, Paul Berti Ari Hietanen[1], Esa Kumpulainen[2], Anniina Vesalainen[1], Josef Messinger[2]

Department of Chemistry and Chemical Biology, McMaster University, 1280 Main St. W., Hamilton, ON L8S 4L8, Canada 1) Medicinal Chemistry, Orion Corporation Tengströminkatu 8, 20101 Turku, Finland 2) Medicinal Chemistry, Orion Corporation Orionintie 1, 02200 Espoo, Finland

Expanded bacterial resistance towards conventional antibiotics is creating an urgent need for new strategies against bacteria. DAHP (3-deoxy-D-arabinoheptulosonate-7-phosphate) oxime is a transition state inhibitor for DAHP synthase (DAHPS). To better understand inhibition, the pH profiles of catalysis and inhibition were Chemical Manufacturing Methods for the 21st Century Pharmaceutical Industries (CHEM21) is a multipartner investigated. While the enzyme’s kcat/KM value decreased at high pH, inhibition (1/Ki) by DAHP oxime project under Innovative Medicines Initiative consisting of collaborators from academia and pharmaceutical increased. Ionization of enzymatic residues could not be identified as responsible for stronger inhibition, so the industry across Europe. The objective of the project is to develop and implement sustainable technologies for ionization of the inhibitor itself was investigated. The Ki value of the derivative DAHP O-methyloxime showed green pharmaceutical synthesis and manufacture. The concortium has divided key technologies and activities in little pH dependence. Therefore, we hypothesized that the anionic DAHP oximate (C=N−O−) inhibits DAHPS work packages (WPs), such as WP2 focusing on chemical catalysis and continuous flow processing; WP3 and more strongly than the neutral form, and that a molecule with a lower oxime pKa would be a tighter inhibitor. WP4 focusing on biocatalysis and synthetic biology, respectively; and WP5 focusing on green chemistry The analysis of the pKa and Ki values of pyruvate oxime, 3-fluoropyruvate oxime, and 3,3,3-trifluoropyruvate training, metrics development and results dissemination. oxime confirmed that fluorine bonded to the oxime α-carbon lowers the inhibitor’s pKa and improves Ki. An enzymatic synthesis of 3-fluoro-DAHP oxime is being developed. Furthermore, 3,3,3-trifluoropyruvate oxime Despite green chemistry activities often being targeted for process scale operations, there is an increasing ethyl ester is the first oxime-based inhibitor to reduce E. coli growth in culture (IC50 = 0.2 mg/mL), and its awareness on the importance of applying sustainable technologies already in medicinal chemistry phase of drug effects were mitigated by DAHPS overexpression. This is evidence that this oxime-based inhibitor targets discovery projects. Medicinal chemistry at Orion is committed to environmental sustainability as this is an DAHPS. important component in Orion corporate strategy. The poster presents strategies with examples how some of the key technologies of CHEM21 have been and will be implemented at Orion medicinal chemistry. These include applications in C–H activation chemistry, flow synthesis and biocatalytic synthesis as well as training of research personnel in green chemistry and adaptation of best sustainability practices in Orion medicinal chemistry laboratories. The research for this work has received funding from the Innovative Medicines Initiative joint undertaking project CHEM21 (https://www.chem21.eu/) grant agreement n°115360, resources of which are composed of financial contribution from the European Union’s Seventh Framework Programme (FP7/2007-2013) and EFPIA companies in kind contribution.

82 P023 P024 BACTERIAL GROWTH INHIBITION BY TARGETING DAHP INNOVATIVE MEDICINES INITIATIVE CHEM21: SYNTHASE WITH A RATIONALLY DESIGNED OXIME-BASED IMPLEMENTATION OF KEY TECHNOLOGIES AT ORION PHARMA INHIBITOR MEDICINAL CHEMISTRY

Maren Heimhalt, Paul Berti Ari Hietanen[1], Esa Kumpulainen[2], Anniina Vesalainen[1], Josef Messinger[2]

Department of Chemistry and Chemical Biology, McMaster University, 1280 Main St. W., Hamilton, ON L8S 4L8, Canada 1) Medicinal Chemistry, Orion Corporation Tengströminkatu 8, 20101 Turku, Finland 2) Medicinal Chemistry, Orion Corporation Orionintie 1, 02200 Espoo, Finland

Expanded bacterial resistance towards conventional antibiotics is creating an urgent need for new strategies against bacteria. DAHP (3-deoxy-D-arabinoheptulosonate-7-phosphate) oxime is a transition state inhibitor for DAHP synthase (DAHPS). To better understand inhibition, the pH profiles of catalysis and inhibition were Chemical Manufacturing Methods for the 21st Century Pharmaceutical Industries (CHEM21) is a multipartner investigated. While the enzyme’s kcat/KM value decreased at high pH, inhibition (1/Ki) by DAHP oxime project under Innovative Medicines Initiative consisting of collaborators from academia and pharmaceutical increased. Ionization of enzymatic residues could not be identified as responsible for stronger inhibition, so the industry across Europe. The objective of the project is to develop and implement sustainable technologies for ionization of the inhibitor itself was investigated. The Ki value of the derivative DAHP O-methyloxime showed green pharmaceutical synthesis and manufacture. The concortium has divided key technologies and activities in little pH dependence. Therefore, we hypothesized that the anionic DAHP oximate (C=N−O−) inhibits DAHPS work packages (WPs), such as WP2 focusing on chemical catalysis and continuous flow processing; WP3 and more strongly than the neutral form, and that a molecule with a lower oxime pKa would be a tighter inhibitor. WP4 focusing on biocatalysis and synthetic biology, respectively; and WP5 focusing on green chemistry The analysis of the pKa and Ki values of pyruvate oxime, 3-fluoropyruvate oxime, and 3,3,3-trifluoropyruvate training, metrics development and results dissemination. oxime confirmed that fluorine bonded to the oxime α-carbon lowers the inhibitor’s pKa and improves Ki. An enzymatic synthesis of 3-fluoro-DAHP oxime is being developed. Furthermore, 3,3,3-trifluoropyruvate oxime Despite green chemistry activities often being targeted for process scale operations, there is an increasing ethyl ester is the first oxime-based inhibitor to reduce E. coli growth in culture (IC50 = 0.2 mg/mL), and its awareness on the importance of applying sustainable technologies already in medicinal chemistry phase of drug effects were mitigated by DAHPS overexpression. This is evidence that this oxime-based inhibitor targets discovery projects. Medicinal chemistry at Orion is committed to environmental sustainability as this is an DAHPS. important component in Orion corporate strategy. The poster presents strategies with examples how some of the key technologies of CHEM21 have been and will be implemented at Orion medicinal chemistry. These include applications in C–H activation chemistry, flow synthesis and biocatalytic synthesis as well as training of research personnel in green chemistry and adaptation of best sustainability practices in Orion medicinal chemistry laboratories. The research for this work has received funding from the Innovative Medicines Initiative joint undertaking project CHEM21 (https://www.chem21.eu/) grant agreement n°115360, resources of which are composed of financial contribution from the European Union’s Seventh Framework Programme (FP7/2007-2013) and EFPIA companies in kind contribution.

83 P025 P027 LEAD OPTIMIZATION OF GABAAa5 RECEPTOR NEGATIVE NEW METAL CHELATING PYRAZOLONE BASED LIGANDS AND ALLOSTERIC MODULATORS THEIR Co/Cu COMPLEXES OF THERAPEUTIC PROPERTIES

Masato Higashino (1), Tetsuji Saito (1), Akira Imagawa (1), Hiromu Habashita (1), Soichi Kawaharada Omoruyi Idemudia, Alexander Sadimenko (1), Miki Nakanishi (1), Angus M Macleod (2), Mark S Chambers (2), Arwel Lewis (2), Alastair Rae (2), Kim L Hirst (2), Grant Wishart (2), Gary S Clark (2), Scott A Maidment (2) Chemistry Department, University of Fort Hare 1 King Williams Town Road, Private Bag x1314, Alice 5700, South Africa 1) Ono Pharmaceutical Co., Ltd., 3-1-1 Sakurai, Shimamoto-cho, Mishima-gun, Osaka, Japan 2) Charles River, Chesterford Research Park, CB10 1XL, United Kingdom

The need for the design and synthesis of new bioactive compounds with novel characteristics that may become alternative replacement to commonly used medicinal drugs due to the problems of disease resistance to the drugs From HTS of 120,000 compounds using both binding and functional assays against the GABAAα5 receptor, we and their toxicity effects is still a huge concern for synthetic as well as medicinal chemists. These compounds identified several chemical series of GABAAα5 receptor negative allosteric modulators (NAMs) as initial hit have further been coordinated with transition metal ions as this may increase their pharmacological activities. compounds. Optimization of the hit compounds led us to identify a potent and selective GABAAα5 receptor Acylpyrazolones are good biological, metal ion chelating and analytical reagents. Antipyrine, a derivative of NAM. This compound significantly enhanced LTP (Long-term potentiation) in rat hippocampus slice and pyrazolone has been employed as a clinical drugs and this work is based on its structural modifications towards a improved cognition in several animal models without anxiogenic or proconvulsant side effects. Currently further possible increase in its therapeutic applications. Via a condensation reaction with amines, acylpyrazolones form optimization is ongoing. a more chelating and superior group of compounds known as azomethines. 4-acyl-3-methyl-1-phenyl-2-pyrazolin-5-ones were reacted with corresponding phenylhydrazine derivatives to get a new phenylhydrazone (azomethine), which were further reacted with aqueous solutions of cobalt and cuopper to afford their metal complexes. The compounds were characterized/identified by analytical, spectroscopic, TGA, as well as x-ray crystallography. The bidentate ON ligand formed stable octahedral geometry with metal ions. Using the disc diffusion technique to screen the synthesized compounds at 20 mg/ml against selected bacterial isolates in triplicates, potential bactericides were identified. Their bioactivity varies, with the metal complexes showing higher antibacterial activity at an MIC value of 0.63 mg/ml for Co(II). Similarly, ligands and complexes also showed antioxidant scavenging properties against 2, 2-diphenyl-1-picrylhydrazyl DPPH radical at 0.5mg/ml relative to Ascorbic acid. Anticancer studies of synthesized compounds are ongoing as possible antitumour leads are proposed

84 P025 P027 LEAD OPTIMIZATION OF GABAAa5 RECEPTOR NEGATIVE NEW METAL CHELATING PYRAZOLONE BASED LIGANDS AND ALLOSTERIC MODULATORS THEIR Co/Cu COMPLEXES OF THERAPEUTIC PROPERTIES

Masato Higashino (1), Tetsuji Saito (1), Akira Imagawa (1), Hiromu Habashita (1), Soichi Kawaharada Omoruyi Idemudia, Alexander Sadimenko (1), Miki Nakanishi (1), Angus M Macleod (2), Mark S Chambers (2), Arwel Lewis (2), Alastair Rae (2), Kim L Hirst (2), Grant Wishart (2), Gary S Clark (2), Scott A Maidment (2) Chemistry Department, University of Fort Hare 1 King Williams Town Road, Private Bag x1314, Alice 5700, South Africa 1) Ono Pharmaceutical Co., Ltd., 3-1-1 Sakurai, Shimamoto-cho, Mishima-gun, Osaka, Japan 2) Charles River, Chesterford Research Park, CB10 1XL, United Kingdom

The need for the design and synthesis of new bioactive compounds with novel characteristics that may become alternative replacement to commonly used medicinal drugs due to the problems of disease resistance to the drugs From HTS of 120,000 compounds using both binding and functional assays against the GABAAα5 receptor, we and their toxicity effects is still a huge concern for synthetic as well as medicinal chemists. These compounds identified several chemical series of GABAAα5 receptor negative allosteric modulators (NAMs) as initial hit have further been coordinated with transition metal ions as this may increase their pharmacological activities. compounds. Optimization of the hit compounds led us to identify a potent and selective GABAAα5 receptor Acylpyrazolones are good biological, metal ion chelating and analytical reagents. Antipyrine, a derivative of NAM. This compound significantly enhanced LTP (Long-term potentiation) in rat hippocampus slice and pyrazolone has been employed as a clinical drugs and this work is based on its structural modifications towards a improved cognition in several animal models without anxiogenic or proconvulsant side effects. Currently further possible increase in its therapeutic applications. Via a condensation reaction with amines, acylpyrazolones form optimization is ongoing. a more chelating and superior group of compounds known as azomethines. 4-acyl-3-methyl-1-phenyl-2-pyrazolin-5-ones were reacted with corresponding phenylhydrazine derivatives to get a new phenylhydrazone (azomethine), which were further reacted with aqueous solutions of cobalt and cuopper to afford their metal complexes. The compounds were characterized/identified by analytical, spectroscopic, TGA, as well as x-ray crystallography. The bidentate ON ligand formed stable octahedral geometry with metal ions. Using the disc diffusion technique to screen the synthesized compounds at 20 mg/ml against selected bacterial isolates in triplicates, potential bactericides were identified. Their bioactivity varies, with the metal complexes showing higher antibacterial activity at an MIC value of 0.63 mg/ml for Co(II). Similarly, ligands and complexes also showed antioxidant scavenging properties against 2, 2-diphenyl-1-picrylhydrazyl DPPH radical at 0.5mg/ml relative to Ascorbic acid. Anticancer studies of synthesized compounds are ongoing as possible antitumour leads are proposed

85 P028 P029 MOLECULAR DOCKING OF NOVEL NON-COMPETITIVE hBGT1 THE MELLEOLIDE DEHYDROARMELLYLORSELLINATE (DAO) INHIBITORS TO EXPLORE THEIR STRUCTURE-ACTIVITY INHIBITS 5-LIPOXYGENASE AND DECREASES EICOSANOID RELATIONSHIP BIOSYNTHESIS IN HUMAN LEUKOCYTES

Stefanie Kickinger (1), Anas Al-Khawaja (2), Margot Ernst (3), Rasmus P. Clausen (2), Petrine Stefanie König (1), Maximilian Dörfer (2), Dirk Hoffmeister (2), Oliver Werz (1) Wellendorph (2), Gerhard F. Ecker (1) 1) Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Germany 1) University of Vienna, Dept. of Pharmaceutical Chemistry, Austria 2) Department of Pharmaceutical Microbiology, Leibniz Institute for Natural Product Research and Infection Biology, 2) University of Copenhagen, Dept. of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, Denmark Friedrich Schiller University Jena, Germany 3) Medical University of Vienna, Center for Brain Research, Dept. of Biochemistry and Molecular Biology, Austria

In mammalian cells, eicosanoids play distinguished roles in the regulation of inflammatory processes and may The human betaine/γ-aminobutyric acid-transporter 1 (hBGT1) facilitates the reuptake of γ-aminobutyric acid increase or attenuate inflammatory responses, with major impact on cell proliferation, cell migration, (GABA) into neuronal and glial cells [1]. The inhibition of hBGT1 increases GABAergic transmission and has phagocytosis and cytokine production. Arachidonic acid (AA), a polyunsaturated fatty acid, is the precursor of therefore emerged as a promising target for the treatment of epilepsy [2,3]. eicosanoids that include leukotrienes (LTs), prostaglandins (PGs), thromboxanes (TXs), lipoxins (LXs) and other hydroxylated AA derivatives. AA is stored in membrane phospholipids and is released by cytosolic In this study, we explored the structure-activity relationship of new analogs of the first known non-competitive phospholipase A2 (cPLA2) upon stimulation. Three different enzymes transform liberated AA to eicosanoids: hBGT1 inhibitor N-(1-benzyl-4-piperidinyl)-2,4-dichlorobenzamide (BPDBA) by computational methods [4]. BPDBA shows selectivity for BGT1 over the other three GATs. The recently released crystal structure of human lipoxygenases (LOX), cyclooxygenases (COX) and cytochromes P450 (CYP 450). Here, we present the serotonin transporter (hSERT) shares high sequence identity (44.7%) and similarity (66.4%) with hBGT1 and melleolide dehydroarmillylorsellinate (DAO) as a potent suppressor of eicosanoid biosynthesis. Melleolides are was used as a template for generating hBGT1 homology models with Modeller [5]. Noteworthy, the structure of structurally unique and bioactive natural products of the basidiomycete genus Armillaria. DAO showes hSERT was co-crystalized with a ligand revealing an allosteric binding site [6]. Therefore, active BPDBA antifungal as well as cytotoxic effects [1], but it may also exhibit anti-inflammatory properties as it effectively analogs were docked into thispostulated corresponding allosteric binding site of hBGT1 between transmembrane inhibits 5-LOX activity. We observed a strong suppression of 5-LOX product formation in A23187 stimulated regions 1, 6, 10, 11. Docking with flexible side chains was performed in the software package GOLD [7] and human neutrophils by DAO (IC50 = 0.3 µM). This inhibitory effect by DAO was irreversible (demonstrated by 100 poses per compound were generated. The docking poses were analyzed using an in-house protocol for wash-out experiments) and supplementation of neutrophils with exogenous AA impaired the potency of DAO common scaffold clustering with an RMSD of less than 3Å [8]. Calculation of (IC50= 2 µM). Furthermore, DAO inhibited 5-LOX activity in a cell-free assay with lower effectiveness (IC50= 3 Protein-Ligand-Interaction-Fingerprints (PLIFs) and visual inspection was carried out with MOE [9]. 1000 poses µM). Using an UPLC-MS/MS-based lipidomics method we determined the effects of DAO on overall eicosanoid of 10 active BPDBA analogs were assembled into 87 clusters. formation in A23187 activated human neutrophils and monocytes. DAO essentially suppressed all monitored. The two most populated clusters, which contained poses of all docked compounds, were able to explain the Again, the inhibitory potency of DAO was impaired when exogenous AA was added to the leukocytes, structure-activity relationship of an experimentally tested BPDBA analog series. Accordingly, the suggesting that DAO may negatively influence other key enzymes along the AA pathway like cPLA2. 2,4-dichlorobenzamide ring of BPDBA fits into a lipophilic pocket that is stabilized by pi-pi interactions with Interestingly, when exogenous AA was provided an increase of PGs and 12-/15-LOX products were evident by Tyr453. Removing the Cl substituents in the series of BPDBA compounds leads to a loss of van der Waals DAO treatment. In summary, the natural product DAO is a direct inhibitor of 5-LOX that in addition exhibits a (vdW) interaction and gradually decreases affinity. Introducing a third Cl substituent in the meta position broad activity spectrum on eicosanoid biosynthesis. increases affinity due to increased vdW interaction. However, introduction of a trifluoromethyl substituent in the ortho-position leads to a slight decrease in affinity due to steric hindrance. In addition, the removal of the benzyl ring of BPDBA leads to a drastic affinity drop due to the loss of vdW interaction. This docking study reveals the References first insight into a new possible molecular binding mode of non-competitive hBGT1 inhibitors that might be 1) Bohnert, Markus, et al. "Melleolides induce rapid cell death in human primary monocytes and cancer cells." Bioorganic & driven by vdW interaction. medicinal chemistry 22.15 (2014): 3856-3861 Acknowledgement We gratefully acknowledge financial support provided by the Austrian Science Fund grant number W1232.

References 1) Kristensen, A. S. et al., Pharmacological Reviews 63, 585–640 (2011). 2) White, H. S, Journal of Pharmacology and Experimental Therapeutics 312, 866–874 (2004). 3) Madsen, K. K. et al, Journal of Pharmacology and Experimental Therapeutics 338, 214–219 (2011). 4) Kragholm, B. et al, Biochemical Pharmacology 86, 521–528 (2013). 5) Webb, B.& Sali, A., Bioinformatics, John Wiley & Sons, Inc., 5.6.1-5.6.32, 2014. 6) Coleman, J. A., Green, E. M. & Gouaux, Nature 532, 334–339 (2016). 7) Hartshorn, M. J. et al, Journal of Medicinal Chemistry 50, 726-741, 2007. 8) Rudolph, U. & Knoflach, F., Nature Reviews Drug Discovery 10, 685–697 (2011). 9) Molecular Operating Environment (MOE), 2013.08; Chemical Computing Group Inc., 1010 Sherbooke St. West, Suite #910, Montreal, QC, Canada, H3A 2R7 (2016).

86 P028 P029 MOLECULAR DOCKING OF NOVEL NON-COMPETITIVE hBGT1 THE MELLEOLIDE DEHYDROARMELLYLORSELLINATE (DAO) INHIBITORS TO EXPLORE THEIR STRUCTURE-ACTIVITY INHIBITS 5-LIPOXYGENASE AND DECREASES EICOSANOID RELATIONSHIP BIOSYNTHESIS IN HUMAN LEUKOCYTES

Stefanie Kickinger (1), Anas Al-Khawaja (2), Margot Ernst (3), Rasmus P. Clausen (2), Petrine Stefanie König (1), Maximilian Dörfer (2), Dirk Hoffmeister (2), Oliver Werz (1) Wellendorph (2), Gerhard F. Ecker (1) 1) Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Germany 1) University of Vienna, Dept. of Pharmaceutical Chemistry, Austria 2) Department of Pharmaceutical Microbiology, Leibniz Institute for Natural Product Research and Infection Biology, 2) University of Copenhagen, Dept. of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, Denmark Friedrich Schiller University Jena, Germany 3) Medical University of Vienna, Center for Brain Research, Dept. of Biochemistry and Molecular Biology, Austria

In mammalian cells, eicosanoids play distinguished roles in the regulation of inflammatory processes and may The human betaine/γ-aminobutyric acid-transporter 1 (hBGT1) facilitates the reuptake of γ-aminobutyric acid increase or attenuate inflammatory responses, with major impact on cell proliferation, cell migration, (GABA) into neuronal and glial cells [1]. The inhibition of hBGT1 increases GABAergic transmission and has phagocytosis and cytokine production. Arachidonic acid (AA), a polyunsaturated fatty acid, is the precursor of therefore emerged as a promising target for the treatment of epilepsy [2,3]. eicosanoids that include leukotrienes (LTs), prostaglandins (PGs), thromboxanes (TXs), lipoxins (LXs) and other hydroxylated AA derivatives. AA is stored in membrane phospholipids and is released by cytosolic In this study, we explored the structure-activity relationship of new analogs of the first known non-competitive phospholipase A2 (cPLA2) upon stimulation. Three different enzymes transform liberated AA to eicosanoids: hBGT1 inhibitor N-(1-benzyl-4-piperidinyl)-2,4-dichlorobenzamide (BPDBA) by computational methods [4]. BPDBA shows selectivity for BGT1 over the other three GATs. The recently released crystal structure of human lipoxygenases (LOX), cyclooxygenases (COX) and cytochromes P450 (CYP 450). Here, we present the serotonin transporter (hSERT) shares high sequence identity (44.7%) and similarity (66.4%) with hBGT1 and melleolide dehydroarmillylorsellinate (DAO) as a potent suppressor of eicosanoid biosynthesis. Melleolides are was used as a template for generating hBGT1 homology models with Modeller [5]. Noteworthy, the structure of structurally unique and bioactive natural products of the basidiomycete genus Armillaria. DAO showes hSERT was co-crystalized with a ligand revealing an allosteric binding site [6]. Therefore, active BPDBA antifungal as well as cytotoxic effects [1], but it may also exhibit anti-inflammatory properties as it effectively analogs were docked into thispostulated corresponding allosteric binding site of hBGT1 between transmembrane inhibits 5-LOX activity. We observed a strong suppression of 5-LOX product formation in A23187 stimulated regions 1, 6, 10, 11. Docking with flexible side chains was performed in the software package GOLD [7] and human neutrophils by DAO (IC50 = 0.3 µM). This inhibitory effect by DAO was irreversible (demonstrated by 100 poses per compound were generated. The docking poses were analyzed using an in-house protocol for wash-out experiments) and supplementation of neutrophils with exogenous AA impaired the potency of DAO common scaffold clustering with an RMSD of less than 3Å [8]. Calculation of (IC50= 2 µM). Furthermore, DAO inhibited 5-LOX activity in a cell-free assay with lower effectiveness (IC50= 3 Protein-Ligand-Interaction-Fingerprints (PLIFs) and visual inspection was carried out with MOE [9]. 1000 poses µM). Using an UPLC-MS/MS-based lipidomics method we determined the effects of DAO on overall eicosanoid of 10 active BPDBA analogs were assembled into 87 clusters. formation in A23187 activated human neutrophils and monocytes. DAO essentially suppressed all monitored. The two most populated clusters, which contained poses of all docked compounds, were able to explain the Again, the inhibitory potency of DAO was impaired when exogenous AA was added to the leukocytes, structure-activity relationship of an experimentally tested BPDBA analog series. Accordingly, the suggesting that DAO may negatively influence other key enzymes along the AA pathway like cPLA2. 2,4-dichlorobenzamide ring of BPDBA fits into a lipophilic pocket that is stabilized by pi-pi interactions with Interestingly, when exogenous AA was provided an increase of PGs and 12-/15-LOX products were evident by Tyr453. Removing the Cl substituents in the series of BPDBA compounds leads to a loss of van der Waals DAO treatment. In summary, the natural product DAO is a direct inhibitor of 5-LOX that in addition exhibits a (vdW) interaction and gradually decreases affinity. Introducing a third Cl substituent in the meta position broad activity spectrum on eicosanoid biosynthesis. increases affinity due to increased vdW interaction. However, introduction of a trifluoromethyl substituent in the ortho-position leads to a slight decrease in affinity due to steric hindrance. In addition, the removal of the benzyl ring of BPDBA leads to a drastic affinity drop due to the loss of vdW interaction. This docking study reveals the References first insight into a new possible molecular binding mode of non-competitive hBGT1 inhibitors that might be 1) Bohnert, Markus, et al. "Melleolides induce rapid cell death in human primary monocytes and cancer cells." Bioorganic & driven by vdW interaction. medicinal chemistry 22.15 (2014): 3856-3861 Acknowledgement We gratefully acknowledge financial support provided by the Austrian Science Fund grant number W1232.

References 1) Kristensen, A. S. et al., Pharmacological Reviews 63, 585–640 (2011). 2) White, H. S, Journal of Pharmacology and Experimental Therapeutics 312, 866–874 (2004). 3) Madsen, K. K. et al, Journal of Pharmacology and Experimental Therapeutics 338, 214–219 (2011). 4) Kragholm, B. et al, Biochemical Pharmacology 86, 521–528 (2013). 5) Webb, B.& Sali, A., Bioinformatics, John Wiley & Sons, Inc., 5.6.1-5.6.32, 2014. 6) Coleman, J. A., Green, E. M. & Gouaux, Nature 532, 334–339 (2016). 7) Hartshorn, M. J. et al, Journal of Medicinal Chemistry 50, 726-741, 2007. 8) Rudolph, U. & Knoflach, F., Nature Reviews Drug Discovery 10, 685–697 (2011). 9) Molecular Operating Environment (MOE), 2013.08; Chemical Computing Group Inc., 1010 Sherbooke St. West, Suite #910, Montreal, QC, Canada, H3A 2R7 (2016).

87 P030 P031 SCREENING OF NATURE BANK AND DIVERSE SCAFFOLD ANTICANCER AND STRUCTURE ACTIVITY RELATIONSHIP OF LIBRARY FROM COMPOUNDS AUSTRALIA AGAINST AMPK AMINOPHENOLS CHOLINE KINASE INHIBITORS

Jan Lanz (1,2), Ronald J. Quinn (2), Trond Ulven (1) LUISA CARLOTA LOPEZ-CARA (1), MARIA KIMATRAI SALVADOR (1), SANTIAGO SCHIAFFINO ORTEGA (1), ANTONIO ENTRENA (1), MIGUEL ANGEL GALLO (1), SERGIO 1) Department of Physics, Chemistry and Pharmacy, Faculty of Science, University of Southern Denmark, Odense, Denmark PORTILLO HARO (1), GIANLUCA RUBBINI (1), MARIA PAZ CARRASCO JIMENEZ (2), PABLO 2) Eskitis Institute for Drug Discovery, Griffith University, Brisbane, Australia RIOS (2), CARMEN MARCO (2), ELENA MARIOTTO (3), ROBERTA BORTOLOZZI (3), GIUSEPPE BASSO (3), GIAMPIETRO VIOLA (3)

1) Departamento de Química Orgánica y Farmacéutica, Facultad de Farmacia, Campus de Cartuja s/n, 18071, Granada, Spain Adenosine 5’-monophosphate-activated protein kinase (AMPK), a sensor of cellular energy status, plays a key 2) Departamento de Bioquímica y Biología Molecular I, Facultad de Ciencias, Campus Fuentenueva, 18071 Granada, Spain role in whole-body energy homeostasis. Therefore, AMPK is an interesting target for the treatment of 3) Dipartimento di Salute della Donna e del Bambino, Laboratorio di Oncoematologia, Università di Padova, 35131 pathological conditions involving abnormal energy regulation like metabolic disorders including type-2-diabetes Padova, Italy and obesity. AMPK is a promising target to fight cancer due to its influence on cell proliferation. Additionally, pharmacological targeting of AMPK could be beneficial for the treatment of Alzheimer´s disease and other neurological diseases.[1] A common theme emerging from metabolic reprogramming of cancer is that when nutrients are abundant, A high-throughput screening method was set up using the luciferase-based Kinase-Glo® assay kit from Promega oncogenic signalling pathways direct enhanced nutrient acquisition and facilitate assimilation of carbon into on an Agilent BioCel™ automation system. The assay conditions were optimized so AMPK activators and macromolecules such as lipids, proteins and nucleic acids to support cell growth and proliferation. Lipids inhibitors can be detected using the same conditions. compose a substantial fraction of the dry weight of mammalian cells, and maintaining a supply of lipids is Selected compound libraries from Nature Bank[2], a unique source of natural products and natural product essential for cell proliferation1. To produce the new phospholipid bilayers that are necessary for cell division, extracts, and Compounds Australia[3] were then screened against one isoform of AMPK in order to discover cancer cell must increase de novo generation of lipids and steroids. Actually, several enzymes involved in these novel AMPK activators and inhibitors for further development in biological and medicinal chemistry projects. pathways such as fatty acid synthase, ATP citrate lyase and choline kinase (ChoK) among others, have been associated with tumor development and progression in vitro and in vivo2. Choline is essential for the synthesis of So far, >700 compounds from Nature Bank and >5´300 compounds from a diverse scaffold library from the major membrane phospholipid phosphatidylcholine and choline kinase activity has been shown to be Compounds Australia were screened. Five scaffolds including >40 molecules from the diverse scaffold library upregulated in human cancers and is requires for tumor-cell proliferation2,3. Herein we report a novel and >20 natural products were identified to modulate the activity of AMPK. The structure-activity relationship family of structurally related to hemicolinium-3 (HC-3, a classical ChoK inhibitor known by its neurotoxicity) of information that is already present in the scaffold hits as well as molecular modeling studies of the hit structures non-symmetrical monocationic compounds with antitumor activity. Compound known as 4f, (compared to will be used for the development of more potent AMPK modulators. other, a priori, better candidates from the same series) shows the best IC50 (0.99±0.17) in the inhibition of Human choline quinasa, HsChoKα1 (the isoform upregulated in cancer) something that can be explained References because docking studies show that two wholes molecules of compound are able to take up both binding sites of 1) Rana, S.; Blowers, E. C.; Natarajan, A.; Small Molecule Adenosine 5'-Monophosphate Activated Protein Kinase (AMPK) the enzyme, simultaneously, one molecule binds the ATP binding site of the enzyme and another one binds the Modulators and Human Diseases. J. Med. Chem. 2015, 58, 2-29. choline binding site, the 3-aminophenol fragment of compound 4f is stabilized by two H-bonds, with Tyr354 2) www.griffith.edu.au/science-aviation/eskitis-institute/nature-bank and Glu434 provided by both hydrogens from –NH- and OH groups. All together would explain the better IC50 3) www.griffith.edu.au/science-aviation/compounds-australia of 4f.

References 1) Lindsey, K. Nature Cell Biology 2015, 17, 351-359. 2) Martinez-Outschoorn , U. et al. Nature Reviews Clinical Oncology 2016. doi:10.1038/nrclinonc.2016.60 3) Schiaffino-Ortega, R.et al. Bioorganic & Medicinal Chemistry 2013, 21, 7146-7154

88 P030 P031 SCREENING OF NATURE BANK AND DIVERSE SCAFFOLD ANTICANCER AND STRUCTURE ACTIVITY RELATIONSHIP OF LIBRARY FROM COMPOUNDS AUSTRALIA AGAINST AMPK AMINOPHENOLS CHOLINE KINASE INHIBITORS

Jan Lanz (1,2), Ronald J. Quinn (2), Trond Ulven (1) LUISA CARLOTA LOPEZ-CARA (1), MARIA KIMATRAI SALVADOR (1), SANTIAGO SCHIAFFINO ORTEGA (1), ANTONIO ENTRENA (1), MIGUEL ANGEL GALLO (1), SERGIO 1) Department of Physics, Chemistry and Pharmacy, Faculty of Science, University of Southern Denmark, Odense, Denmark PORTILLO HARO (1), GIANLUCA RUBBINI (1), MARIA PAZ CARRASCO JIMENEZ (2), PABLO 2) Eskitis Institute for Drug Discovery, Griffith University, Brisbane, Australia RIOS (2), CARMEN MARCO (2), ELENA MARIOTTO (3), ROBERTA BORTOLOZZI (3), GIUSEPPE BASSO (3), GIAMPIETRO VIOLA (3)

1) Departamento de Química Orgánica y Farmacéutica, Facultad de Farmacia, Campus de Cartuja s/n, 18071, Granada, Spain Adenosine 5’-monophosphate-activated protein kinase (AMPK), a sensor of cellular energy status, plays a key 2) Departamento de Bioquímica y Biología Molecular I, Facultad de Ciencias, Campus Fuentenueva, 18071 Granada, Spain role in whole-body energy homeostasis. Therefore, AMPK is an interesting target for the treatment of 3) Dipartimento di Salute della Donna e del Bambino, Laboratorio di Oncoematologia, Università di Padova, 35131 pathological conditions involving abnormal energy regulation like metabolic disorders including type-2-diabetes Padova, Italy and obesity. AMPK is a promising target to fight cancer due to its influence on cell proliferation. Additionally, pharmacological targeting of AMPK could be beneficial for the treatment of Alzheimer´s disease and other neurological diseases.[1] A common theme emerging from metabolic reprogramming of cancer is that when nutrients are abundant, A high-throughput screening method was set up using the luciferase-based Kinase-Glo® assay kit from Promega oncogenic signalling pathways direct enhanced nutrient acquisition and facilitate assimilation of carbon into on an Agilent BioCel™ automation system. The assay conditions were optimized so AMPK activators and macromolecules such as lipids, proteins and nucleic acids to support cell growth and proliferation. Lipids inhibitors can be detected using the same conditions. compose a substantial fraction of the dry weight of mammalian cells, and maintaining a supply of lipids is Selected compound libraries from Nature Bank[2], a unique source of natural products and natural product essential for cell proliferation1. To produce the new phospholipid bilayers that are necessary for cell division, extracts, and Compounds Australia[3] were then screened against one isoform of AMPK in order to discover cancer cell must increase de novo generation of lipids and steroids. Actually, several enzymes involved in these novel AMPK activators and inhibitors for further development in biological and medicinal chemistry projects. pathways such as fatty acid synthase, ATP citrate lyase and choline kinase (ChoK) among others, have been associated with tumor development and progression in vitro and in vivo2. Choline is essential for the synthesis of So far, >700 compounds from Nature Bank and >5´300 compounds from a diverse scaffold library from the major membrane phospholipid phosphatidylcholine and choline kinase activity has been shown to be Compounds Australia were screened. Five scaffolds including >40 molecules from the diverse scaffold library upregulated in human cancers and is requires for tumor-cell proliferation2,3. Herein we report a novel and >20 natural products were identified to modulate the activity of AMPK. The structure-activity relationship family of structurally related to hemicolinium-3 (HC-3, a classical ChoK inhibitor known by its neurotoxicity) of information that is already present in the scaffold hits as well as molecular modeling studies of the hit structures non-symmetrical monocationic compounds with antitumor activity. Compound known as 4f, (compared to will be used for the development of more potent AMPK modulators. other, a priori, better candidates from the same series) shows the best IC50 (0.99±0.17) in the inhibition of Human choline quinasa, HsChoKα1 (the isoform upregulated in cancer) something that can be explained References because docking studies show that two wholes molecules of compound are able to take up both binding sites of 1) Rana, S.; Blowers, E. C.; Natarajan, A.; Small Molecule Adenosine 5'-Monophosphate Activated Protein Kinase (AMPK) the enzyme, simultaneously, one molecule binds the ATP binding site of the enzyme and another one binds the Modulators and Human Diseases. J. Med. Chem. 2015, 58, 2-29. choline binding site, the 3-aminophenol fragment of compound 4f is stabilized by two H-bonds, with Tyr354 2) www.griffith.edu.au/science-aviation/eskitis-institute/nature-bank and Glu434 provided by both hydrogens from –NH- and OH groups. All together would explain the better IC50 3) www.griffith.edu.au/science-aviation/compounds-australia of 4f.

References 1) Lindsey, K. Nature Cell Biology 2015, 17, 351-359. 2) Martinez-Outschoorn , U. et al. Nature Reviews Clinical Oncology 2016. doi:10.1038/nrclinonc.2016.60 3) Schiaffino-Ortega, R.et al. Bioorganic & Medicinal Chemistry 2013, 21, 7146-7154

89 P032 P033 ANTICANCER AGENTS BASED ON CIS-RESTRICTED INHIBITION OF AKT BY POLYUNSATURATED COMBRETASTATIN A-4 ANALOGUES PHOSPHATIDYLCHOLINE – A NOVEL APPROACH FOR POTENTIATING LIPOSOMAL ANTI-CANCER THERAPY LUISA CARLOTA LOPEZ CARA (1), MARIA KIMATRAI SALVADOR (1), ROMEO ROMAGNOLI (2), PIER GIOVANNI BARALDI (2), FILLIPPO PRENCIPE (2), STEFANIA BARALDI (2), MOJGAN Konstantin Löser (1), Uli Kazmaier (2), Peter van Hoogevest (3), Oliver Werz (1), Andreas Koeberle (1) AGLHAZADEH TABRIZI (2), SALVATORE FERLA (3), ANDREA BRANCALES (3), ERNEST HAMEL (4), ROBERTO RONCA (5), ROBERTA BORTOLOLOZZI (6), GIUSSEPPE BASSO (6), 1) Chair of Pharmaceutical /Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Germany GIAMPIETRO VIOLA (6) 2) Chair of Organic Chemistry, Saarland University, Saarbrücken, Germany 3) Phospholipid Research Center, Heidelberg, Germany 1) Departamento de Química Orgánica y Farmacéutica, Facultad de Farmacia, Campus de Cartuja s/n, 18071, Granada, Spain 2) Dipartimento di Scienze Farmaceutiche, Università di Ferrara, 44100 Ferrara, Italy 3) School of Pharmacy and Pharmaceutical Sciences, Cardiff University, King Edward VII Avenue, Cardiff, CF10 3NB, UK 4) Screening Technologies Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, Liposomal drug carrier systems are important in cancer chemotherapy, but their own, phospholipid-related, Frederick National Laboratory for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, anti-tumoral potential has not been sufficiently evaluated so far. We have recently shown that Maryland 21702, USA 5) Dipartimento di Medicina molecolare e traslazionale Unità di oncologia sperimentale ed immunologia. Università di phosphatidylcholines with polyunsaturated fatty acids (PUFA-PC) suppress cell proliferation in vitro by Brescia, 25123 Brescia, Italy inhibiting signal transduction of Akt - a key kinase for tumor progression and chemoresistance (protein kinase B) 6) Dipartimento di Salute della Donna e del Bambino, Laboratorio di Oncoematologia, Università di Padova, 35131 1. Whether PUFA-PC sensitizes cells towards anti-tumoral agents by inhibiting the activation of Akt has not Padova, Italy been addressed so far. Here we screened mechanistically diverse small molecule anti-cancer agents in NIH-3T3 fibroblasts and show synergistic suppression of Akt activation and cell proliferation by combined treatment with PC-bound docosahexaenoic acid (22:6-PC) and a myxobacterial miuraenamide derivative which disrupts the actin cytoskeleton. 22:6-PC is efficiently taken up by cells and not significantly degraded to free 22:6 as shown Cancer has a prodigious socioeconomic impact on society as evidenced by the estimated $125 billion spent on by ultraperformance liquid-chromatography ESI tandem mass spectrometry. Moreover, miuraenamides cancer care each year in the United States alone. Actually if one lives long enough, he or she will eventually be concentration-dependently increase the cellular proportion of PUFA-PC along with decreasing Akt activation, impacted by the debilitating malignancy known as cancer. Even if the individual is lucky enough to elude cancer, even without supplementation of 22:6-PC. Conclusively, we demonstrate that the cellular PUFA-PC ratio is it is likely that a close friend or family member will be stricken by a malignant neoplasm1. Natural products, targetable by both small molecules and phospholipids and that a combined strategy is superior in interfering with developed by organism through years of evolutionary selection pressures to combat potential predators have the survival kinase Akt. Therefore, it is tempting to speculate that dietary phospholipids modulate the efficacy of been vital for the development of novel compounds with promising activities against malignancies2. The natural anti-tumoral therapies and that liposomal anti-cancer drugs benefit from 22:6-based cationic phospholipids as product Combretastatin A-4 (CA-4, a tubulin polymerization inhibitir) shows potent cytotoxicity against a drug delivery system. variety of human cancer cell lines and also fascinating vascular disrupting properties that represent a new approach for cancer treatment. A novel series of tubulin polymerization inhibitors, based on the 1-(3′,4′,5′ -trimethoxyphenyl)-2-aryl-1H-imidazole scaffold and designed as cis-restricted combretastatin A-4 analogues, References was synthesized with the goal of evaluating the effects of various patterns of substitution on the phenyl at the 1) Koeberle, A., et al., Arachidonoyl-phosphatidylcholine oscillates during the cell cycle and counteracts proliferation by 2-position of the imidazole ring on biological activity. The best results for inhibition of antiproliferative activity suppressing Akt membrane binding. Proc Natl Acad Sci U S A, 2013. 110(7): p. 2546-51. was obtained with 4′-OEt, 3′ -F-4′ -OEt and 3′ -Cl-4′ -OEt phenyl analogues 4k, 4n and 4o, respectively. In particular, compound 4o exhibited the strongest growth inhibitory activity in the series, with IC50 values ranging from 0.4 to 3.8 nM against seven cancer cells lines. Importantly, 4o showed very low cytotoxicity in proliferating lymphocytes obtained from healthy volunteers, and it was practically ineffective in resting lymphocytes, suggesting that it preferentially was toxic in proliferating cells as compared with quiescent cells. This excellent pharmacological profile was confirmed through in vivo experiments in which the compound was very effective in reducing tumor mass at doses 3-30 times lower in comparison with CA-4P. Our findings suggest that 4o is a promising anticancer drug candidate that warrants further preclinical evaluation3.

References 1) Trendowski, M. Cancer Causes Control 2014, 10, 1243-6.1 2) Trendowski, M. Drugs 2015, 45, 1993-2016. 3) Romagnoli, R.et al. Scientific Reports 2016, 45, 1993-2016

90 P032 P033 ANTICANCER AGENTS BASED ON CIS-RESTRICTED INHIBITION OF AKT BY POLYUNSATURATED COMBRETASTATIN A-4 ANALOGUES PHOSPHATIDYLCHOLINE – A NOVEL APPROACH FOR POTENTIATING LIPOSOMAL ANTI-CANCER THERAPY LUISA CARLOTA LOPEZ CARA (1), MARIA KIMATRAI SALVADOR (1), ROMEO ROMAGNOLI (2), PIER GIOVANNI BARALDI (2), FILLIPPO PRENCIPE (2), STEFANIA BARALDI (2), MOJGAN Konstantin Löser (1), Uli Kazmaier (2), Peter van Hoogevest (3), Oliver Werz (1), Andreas Koeberle (1) AGLHAZADEH TABRIZI (2), SALVATORE FERLA (3), ANDREA BRANCALES (3), ERNEST HAMEL (4), ROBERTO RONCA (5), ROBERTA BORTOLOLOZZI (6), GIUSSEPPE BASSO (6), 1) Chair of Pharmaceutical /Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Germany GIAMPIETRO VIOLA (6) 2) Chair of Organic Chemistry, Saarland University, Saarbrücken, Germany 3) Phospholipid Research Center, Heidelberg, Germany 1) Departamento de Química Orgánica y Farmacéutica, Facultad de Farmacia, Campus de Cartuja s/n, 18071, Granada, Spain 2) Dipartimento di Scienze Farmaceutiche, Università di Ferrara, 44100 Ferrara, Italy 3) School of Pharmacy and Pharmaceutical Sciences, Cardiff University, King Edward VII Avenue, Cardiff, CF10 3NB, UK 4) Screening Technologies Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, Liposomal drug carrier systems are important in cancer chemotherapy, but their own, phospholipid-related, Frederick National Laboratory for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, anti-tumoral potential has not been sufficiently evaluated so far. We have recently shown that Maryland 21702, USA 5) Dipartimento di Medicina molecolare e traslazionale Unità di oncologia sperimentale ed immunologia. Università di phosphatidylcholines with polyunsaturated fatty acids (PUFA-PC) suppress cell proliferation in vitro by Brescia, 25123 Brescia, Italy inhibiting signal transduction of Akt - a key kinase for tumor progression and chemoresistance (protein kinase B) 6) Dipartimento di Salute della Donna e del Bambino, Laboratorio di Oncoematologia, Università di Padova, 35131 1. Whether PUFA-PC sensitizes cells towards anti-tumoral agents by inhibiting the activation of Akt has not Padova, Italy been addressed so far. Here we screened mechanistically diverse small molecule anti-cancer agents in NIH-3T3 fibroblasts and show synergistic suppression of Akt activation and cell proliferation by combined treatment with PC-bound docosahexaenoic acid (22:6-PC) and a myxobacterial miuraenamide derivative which disrupts the actin cytoskeleton. 22:6-PC is efficiently taken up by cells and not significantly degraded to free 22:6 as shown Cancer has a prodigious socioeconomic impact on society as evidenced by the estimated $125 billion spent on by ultraperformance liquid-chromatography ESI tandem mass spectrometry. Moreover, miuraenamides cancer care each year in the United States alone. Actually if one lives long enough, he or she will eventually be concentration-dependently increase the cellular proportion of PUFA-PC along with decreasing Akt activation, impacted by the debilitating malignancy known as cancer. Even if the individual is lucky enough to elude cancer, even without supplementation of 22:6-PC. Conclusively, we demonstrate that the cellular PUFA-PC ratio is it is likely that a close friend or family member will be stricken by a malignant neoplasm1. Natural products, targetable by both small molecules and phospholipids and that a combined strategy is superior in interfering with developed by organism through years of evolutionary selection pressures to combat potential predators have the survival kinase Akt. Therefore, it is tempting to speculate that dietary phospholipids modulate the efficacy of been vital for the development of novel compounds with promising activities against malignancies2. The natural anti-tumoral therapies and that liposomal anti-cancer drugs benefit from 22:6-based cationic phospholipids as product Combretastatin A-4 (CA-4, a tubulin polymerization inhibitir) shows potent cytotoxicity against a drug delivery system. variety of human cancer cell lines and also fascinating vascular disrupting properties that represent a new approach for cancer treatment. A novel series of tubulin polymerization inhibitors, based on the 1-(3′,4′,5′ -trimethoxyphenyl)-2-aryl-1H-imidazole scaffold and designed as cis-restricted combretastatin A-4 analogues, References was synthesized with the goal of evaluating the effects of various patterns of substitution on the phenyl at the 1) Koeberle, A., et al., Arachidonoyl-phosphatidylcholine oscillates during the cell cycle and counteracts proliferation by 2-position of the imidazole ring on biological activity. The best results for inhibition of antiproliferative activity suppressing Akt membrane binding. Proc Natl Acad Sci U S A, 2013. 110(7): p. 2546-51. was obtained with 4′-OEt, 3′ -F-4′ -OEt and 3′ -Cl-4′ -OEt phenyl analogues 4k, 4n and 4o, respectively. In particular, compound 4o exhibited the strongest growth inhibitory activity in the series, with IC50 values ranging from 0.4 to 3.8 nM against seven cancer cells lines. Importantly, 4o showed very low cytotoxicity in proliferating lymphocytes obtained from healthy volunteers, and it was practically ineffective in resting lymphocytes, suggesting that it preferentially was toxic in proliferating cells as compared with quiescent cells. This excellent pharmacological profile was confirmed through in vivo experiments in which the compound was very effective in reducing tumor mass at doses 3-30 times lower in comparison with CA-4P. Our findings suggest that 4o is a promising anticancer drug candidate that warrants further preclinical evaluation3.

References 1) Trendowski, M. Cancer Causes Control 2014, 10, 1243-6.1 2) Trendowski, M. Drugs 2015, 45, 1993-2016. 3) Romagnoli, R.et al. Scientific Reports 2016, 45, 1993-2016

91 P034 P035 NEW MOLECULES DERIVED FROM PREVIOUSLY REPORTED NEW SYMMETRICAL BISCATIONIC CHOLINE KINASE ANTICANCER AGENT WITH POTENT ANTILEUKEMIC ACTIVITY INHIBITORS WITH ANTIPLASMODIAL ACTIVITY ARE INHIBITORS OF STAT5 PHOSPHORYLATION MARIA KIMATRAI SALVADOR (1), LUISA CARLOTA LOPEZ CARA (1), ANTONIO ENTRENA (1), MIGUEL ANGEL GALLO (1), GIANLUCA RUBBINI (1), SERGIO PORTILLO HARO (1), DOLORES MARIA KIMATRAI SALVADOR (1), LUISA CARLOTA LOPEZ CARA (1), ROMEO ROMAGNOLI GONZALEZ PACANOWSKA (2), GUIOMAR PEREZ MORENO (2) (2), PIER GIOVANNI BARALDI (2), FILLIPPO PRENCIPE (2), RICARDO RONDANIN (2), DANIELE SIMONI (2), ERNETS HAMEL (3), STEFANIA GRIMAUDO (4), ROSARIA MARIA PIPITONE (3), MARIA MELI (5), MANLIO TOLOMEO (6) 1) Departamento de Química Orgánica y Farmacéutica, Facultad de Farmacia, Campus de Cartuja s/n, 18071, Granada, Spain 2) Instituto de Parasitología y Biomedicina "López - Neyra", Campus Tecnologico de la Salud, Granada, Spain 1) Departamento de Química Farmacéutica y Orgánica, Facultad de Farmacia, Campus de Cartuja s/n, 18071, Granada, Spain 2) Dipartimento di Scienze Chimiche e Farmaceutiche, Universit_a di Ferrara, 44121 Ferrara, Italy 3) Screening Technologies Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, Frederick National Laboratory for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD According to world malaria report published in 2012 by World Health Organization (WHO) In 2010, 21702, USA there were an estimated 219 million cases of malaria 4) Dipartimento Biomedico di Medicina Interna e Specialistica, Universita di Palermo, 90125 Palermo, Italy (range 154–289 million), and 660 000 malaria deaths 5) Dipartimento di Scienze per la Promozione della Salute e Materno Infantile, Area di Farmacologia, Universita di globally (range 490 000–836 000). Approximately 80% of cases and 90% of deaths are estimated to occur in the Palermo, 90125 Palermo, Italy WHO African Region, with children under five years of age and pregnant women most severely affected1. A 6) Centro Interdipartimentale di Ricerca in Oncologia Clinica e Dipartimento Biomedico di Medicina Interna e major obstacle to the eradication of this disease is the emerging resistance of the malaria-causing parasite Specialistica, Sezione di Malattie Infettive, Universita di Palermo, 90125 Palermo, Italy Plasmodium falciparum to most marketed antimalarial drugs. As such, new compounds acting through novel mechanisms of action are urgently needed2. The proliferation of this parasite within erythrocytes is concomitant with a massive increase of phosphatidylcholine biosynthesis which depends on Choline Kinase enzyme activity the first enzyme in the Kennedy pathway for the biosynthesis of phosphatidylcholine, the most Leukemias continue to cause significat mortality (a heterogeneous group of clonal hematologic malignancies) essential phospholipid in P. falciparum. The inhibition of this enzyme in Plasmodium (PfChoK) disrupts the in adults and in children . The underlying biology of these diseases will be decisive in the course and Kennedy pathway, which results in parasite death2. We have synthesized and published3 a new symmetrical progression of the malady, thus, there is a great interest in treatments directed against inappropriately activated biscationic compounds containing a pair of oxygen atoms in the spacer of the linker between the 1 cell signalig pathways which stimulate the uncontrolled growth of neoplastic cells . Great attention has been biscationic moieties. These compounds resulted be very active as inhibitors of human Choline Kinase, HsChoK STAT S T A T focused on ( ignal ransducer and ctivator of ranscription) proteins that play a crucial role in and antiproliferatives. Herein we present the evaluation of these compounds as antimalarial. The most 1, 2 constitutive mediating the response of hematopoietic cells to certain cytokines . Many studies have shown that promising compound which inhibits the growth of Plasmodium was 10a 1,1′-(((ethane-1,2-diylbis(oxy)) activation of the gene signal transducer and activator of transcription 5 (STAT5) plays an essential role in bis(4,1-phenylene))bis(methylene))bis(4-(dimethylamino)pyridinium) with an IC50= 16nM. In order the pathogenesis of chronic myelogenous leukemia (CML) induced by BCR-ABL STAT5 protein is . to determine the mechanism of action of this compound against P. falciparum we are currently performing considered to be an attractive therapeutic target even more since drug resistance that can be developed more assays with the object of confirming that the antimalarial activity could be due to the inhibition of the Pf through BCR-ABL gene mutations make this oncoprotein refractory to inhibition by tyrosine kinase inhibitors ChoK. such as imatinib2. Molecules that target microtubules have an important role in the treatment of cancer. The 2-(3´,4´,5´-trimethoxybenzoyl)-3-iodoacetylamino-6-methoxybenzo[b]furan derivative (1) (an analogue of the combretastatin natural compound) showed somewhat greater antiproliferative activity than imatinib and had the ability to decrease intracellular levels of phosphorylated STAT5 (pSTAT). We designed a new series of 2-aroyl benzoheterocyclic derivatives structurally related to compound 1 and examined them for their ability to decrease pSTAT5 levels in K562 cells. Preliminary Structure-Activity relationship analysis indicated that (i) the benzo[b]furan nucleus was not indispensable for STAT5 inhibitory activity, (ii) the ortho relationship between the benzoyl and the iodoacetylamino moieties at the C-2 and C-3 positions, respectively, of the benzoheterocyclic nucleus was essential for the STAT5 activity, and (iii) replacing the 3´-4´-5´-trimethoxybenzoyl moiety (a group found in many antitubulin compounds such as colchicine, References podophyllotoxin and combretastatin-A-4) at the 2-position of benzo[b]furan skeleton with a benzoyl group 2 1) http://www.who.int/malaria/publications/world_malaria_report_2012/wmr2012_summary_en.pdf maintained STAT5 inhibitory activity . 2) Rubio-Ruiz. B et al. European Journal of Medicinal Chemistry 2014, 85,289-292. 3) Schiaffino-Ortega .S et al. Scientific Reports, 2016, | 6:23793

References 1) Thomas S Lin et al. Oncogene 2000, 19, 2496-2504. 2) Romagnoli, R. et al. European Journal of Medicinal Chemistry 2016, 108,39-52 92 P034 P035 NEW MOLECULES DERIVED FROM PREVIOUSLY REPORTED NEW SYMMETRICAL BISCATIONIC CHOLINE KINASE ANTICANCER AGENT WITH POTENT ANTILEUKEMIC ACTIVITY INHIBITORS WITH ANTIPLASMODIAL ACTIVITY ARE INHIBITORS OF STAT5 PHOSPHORYLATION MARIA KIMATRAI SALVADOR (1), LUISA CARLOTA LOPEZ CARA (1), ANTONIO ENTRENA (1), MIGUEL ANGEL GALLO (1), GIANLUCA RUBBINI (1), SERGIO PORTILLO HARO (1), DOLORES MARIA KIMATRAI SALVADOR (1), LUISA CARLOTA LOPEZ CARA (1), ROMEO ROMAGNOLI GONZALEZ PACANOWSKA (2), GUIOMAR PEREZ MORENO (2) (2), PIER GIOVANNI BARALDI (2), FILLIPPO PRENCIPE (2), RICARDO RONDANIN (2), DANIELE SIMONI (2), ERNETS HAMEL (3), STEFANIA GRIMAUDO (4), ROSARIA MARIA PIPITONE (3), MARIA MELI (5), MANLIO TOLOMEO (6) 1) Departamento de Química Orgánica y Farmacéutica, Facultad de Farmacia, Campus de Cartuja s/n, 18071, Granada, Spain 2) Instituto de Parasitología y Biomedicina "López - Neyra", Campus Tecnologico de la Salud, Granada, Spain 1) Departamento de Química Farmacéutica y Orgánica, Facultad de Farmacia, Campus de Cartuja s/n, 18071, Granada, Spain 2) Dipartimento di Scienze Chimiche e Farmaceutiche, Universit_a di Ferrara, 44121 Ferrara, Italy 3) Screening Technologies Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, Frederick National Laboratory for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD According to world malaria report published in 2012 by World Health Organization (WHO) In 2010, 21702, USA there were an estimated 219 million cases of malaria 4) Dipartimento Biomedico di Medicina Interna e Specialistica, Universita di Palermo, 90125 Palermo, Italy (range 154–289 million), and 660 000 malaria deaths 5) Dipartimento di Scienze per la Promozione della Salute e Materno Infantile, Area di Farmacologia, Universita di globally (range 490 000–836 000). Approximately 80% of cases and 90% of deaths are estimated to occur in the Palermo, 90125 Palermo, Italy WHO African Region, with children under five years of age and pregnant women most severely affected1. A 6) Centro Interdipartimentale di Ricerca in Oncologia Clinica e Dipartimento Biomedico di Medicina Interna e major obstacle to the eradication of this disease is the emerging resistance of the malaria-causing parasite Specialistica, Sezione di Malattie Infettive, Universita di Palermo, 90125 Palermo, Italy Plasmodium falciparum to most marketed antimalarial drugs. As such, new compounds acting through novel mechanisms of action are urgently needed2. The proliferation of this parasite within erythrocytes is concomitant with a massive increase of phosphatidylcholine biosynthesis which depends on Choline Kinase enzyme activity the first enzyme in the Kennedy pathway for the biosynthesis of phosphatidylcholine, the most Leukemias continue to cause significat mortality (a heterogeneous group of clonal hematologic malignancies) essential phospholipid in P. falciparum. The inhibition of this enzyme in Plasmodium (PfChoK) disrupts the in adults and in children . The underlying biology of these diseases will be decisive in the course and Kennedy pathway, which results in parasite death2. We have synthesized and published3 a new symmetrical progression of the malady, thus, there is a great interest in treatments directed against inappropriately activated biscationic compounds containing a pair of oxygen atoms in the spacer of the linker between the 1 cell signalig pathways which stimulate the uncontrolled growth of neoplastic cells . Great attention has been biscationic moieties. These compounds resulted be very active as inhibitors of human Choline Kinase, HsChoK STAT S T A T focused on ( ignal ransducer and ctivator of ranscription) proteins that play a crucial role in and antiproliferatives. Herein we present the evaluation of these compounds as antimalarial. The most 1, 2 constitutive mediating the response of hematopoietic cells to certain cytokines . Many studies have shown that promising compound which inhibits the growth of Plasmodium was 10a 1,1′-(((ethane-1,2-diylbis(oxy)) activation of the gene signal transducer and activator of transcription 5 (STAT5) plays an essential role in bis(4,1-phenylene))bis(methylene))bis(4-(dimethylamino)pyridinium) bromide with an IC50= 16nM. In order the pathogenesis of chronic myelogenous leukemia (CML) induced by BCR-ABL STAT5 protein is . to determine the mechanism of action of this compound against P. falciparum we are currently performing considered to be an attractive therapeutic target even more since drug resistance that can be developed more assays with the object of confirming that the antimalarial activity could be due to the inhibition of the Pf through BCR-ABL gene mutations make this oncoprotein refractory to inhibition by tyrosine kinase inhibitors ChoK. such as imatinib2. Molecules that target microtubules have an important role in the treatment of cancer. The 2-(3´,4´,5´-trimethoxybenzoyl)-3-iodoacetylamino-6-methoxybenzo[b]furan derivative (1) (an analogue of the combretastatin natural compound) showed somewhat greater antiproliferative activity than imatinib and had the ability to decrease intracellular levels of phosphorylated STAT5 (pSTAT). We designed a new series of 2-aroyl benzoheterocyclic derivatives structurally related to compound 1 and examined them for their ability to decrease pSTAT5 levels in K562 cells. Preliminary Structure-Activity relationship analysis indicated that (i) the benzo[b]furan nucleus was not indispensable for STAT5 inhibitory activity, (ii) the ortho relationship between the benzoyl and the iodoacetylamino moieties at the C-2 and C-3 positions, respectively, of the benzoheterocyclic nucleus was essential for the STAT5 activity, and (iii) replacing the 3´-4´-5´-trimethoxybenzoyl moiety (a group found in many antitubulin compounds such as colchicine, References podophyllotoxin and combretastatin-A-4) at the 2-position of benzo[b]furan skeleton with a benzoyl group 2 1) http://www.who.int/malaria/publications/world_malaria_report_2012/wmr2012_summary_en.pdf maintained STAT5 inhibitory activity . 2) Rubio-Ruiz. B et al. European Journal of Medicinal Chemistry 2014, 85,289-292. 3) Schiaffino-Ortega .S et al. Scientific Reports, 2016, | 6:23793

References 1) Thomas S Lin et al. Oncogene 2000, 19, 2496-2504. 2) Romagnoli, R. et al. European Journal of Medicinal Chemistry 2016, 108,39-52 93 P036 P037 A SUSTAINABLE APPROACH TO RECYCLING CLASSIC DESIGN AND EVALUATION OF NOVEL THERANOSTIC ANTIMALARIALS TOWARDS NEW ANTIPROLIFERATIVE DRUGS FLUOROGENIC DUAL PROBE-PRODRUG IN CANCER

Ana Gomes (1), Iva Fernandes (2), Cátia Teixeira (1,3), M. J. Sottomayor (4), Nuno Mateus (2), Paula Sunil Mathur, Agnes Turnbull, Craig Stevens, David Mincher Gomes (1) Molecular Drug Design Laboratory, Edinburgh Napier University, Edinburgh, UK 1) UCIBIO-REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Porto, Portugal 2) LAQV-REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Porto, Portugal 3) CICECO, Universidade de Aveiro, Aveiro, Portugal. Background: In spite of major advances in the diagnosis and treatment of cancer, there remains a paucity of 4) CIQ-UP, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Porto, Portugal anticancer compounds (adverse drug effects and dose-limiting toxicities) [1]. Legumain is a potential cancer biomarker and a molecular target for imaging and drug targeting. Legumain is a lysosomal protease with a remarkably restricted specificity, as it cleaves only substrate sequences having an asparagine (Asn) at the P1 site [2]. We have previously shown that a legumain substrate probe (SM9), Pro-Ala-Asn~PEG-AQ, is quenched until Recycling classical drugs, by performing simple chemical modifications in order to improve their activity or activated by proteolytic action of legumain [3]. Design of first generation molecular probe SM9 has been repurposing them for other therapeutic targets, could be a sustainable manner to find new drugs.[1] Based on that, extended to a second generation theranostic dual probe-prodrug SM20 (Rho-Pro-Ala-Asn~Lys-PEG-AQ) that in the past few years, our group has been casting on classical antimalarials, such as chloroquine (1) and serves as both a diagnostic and therapeutic (theranostic) tool for cancer. mepacrine (2), and discovered that a simple N-cinnamoylation on those scaffolds (compounds 3 and 4, Figure 1) Material and Methods: Solution and solid phase peptide methods have been used to create a novel improved significantly their antimalarial activity.[2] When evaluated as antiproliferative drugs, compounds 3 and rhodamine-labelled tetra peptide second generation theranostic dual probe-prodrug (SM20) of legumain, which 4 showed activities in micromolar range against three different cancer cell lines (MKN-28, Caco-2 and MCF-7). was purified by chromatographic methods and characterised by high resolution mass spectrometry, together with [3] One of the most interesting and selective hits, from the family of compounds 4 (R = pF), was used to study all synthetic intermediates. Fluorescence spectroscopic methods were used to determine the efficiency of FRET the possible mechanism of action (MOA) behind the antimalarial and antiproliferative activity. The results that in the prodrug (SM20). In vitro fluorimetric assay was developed with human recombinant legumain at 370C in will be presented bring a new hope to search for antitumor leads with DNA binding, as possible MOA. MES assay buffer (pH 5.0). The lipophilic character (Log D) of the SM20 was assessed by distribution coefficient measurements. Confocal microscopy studies were performed to investigate the cellular uptake and sub-cellular localization of both SM9 and SM20 and their legumain-mediated cleavage fragments in PC3 prostate cancer cells. Results: The fluorogenic second generation prodrug (SM20) is an efficient FRET substrate and affords good restoration of fluorescence when incubated at 10μM with rh-legumain (40ng) in legumain assay buffer (pH-5.0; λex 544nm, λem 585nm). Cleavage at the designed Asn~Lys (SM20) cleavage ‘hotspot' was demonstrated at concentrations of legumain in the 5ng-40ng range in in vitro metabolism experiments using recombinant legumain. The second generation theranostic prodrug (SM20) is structurally similar to the SM9 probe, with a different aminoanthraquinone quencher, which is lysosomotropic. Measurement of the distribution coefficients have shown that SM20 is more lipophilic than the Lys-PEG-AQ cleavage product from which it is derived. Confocal studies for 0-1hr at 63x magnification have shown that both SM9 (1M) and SM20 (1M) were localized in the lysosomes of PC3 prostate cancer cells. Conclusions: The theranostic dual probe-prodrug (SM20) is an efficient substrate for sensitive and early detection of legumain and a smart therapeutic agent for cancer. Work is ongoing to optimize the dual probe-prodrug and to extend the studies in vivo.

References 1) Drucker, E. and Krapfenbauer, K. (2013) Pitfalls and limitations in translation from biomarker discovery to clinical utility Figure 1: Chloroquine (1), mepacrine (2) and respective surrogates 3 and 4. in predictive and personalised medicine. EPMA J, 4(1). 7. 2) Dall, E. and Brandstetter, H. (2015) Structure and function of legumain in health and disease. Biochimie. 9084(15), 00297-7. Acknowledgements: Thanks are due to Fundação para a Ciência e Tecnologia (FCT, Portugal) for funding 3) Mathur, S., Turnbull, A., Stevens, C., Poole, A and Mincher, J.D. (2014) Design and Evaluation of New Theranostic Molecular Probes in Cancer. In: APS PharmSci 2014, 8-10 Sep 2014, University of Hertfordshire, Hatfield, UK. research units UCIBIO-REQUIMTE (ref. UID/MULTI/04378/2013), LAQV-REQUMTE (ref. UID/QUI/50006/2013) and CIQ-UP (ref. UID/QUI/0081/2013). CT and IF thank FCT for post-doctoral grants SFRH/BPD/62967/2009 and SFRH/BPD/86173/2012, respectively.

References

1) (a) C. Teixeira, et al. Chem. Rev. 2014, 114, 11164-11220; (b) K.T. Andrews et al. Int. J. Parasitol. Drugs Drug Resist. 2014, 4, 95–111; 2) (a) B. Pérez, et al., J. Med. Chem. 2013, 56, 556-567; (b) A. Gomes, et al., Chem. Med. Chem. 2014, 9, 305-310; 3) B. Pérez, et al., Bioorg. Med. Chem. Lett. 2013, 23, 6769-6772.

94 P036 P037 A SUSTAINABLE APPROACH TO RECYCLING CLASSIC DESIGN AND EVALUATION OF NOVEL THERANOSTIC ANTIMALARIALS TOWARDS NEW ANTIPROLIFERATIVE DRUGS FLUOROGENIC DUAL PROBE-PRODRUG IN CANCER

Ana Gomes (1), Iva Fernandes (2), Cátia Teixeira (1,3), M. J. Sottomayor (4), Nuno Mateus (2), Paula Sunil Mathur, Agnes Turnbull, Craig Stevens, David Mincher Gomes (1) Molecular Drug Design Laboratory, Edinburgh Napier University, Edinburgh, UK 1) UCIBIO-REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Porto, Portugal 2) LAQV-REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Porto, Portugal 3) CICECO, Universidade de Aveiro, Aveiro, Portugal. Background: In spite of major advances in the diagnosis and treatment of cancer, there remains a paucity of 4) CIQ-UP, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Porto, Portugal anticancer compounds (adverse drug effects and dose-limiting toxicities) [1]. Legumain is a potential cancer biomarker and a molecular target for imaging and drug targeting. Legumain is a lysosomal protease with a remarkably restricted specificity, as it cleaves only substrate sequences having an asparagine (Asn) at the P1 site [2]. We have previously shown that a legumain substrate probe (SM9), Pro-Ala-Asn~PEG-AQ, is quenched until Recycling classical drugs, by performing simple chemical modifications in order to improve their activity or activated by proteolytic action of legumain [3]. Design of first generation molecular probe SM9 has been repurposing them for other therapeutic targets, could be a sustainable manner to find new drugs.[1] Based on that, extended to a second generation theranostic dual probe-prodrug SM20 (Rho-Pro-Ala-Asn~Lys-PEG-AQ) that in the past few years, our group has been casting on classical antimalarials, such as chloroquine (1) and serves as both a diagnostic and therapeutic (theranostic) tool for cancer. mepacrine (2), and discovered that a simple N-cinnamoylation on those scaffolds (compounds 3 and 4, Figure 1) Material and Methods: Solution and solid phase peptide methods have been used to create a novel improved significantly their antimalarial activity.[2] When evaluated as antiproliferative drugs, compounds 3 and rhodamine-labelled tetra peptide second generation theranostic dual probe-prodrug (SM20) of legumain, which 4 showed activities in micromolar range against three different cancer cell lines (MKN-28, Caco-2 and MCF-7). was purified by chromatographic methods and characterised by high resolution mass spectrometry, together with [3] One of the most interesting and selective hits, from the family of compounds 4 (R = pF), was used to study all synthetic intermediates. Fluorescence spectroscopic methods were used to determine the efficiency of FRET the possible mechanism of action (MOA) behind the antimalarial and antiproliferative activity. The results that in the prodrug (SM20). In vitro fluorimetric assay was developed with human recombinant legumain at 370C in will be presented bring a new hope to search for antitumor leads with DNA binding, as possible MOA. MES assay buffer (pH 5.0). The lipophilic character (Log D) of the SM20 was assessed by distribution coefficient measurements. Confocal microscopy studies were performed to investigate the cellular uptake and sub-cellular localization of both SM9 and SM20 and their legumain-mediated cleavage fragments in PC3 prostate cancer cells. Results: The fluorogenic second generation prodrug (SM20) is an efficient FRET substrate and affords good restoration of fluorescence when incubated at 10μM with rh-legumain (40ng) in legumain assay buffer (pH-5.0; λex 544nm, λem 585nm). Cleavage at the designed Asn~Lys (SM20) cleavage ‘hotspot' was demonstrated at concentrations of legumain in the 5ng-40ng range in in vitro metabolism experiments using recombinant legumain. The second generation theranostic prodrug (SM20) is structurally similar to the SM9 probe, with a different aminoanthraquinone quencher, which is lysosomotropic. Measurement of the distribution coefficients have shown that SM20 is more lipophilic than the Lys-PEG-AQ cleavage product from which it is derived. Confocal studies for 0-1hr at 63x magnification have shown that both SM9 (1M) and SM20 (1M) were localized in the lysosomes of PC3 prostate cancer cells. Conclusions: The theranostic dual probe-prodrug (SM20) is an efficient substrate for sensitive and early detection of legumain and a smart therapeutic agent for cancer. Work is ongoing to optimize the dual probe-prodrug and to extend the studies in vivo.

References 1) Drucker, E. and Krapfenbauer, K. (2013) Pitfalls and limitations in translation from biomarker discovery to clinical utility Figure 1: Chloroquine (1), mepacrine (2) and respective surrogates 3 and 4. in predictive and personalised medicine. EPMA J, 4(1). 7. 2) Dall, E. and Brandstetter, H. (2015) Structure and function of legumain in health and disease. Biochimie. 9084(15), 00297-7. Acknowledgements: Thanks are due to Fundação para a Ciência e Tecnologia (FCT, Portugal) for funding 3) Mathur, S., Turnbull, A., Stevens, C., Poole, A and Mincher, J.D. (2014) Design and Evaluation of New Theranostic Molecular Probes in Cancer. In: APS PharmSci 2014, 8-10 Sep 2014, University of Hertfordshire, Hatfield, UK. research units UCIBIO-REQUIMTE (ref. UID/MULTI/04378/2013), LAQV-REQUMTE (ref. UID/QUI/50006/2013) and CIQ-UP (ref. UID/QUI/0081/2013). CT and IF thank FCT for post-doctoral grants SFRH/BPD/62967/2009 and SFRH/BPD/86173/2012, respectively.

References

1) (a) C. Teixeira, et al. Chem. Rev. 2014, 114, 11164-11220; (b) K.T. Andrews et al. Int. J. Parasitol. Drugs Drug Resist. 2014, 4, 95–111; 2) (a) B. Pérez, et al., J. Med. Chem. 2013, 56, 556-567; (b) A. Gomes, et al., Chem. Med. Chem. 2014, 9, 305-310; 3) B. Pérez, et al., Bioorg. Med. Chem. Lett. 2013, 23, 6769-6772.

95 P038 P039 DENDRIMER-SUBSTITUTED SULFANYL PORPHYRAZINES AS DIAZEPINOPORPHYRAZINES WITH HYPERBRANCHED PROMISING PHOTOSENSITIZERS FOR PDT SUBSTITUENTS AS POTENTIAL PHOTOSENSITIZERS FOR PHOTODYNAMIC THERAPY (PDT) Dariusz T. Mlynarczyk (1), Jaroslaw Piskorz (2), Sebastian Lijewski (1), Michal Falkowski (1), Magdalena Stolarska (1,3), Wojciech Szczolko (1), Lukasz Sobotta (2), Lukasz Popenda (4), Stefan Jurga (4,5), Ewelina Wieczorek (1), Dariusz T. Mlynarczyk (1), Jaroslaw Piskorz (2), Lukasz Popenda (3), Anna Krystyna Konopka (6), Nejat Düzgüneş (6), Jadwiga Mielcarek (2), Tomasz Goslinski (1) Wzgarda (4), Stefan Jurga (3,5), Jadwiga Mielcarek (2), Tomasz Goslinski (1)

1) Department of Chemical Technology of Drugs, Poznan University of Medical Sciences, Grunwaldzka 6, 60-780 Poznan 1) Department of Chemical Technology of Drugs, Poznan University of Medical Sciences, Grunwaldzka 6, 60-780 Poznan, 2) Department of Inorganic and Analytical Chemistry, Poznan University of Medical Sciences, Grunwaldzka 6, 60-780 Poland Poznan 2) Department of Inorganic and Analytical Chemistry, Poznan University of Medical Sciences, Grunwaldzka 6, 60-780 3) Department of Pharmaceutical Technology, Poznan University of Medical Sciences, Grunwaldzka 6, 61-780 Poznan, Poznan, Poland Poland 3) NanoBioMedical Centre, Adam Mickiewicz University, Umultowska 85, 61-614 Poznan, Poland 4) NanoBioMedical Centre, Adam Mickiewicz University, Umultowska 85, 61-614 Poznan 4) Department of Pharmaceutical Chemistry, Poznan University of Medical Sciences, Grunwaldzka 6 Street, 60-780 Poznan, 5) Department of Macromolecular Physics, Adam Mickiewicz University, Umultowska 85, 61-614 Poznan Poland 6) Department of Biomedical Sciences, University of the Pacific, 2155 Webster Street, San Francisco, CA 94115, USA 5) Department of Macromolecular Physics, Adam Mickiewicz University in Poznan, Umultowska 85, 61-614 Poznan, Poland

Cancer has become the main cause of death in developed countries. Therefore, there is a constant need to Porphyrazines (Pzs) belong to macrocyclic compounds possessing interesting optical properties, especially develop new methods for treatment of this disease. One of the promising techniques is photodynamic therapy fluorescence and an ability to generate singlet oxygen. Therefore Pzs can be considered as potential agents for (PDT). PDT is based on activation of photosensitizer with light of an appropriate wavelength (Figure), followed Photodynamic Therapy (PDT) of cancer [1]. by generation of reactive oxygen species, especially singlet oxygen (1O2), which induce tumor cells death on the basis of necrosis or apoptosis. The main advantage of PDT over other methods is the negligible ability of cancer PDT is based on the irradiation of photosensitizer localized at the affected site of the tissue. This process causes cells to develop specific resistance [1]. generation of the singlet oxygen. It is known that conversion of light energy into chemical energy leads to chemical destruction of tissues or pathogens. Pzs possessing peripherally annulated diazepine rings have been subjected to advanced physical–chemical studies [2]. The UV–vis spectra of these macrocycles revealed a broad Q-band, which in most of the solvents, is divided into two sub-bands located in the range of 630–640 nm and 660–680 nm. The presence of long Q-band absorption wavelengths observed for diazepinoporphyrazines suggests that these macrocycles can be considered as potential photosensitizers for PDT, as light of longer wavelength is capable of deeper penetration of irradiated tissue. In our study we present synthesis of novel diazepineporphyrazines with peripheral 4-methoxyphenyl and 3,5-bis(benzyloxy)benzyloxyphenyl substituents. The synthesis of the expected porphyrazines was performed following a multistep procedure starting from commercially available diaminomaleonitrile. The dimainomaleonitrile derivatives were subjected to the macrocyclization reactions with magnesium n-butanolate in n-butanol to give novel magnesium porphyrazines. Macrocyclic products were carefully purified via flash colum chromatography and characterized using various analytical techniques, especially NMR (1D and 2D). The potential photosensitizing activities of the obtained porphyrazines were evaluated by measuring their abilities for singlet oxygen production, which is the result of an interaction between an activated photosensitizer and oxygen. Our study was focused on the synthesis and characterization of novel photosensitizers suitable for PDT. Symmetrical and unsymmetrical porphyrazines substituted with dendrimeric moieties were synthesized and characterized using spectral techniques with special emphasis on their photophysical properties. Effectiveness of This study was supported by the National Science Centre under grant No 2012/05/E/NZ7/01204. PL and SJ macrocycles was assessed in photodynamic activity assays against human oral squamous carcinoma cell lines. acknowledge financial support from the National Centre for Research and Development under grant No Among the porphyrazines screened, unsymmetrical tribenzoporphyrazines were found to be highly active, with PBS1/A9/13/2012. IC50 values up to nanomolar range, both in their free form and after embedding in liposomes. This study was supported by the National Science Centre under grant No 2012/05/E/NZ7/01204. LP and SJ References acknowledge financial support from the National Centre for Research and Development under grant No 1) S. Yano, S. Hirohara, M. Obata, Y. Hagiya, S. Ogura, A. Ikeda, H. Kataoka, M. Tanaka, T. Joh (2011) Current states and PBS1/A9/13/2012. KK and ND acknowledge funds from the University of the Pacific, Arthur A. Dugoni School future views in phot odynamic therapy, J. Photochem. Photobiol. 12(1):46-67 of Dentistry. 2) J. Piskorz, K. Konopka, N. Düzgüneş, Z. Gdaniec, J. Mielcarek, T. Goslinski (2014) Diazepinoporphyrazines Containing Peripheral Styryl Substituents and Their Promising Nanomolar Photodynamic Activity against Oral Cancer Cells in Liposomal Formulations, ChemMedChem 9(8):1775-1782 References 1) Bonavida B., Rapozzi V., Jori G. (Eds.) Resistance to Photodynamic Therapy in Cancer of Resistance to Targeted Anti-cancer Therapeutics, vol. 5, Springer, 2015. doi:10.1007/978-3-319-12730-9

96 P038 P039 DENDRIMER-SUBSTITUTED SULFANYL PORPHYRAZINES AS DIAZEPINOPORPHYRAZINES WITH HYPERBRANCHED PROMISING PHOTOSENSITIZERS FOR PDT SUBSTITUENTS AS POTENTIAL PHOTOSENSITIZERS FOR PHOTODYNAMIC THERAPY (PDT) Dariusz T. Mlynarczyk (1), Jaroslaw Piskorz (2), Sebastian Lijewski (1), Michal Falkowski (1), Magdalena Stolarska (1,3), Wojciech Szczolko (1), Lukasz Sobotta (2), Lukasz Popenda (4), Stefan Jurga (4,5), Ewelina Wieczorek (1), Dariusz T. Mlynarczyk (1), Jaroslaw Piskorz (2), Lukasz Popenda (3), Anna Krystyna Konopka (6), Nejat Düzgüneş (6), Jadwiga Mielcarek (2), Tomasz Goslinski (1) Wzgarda (4), Stefan Jurga (3,5), Jadwiga Mielcarek (2), Tomasz Goslinski (1)

1) Department of Chemical Technology of Drugs, Poznan University of Medical Sciences, Grunwaldzka 6, 60-780 Poznan 1) Department of Chemical Technology of Drugs, Poznan University of Medical Sciences, Grunwaldzka 6, 60-780 Poznan, 2) Department of Inorganic and Analytical Chemistry, Poznan University of Medical Sciences, Grunwaldzka 6, 60-780 Poland Poznan 2) Department of Inorganic and Analytical Chemistry, Poznan University of Medical Sciences, Grunwaldzka 6, 60-780 3) Department of Pharmaceutical Technology, Poznan University of Medical Sciences, Grunwaldzka 6, 61-780 Poznan, Poznan, Poland Poland 3) NanoBioMedical Centre, Adam Mickiewicz University, Umultowska 85, 61-614 Poznan, Poland 4) NanoBioMedical Centre, Adam Mickiewicz University, Umultowska 85, 61-614 Poznan 4) Department of Pharmaceutical Chemistry, Poznan University of Medical Sciences, Grunwaldzka 6 Street, 60-780 Poznan, 5) Department of Macromolecular Physics, Adam Mickiewicz University, Umultowska 85, 61-614 Poznan Poland 6) Department of Biomedical Sciences, University of the Pacific, 2155 Webster Street, San Francisco, CA 94115, USA 5) Department of Macromolecular Physics, Adam Mickiewicz University in Poznan, Umultowska 85, 61-614 Poznan, Poland

Cancer has become the main cause of death in developed countries. Therefore, there is a constant need to Porphyrazines (Pzs) belong to macrocyclic compounds possessing interesting optical properties, especially develop new methods for treatment of this disease. One of the promising techniques is photodynamic therapy fluorescence and an ability to generate singlet oxygen. Therefore Pzs can be considered as potential agents for (PDT). PDT is based on activation of photosensitizer with light of an appropriate wavelength (Figure), followed Photodynamic Therapy (PDT) of cancer [1]. by generation of reactive oxygen species, especially singlet oxygen (1O2), which induce tumor cells death on the basis of necrosis or apoptosis. The main advantage of PDT over other methods is the negligible ability of cancer PDT is based on the irradiation of photosensitizer localized at the affected site of the tissue. This process causes cells to develop specific resistance [1]. generation of the singlet oxygen. It is known that conversion of light energy into chemical energy leads to chemical destruction of tissues or pathogens. Pzs possessing peripherally annulated diazepine rings have been subjected to advanced physical–chemical studies [2]. The UV–vis spectra of these macrocycles revealed a broad Q-band, which in most of the solvents, is divided into two sub-bands located in the range of 630–640 nm and 660–680 nm. The presence of long Q-band absorption wavelengths observed for diazepinoporphyrazines suggests that these macrocycles can be considered as potential photosensitizers for PDT, as light of longer wavelength is capable of deeper penetration of irradiated tissue. In our study we present synthesis of novel diazepineporphyrazines with peripheral 4-methoxyphenyl and 3,5-bis(benzyloxy)benzyloxyphenyl substituents. The synthesis of the expected porphyrazines was performed following a multistep procedure starting from commercially available diaminomaleonitrile. The dimainomaleonitrile derivatives were subjected to the macrocyclization reactions with magnesium n-butanolate in n-butanol to give novel magnesium porphyrazines. Macrocyclic products were carefully purified via flash colum chromatography and characterized using various analytical techniques, especially NMR (1D and 2D). The potential photosensitizing activities of the obtained porphyrazines were evaluated by measuring their abilities for singlet oxygen production, which is the result of an interaction between an activated photosensitizer and oxygen. Our study was focused on the synthesis and characterization of novel photosensitizers suitable for PDT. Symmetrical and unsymmetrical porphyrazines substituted with dendrimeric moieties were synthesized and characterized using spectral techniques with special emphasis on their photophysical properties. Effectiveness of This study was supported by the National Science Centre under grant No 2012/05/E/NZ7/01204. PL and SJ macrocycles was assessed in photodynamic activity assays against human oral squamous carcinoma cell lines. acknowledge financial support from the National Centre for Research and Development under grant No Among the porphyrazines screened, unsymmetrical tribenzoporphyrazines were found to be highly active, with PBS1/A9/13/2012. IC50 values up to nanomolar range, both in their free form and after embedding in liposomes. This study was supported by the National Science Centre under grant No 2012/05/E/NZ7/01204. LP and SJ References acknowledge financial support from the National Centre for Research and Development under grant No 1) S. Yano, S. Hirohara, M. Obata, Y. Hagiya, S. Ogura, A. Ikeda, H. Kataoka, M. Tanaka, T. Joh (2011) Current states and PBS1/A9/13/2012. KK and ND acknowledge funds from the University of the Pacific, Arthur A. Dugoni School future views in phot odynamic therapy, J. Photochem. Photobiol. 12(1):46-67 of Dentistry. 2) J. Piskorz, K. Konopka, N. Düzgüneş, Z. Gdaniec, J. Mielcarek, T. Goslinski (2014) Diazepinoporphyrazines Containing Peripheral Styryl Substituents and Their Promising Nanomolar Photodynamic Activity against Oral Cancer Cells in Liposomal Formulations, ChemMedChem 9(8):1775-1782 References 1) Bonavida B., Rapozzi V., Jori G. (Eds.) Resistance to Photodynamic Therapy in Cancer of Resistance to Targeted Anti-cancer Therapeutics, vol. 5, Springer, 2015. doi:10.1007/978-3-319-12730-9

97 P040 P041 SYNTHESIS AND PHYSICO-CHEMICAL PROPERTIES OF DENDRIMERIC PHTHALOCYANINE AND DENDRITIC SULFANYL PORPHYRAZINES OF POTENTIAL PYRAZINOPORPHYRAZINE DERIVATIVES AS POTENTIAL APPLICATIONS IN NANOMEDICINE PHOTOSENSITIZERS FOR PHOTODYNAMIC THERAPY (PDT)

Michal Falkowski (1), Lukasz Popenda (2), Tomasz Rebis (1,3), Jaroslaw Piskorz (4), Ewelina Wieczorek Adam Tillo (1), Dariusz T. Mlynarczyk (1), Ewelina Wieczorek (1), Michal Falkowski (1), Lukasz (1), Anna Wzgarda (5), Adam Tillo (1), Dariusz T. Mlynarczyk (1), Stefan Jurga (2,6), Jadwiga Mielcarek Popenda (2), Barbara Wicher (1), Michal Kryjewski (3), Wojciech Szczolko (1), Anna Wzgarda (4), Stefan (4), Grzegorz Milczarek (3), Tomasz Goslinski (1) Jurga (2,5), Jadwiga Mielcarek (3), Tomasz Goslinski (1), Ewa Tykarska (1)

1) Department of Chemical Technology of Drugs, Poznan University of Medical Sciences, Grunwaldzka 6, 60-780 Poznan, 1) Department of Chemical Technology of Drugs, Poznan University of Medical Sciences, Grunwaldzka 6, 60-780 Poznan, Poland Poland 2) NanoBioMedical Centre, Adam Mickiewicz University in Poznan, Umultowska 85, 61-614 Poznan, Poland 2) NanoBioMedical Centre, Adam Mickiewicz University, Umultowska 85, 61-614 Poznan, Poland 3) Institute of Chemistry and Technical Electrochemistry, Poznan University of Technology, Berdychowo 4, 60-965 Poznan, 3) Department of Inorganic and Analytical Chemistry, Poznan University of Medical Sciences, Grunwaldzka 6, 60-780 Poland Poznan, Poland 4) Department of Inorganic and Analytical Chemistry, Poznan University of Medical Sciences, Grunwaldzka 6, 60-780 4) Department of Pharmaceutical Chemistry, Poznan University of Medical Sciences, Grunwaldzka 6 Street, 60-780 Poznan, Poznan, Poland Poland 5) Department of Pharmaceutical Chemistry, Poznan University of Medical Sciences, Grunwaldzka 6 Street, 60-780 Poznan, 5) Department of Macromolecular Physics, Faculty of Physics, Adam Mickiewicz University, Umultowska 85, 61-614 Poland Poznan, Poland 6) Department of Macromolecular Physics, Adam Mickiewicz University in Poznan, Umultowska 85, 61-614 Poznan, Poland

Porphyrazines (Pz) are synthetic macrocycles, which are analogues of naturally occurring porphyrinoids like Porphyrazines (Pzs) are synthetic analogues of naturally occurring porphyrins. They consist of four pyrrole rings heme or chlorophyll. Pyrrole rings found in the studied structures are bound together by aza bridges, in contrast linked together with azamethine groups in place of methine bridges present in porphyrins. Pzs may be modified to methine groups found in porphyrins. Chemically obtained analogues of porphyrinoids have been for many in their periphery by various substituents of which sulfanyl groups are known to enrich them with interesting years a subject of interest both for technical as well as biomedical sciences. Some of the recent applications of physico-chemical and electrochemical properties. They revealed potential towards nanotechnology and synthetic and semi-synthetic porphyrinoids in medicine include: photodynamic therapy (PDT), boron-neutron medicine, especially as photosensitizers for photodynamic therapy of cancer and noncancerous diseases. capture therapy (BNCT), radiation therapy and bioimaging [1]. The aim of this study was to explore the novel Dendrimers are complex, chemical compounds consisting of centrally located core and spherical shell of structures for their possible use in PDT. In this medical therapeutic approach, photosensitizer is delivered to the polymeric chains. These chains, called dendrons can be constructed of many types of monomers what results in tumor tissue, where upon irradiation with light of an appropriate wavelength, generates reactive oxygen species, diverse chemical and physical properties. including singlet oxygen. These have the ability to kill tumor cells leading to necrotic or apoptotic cell death. Carbon nanotubes (CNT) have gained considerable attention in research due to their unique electronic properties, Currently used photosensitizers are still far from being ideal. Improving their biophysical properties, while chemical stability and affinity to biomolecules. It was found that the CNT can promote effective electron transfer maintaining high singlet oxygen generation yield is a subject of ongoing research. One of the main factors, reactions. Moreover, they can be used as a support for immobilization of different electron transfer mediators hampering biological activity of porphyrinoids is their poor solubility in water and the tendency to form onto electrode surfaces able to improve their electrochemical properties. CNT can be easily modified by covalent aggregates. Noteworthy is that aggregation of the macrocycles can lead to significant decrease of their functionalization, electropolymerization or adsorption. photosensitizing efficacy [2]. The resulting products were characterized using various analytical techniques, especially NMR, MALDI and In presented work, six novel porphyrazines, representing A4 and A3B type have been synthesized. The effects of UV-Vis. Moreover, the morphology of hybrid nanostructures was researched using SEM and AFM microscopy porphyrazine symmetry, annulation of the macrocyclic core with pyrazine rings as well as expansion of the and they were subjected to electrochemical study. The modified electrodes were also utilized for electrocatalytic periphery with bulky substituents were studied. The potential application of novel macrocycles in photodynamic determination of hydrogen peroxide, an important compound generated in many enzymatic reactions. The strong therapy was evaluated by examining the yield of singlet oxygen generation and an aggregation tendency. synergic effect between two nanostructural components, namely multi-walled carbon nanotubes and dendrimeric porphyrazines was evaluated. Acknowledgements

This study was supported by the National Science Centre under grant No 2012/05/E/NZ7/01204 and by the National Centre for Research and Development under grant No PBS1/A9/13/2012.

This study was supported by the National Science Centre under grant No 2012/05/E/NZ7/01204. PL and SJ acknowledge financial support from the National Centre for Research and Development under grant No PBS1/A9/13/2012.

References 1) Vicente, M.G. Curr. Med. Chem.-Anti-Cancer Agents 1 (2001) 175–194 98 2) Jaung, J-Y. Dyes Pigm. 75 (2007) 420–425 P040 P041 SYNTHESIS AND PHYSICO-CHEMICAL PROPERTIES OF DENDRIMERIC PHTHALOCYANINE AND DENDRITIC SULFANYL PORPHYRAZINES OF POTENTIAL PYRAZINOPORPHYRAZINE DERIVATIVES AS POTENTIAL APPLICATIONS IN NANOMEDICINE PHOTOSENSITIZERS FOR PHOTODYNAMIC THERAPY (PDT)

Michal Falkowski (1), Lukasz Popenda (2), Tomasz Rebis (1,3), Jaroslaw Piskorz (4), Ewelina Wieczorek Adam Tillo (1), Dariusz T. Mlynarczyk (1), Ewelina Wieczorek (1), Michal Falkowski (1), Lukasz (1), Anna Wzgarda (5), Adam Tillo (1), Dariusz T. Mlynarczyk (1), Stefan Jurga (2,6), Jadwiga Mielcarek Popenda (2), Barbara Wicher (1), Michal Kryjewski (3), Wojciech Szczolko (1), Anna Wzgarda (4), Stefan (4), Grzegorz Milczarek (3), Tomasz Goslinski (1) Jurga (2,5), Jadwiga Mielcarek (3), Tomasz Goslinski (1), Ewa Tykarska (1)

1) Department of Chemical Technology of Drugs, Poznan University of Medical Sciences, Grunwaldzka 6, 60-780 Poznan, 1) Department of Chemical Technology of Drugs, Poznan University of Medical Sciences, Grunwaldzka 6, 60-780 Poznan, Poland Poland 2) NanoBioMedical Centre, Adam Mickiewicz University in Poznan, Umultowska 85, 61-614 Poznan, Poland 2) NanoBioMedical Centre, Adam Mickiewicz University, Umultowska 85, 61-614 Poznan, Poland 3) Institute of Chemistry and Technical Electrochemistry, Poznan University of Technology, Berdychowo 4, 60-965 Poznan, 3) Department of Inorganic and Analytical Chemistry, Poznan University of Medical Sciences, Grunwaldzka 6, 60-780 Poland Poznan, Poland 4) Department of Inorganic and Analytical Chemistry, Poznan University of Medical Sciences, Grunwaldzka 6, 60-780 4) Department of Pharmaceutical Chemistry, Poznan University of Medical Sciences, Grunwaldzka 6 Street, 60-780 Poznan, Poznan, Poland Poland 5) Department of Pharmaceutical Chemistry, Poznan University of Medical Sciences, Grunwaldzka 6 Street, 60-780 Poznan, 5) Department of Macromolecular Physics, Faculty of Physics, Adam Mickiewicz University, Umultowska 85, 61-614 Poland Poznan, Poland 6) Department of Macromolecular Physics, Adam Mickiewicz University in Poznan, Umultowska 85, 61-614 Poznan, Poland

Porphyrazines (Pz) are synthetic macrocycles, which are analogues of naturally occurring porphyrinoids like Porphyrazines (Pzs) are synthetic analogues of naturally occurring porphyrins. They consist of four pyrrole rings heme or chlorophyll. Pyrrole rings found in the studied structures are bound together by aza bridges, in contrast linked together with azamethine groups in place of methine bridges present in porphyrins. Pzs may be modified to methine groups found in porphyrins. Chemically obtained analogues of porphyrinoids have been for many in their periphery by various substituents of which sulfanyl groups are known to enrich them with interesting years a subject of interest both for technical as well as biomedical sciences. Some of the recent applications of physico-chemical and electrochemical properties. They revealed potential towards nanotechnology and synthetic and semi-synthetic porphyrinoids in medicine include: photodynamic therapy (PDT), boron-neutron medicine, especially as photosensitizers for photodynamic therapy of cancer and noncancerous diseases. capture therapy (BNCT), radiation therapy and bioimaging [1]. The aim of this study was to explore the novel Dendrimers are complex, chemical compounds consisting of centrally located core and spherical shell of structures for their possible use in PDT. In this medical therapeutic approach, photosensitizer is delivered to the polymeric chains. These chains, called dendrons can be constructed of many types of monomers what results in tumor tissue, where upon irradiation with light of an appropriate wavelength, generates reactive oxygen species, diverse chemical and physical properties. including singlet oxygen. These have the ability to kill tumor cells leading to necrotic or apoptotic cell death. Carbon nanotubes (CNT) have gained considerable attention in research due to their unique electronic properties, Currently used photosensitizers are still far from being ideal. Improving their biophysical properties, while chemical stability and affinity to biomolecules. It was found that the CNT can promote effective electron transfer maintaining high singlet oxygen generation yield is a subject of ongoing research. One of the main factors, reactions. Moreover, they can be used as a support for immobilization of different electron transfer mediators hampering biological activity of porphyrinoids is their poor solubility in water and the tendency to form onto electrode surfaces able to improve their electrochemical properties. CNT can be easily modified by covalent aggregates. Noteworthy is that aggregation of the macrocycles can lead to significant decrease of their functionalization, electropolymerization or adsorption. photosensitizing efficacy [2]. The resulting products were characterized using various analytical techniques, especially NMR, MALDI and In presented work, six novel porphyrazines, representing A4 and A3B type have been synthesized. The effects of UV-Vis. Moreover, the morphology of hybrid nanostructures was researched using SEM and AFM microscopy porphyrazine symmetry, annulation of the macrocyclic core with pyrazine rings as well as expansion of the and they were subjected to electrochemical study. The modified electrodes were also utilized for electrocatalytic periphery with bulky substituents were studied. The potential application of novel macrocycles in photodynamic determination of hydrogen peroxide, an important compound generated in many enzymatic reactions. The strong therapy was evaluated by examining the yield of singlet oxygen generation and an aggregation tendency. synergic effect between two nanostructural components, namely multi-walled carbon nanotubes and dendrimeric porphyrazines was evaluated. Acknowledgements

This study was supported by the National Science Centre under grant No 2012/05/E/NZ7/01204 and by the National Centre for Research and Development under grant No PBS1/A9/13/2012.

This study was supported by the National Science Centre under grant No 2012/05/E/NZ7/01204. PL and SJ acknowledge financial support from the National Centre for Research and Development under grant No PBS1/A9/13/2012.

References 1) Vicente, M.G. Curr. Med. Chem.-Anti-Cancer Agents 1 (2001) 175–194 2) Jaung, J-Y. Dyes Pigm. 75 (2007) 420–425 99 P042 P043 DESIGN AND DEVELOPMENT OF POTENT SERIES OF IMPAIRED FATTY ACID BIOSYNTHESIS DURING APOPTOSIS REVERSIBLE INHIBITORS OF LYSINE SPECIFIC METHYLASE 1 DECREASES AKT PHOSPHORYLATION THROUGH SPECIFIC CHANGES IN THE CELLULAR PHOSPHOLIPID COMPOSITION Daniel Mould, Alison McGonagle, Allan Jordan, Donald Ogilvie Helmut Pein, Oliver Werz, Andreas Koeberle Drug Discovery Unit, Cancer Research UK Manchester Institute, Wilmslow Road, Manchester, M20 4BX Friedrich-Schiller-University Jena, Philosophenweg 14, 07743 Jena

Background: LSD1 plays a key role in maintaining the balance between haematopoietic stem cell characteristics and differentiation to mature myeloid cells. In AML, LSD1 drives the oncogenic potential of leukaemic stem The membrane phospholipid arachidonyl-phosphatidylcholine (20:4-PC) oscillates during the cell cycle and cells through the formation of immature blast cells by switching off this differentiation programming. counteracts proliferation by suppressing Akt (protein kinase B) membrane binding1. Whether changes in the Mechanism-based inhibitors of LSD1, developed from the monoamine oxidase (MAO) inhibitor phospholipid composition are linked to Akt activation during apoptosis (programmed cell death) has not been tranylcypromine, have recently entered clinical trials. While the mechanism and inhibitory potential of these investigated so far. Thus, we induced apoptosis in mouse NIH-3T3 cells through different pathways and compounds are now well defined, the potential for effective reversible inhibitors of LSD1 as clinical agents is monitored the phospholipid composition by ultraperformance liquid chromatography-coupled ESI tandem mass less clear. spectrometry. More than 100 species of phosphatidylcholines, -ethanolamines, -serines, -inositols, -gycerols and Methods: Starting from existing literature and patent series, we employed rational medicinal chemistry and sphingomyelins were quantified. The total amount of phospholipid subclasses was either decreased or remained computational design using Cresset software to scaffold-hop into free IP space, while retaining activity against unaffected depending on apoptotic inducer and phospholipid subclass. Most remarkable was the time-dependent LSD1 in biochemical assays and by surface plasmon resonance (SPR). Selected compounds were tested in increase of polyunsaturated phospholipids (including 20:4-PC) relative to palmitoleate-containing species along cellular assays and evaluated for their physico-chemical properties in vitro and in vivo. with an opposing regulation of Akt. The initial enzyme of fatty acid biosynthesis acetyl-CoA carboxylase (ACC) was inhibited by phosphorylation during apoptosis, and its specific inhibition mimicked the apoptotic effects on Results: Several series of reversible LSD1 inhibitors have been designed and synthesised. These novel series phospholipid composition and kinase activation. We conclude that the decrease of fatty acid biosynthesis demonstrate a clear pharmacophore for effective inhibition. The most active series displays KD values of <10 nM associated with apoptosis evokes specific changes in the phospholipid profile, thereby suppressing Akt by SPR, and IC50 values of <300 nM in a CD86 cell assay. Compounds showed good metabolic stability in signalling, which is a convenient strategy for impairing cell survival. mouse hepatocytes, and despite displaying high efflux ratios were shown to be moderately bioavailable. Given the paucity of reversible and selective inhibitors of LSD1 available these compounds may be effective tools to drive further research into the use of LSD1 inhibitors as single agents, or in combination with other therapies. References 1) Koeberle et al. (2013) Arachidonoyl-phosphatidylcholine oscillates during the cell cycle and counteracts proliferation by suppressing Akt membrane binding. PNAS 110(7):2546-2551

100 P042 P043 DESIGN AND DEVELOPMENT OF POTENT SERIES OF IMPAIRED FATTY ACID BIOSYNTHESIS DURING APOPTOSIS REVERSIBLE INHIBITORS OF LYSINE SPECIFIC METHYLASE 1 DECREASES AKT PHOSPHORYLATION THROUGH SPECIFIC CHANGES IN THE CELLULAR PHOSPHOLIPID COMPOSITION Daniel Mould, Alison McGonagle, Allan Jordan, Donald Ogilvie Helmut Pein, Oliver Werz, Andreas Koeberle Drug Discovery Unit, Cancer Research UK Manchester Institute, Wilmslow Road, Manchester, M20 4BX Friedrich-Schiller-University Jena, Philosophenweg 14, 07743 Jena

Background: LSD1 plays a key role in maintaining the balance between haematopoietic stem cell characteristics and differentiation to mature myeloid cells. In AML, LSD1 drives the oncogenic potential of leukaemic stem The membrane phospholipid arachidonyl-phosphatidylcholine (20:4-PC) oscillates during the cell cycle and cells through the formation of immature blast cells by switching off this differentiation programming. counteracts proliferation by suppressing Akt (protein kinase B) membrane binding1. Whether changes in the Mechanism-based inhibitors of LSD1, developed from the monoamine oxidase (MAO) inhibitor phospholipid composition are linked to Akt activation during apoptosis (programmed cell death) has not been tranylcypromine, have recently entered clinical trials. While the mechanism and inhibitory potential of these investigated so far. Thus, we induced apoptosis in mouse NIH-3T3 cells through different pathways and compounds are now well defined, the potential for effective reversible inhibitors of LSD1 as clinical agents is monitored the phospholipid composition by ultraperformance liquid chromatography-coupled ESI tandem mass less clear. spectrometry. More than 100 species of phosphatidylcholines, -ethanolamines, -serines, -inositols, -gycerols and Methods: Starting from existing literature and patent series, we employed rational medicinal chemistry and sphingomyelins were quantified. The total amount of phospholipid subclasses was either decreased or remained computational design using Cresset software to scaffold-hop into free IP space, while retaining activity against unaffected depending on apoptotic inducer and phospholipid subclass. Most remarkable was the time-dependent LSD1 in biochemical assays and by surface plasmon resonance (SPR). Selected compounds were tested in increase of polyunsaturated phospholipids (including 20:4-PC) relative to palmitoleate-containing species along cellular assays and evaluated for their physico-chemical properties in vitro and in vivo. with an opposing regulation of Akt. The initial enzyme of fatty acid biosynthesis acetyl-CoA carboxylase (ACC) was inhibited by phosphorylation during apoptosis, and its specific inhibition mimicked the apoptotic effects on Results: Several series of reversible LSD1 inhibitors have been designed and synthesised. These novel series phospholipid composition and kinase activation. We conclude that the decrease of fatty acid biosynthesis demonstrate a clear pharmacophore for effective inhibition. The most active series displays KD values of <10 nM associated with apoptosis evokes specific changes in the phospholipid profile, thereby suppressing Akt by SPR, and IC50 values of <300 nM in a CD86 cell assay. Compounds showed good metabolic stability in signalling, which is a convenient strategy for impairing cell survival. mouse hepatocytes, and despite displaying high efflux ratios were shown to be moderately bioavailable. Given the paucity of reversible and selective inhibitors of LSD1 available these compounds may be effective tools to drive further research into the use of LSD1 inhibitors as single agents, or in combination with other therapies. References 1) Koeberle et al. (2013) Arachidonoyl-phosphatidylcholine oscillates during the cell cycle and counteracts proliferation by suppressing Akt membrane binding. PNAS 110(7):2546-2551

101 P044 P045 PRODRUGS SENSITIVE TO REACTIVE OXYGEN SPECIES FOR THE EXTENDED SAR EXPLORATION AND STRUCTURAL TREATMENT OF CHRONIC INFLAMMATORY DISEASES AND OPTIMIZATION OF A NOVEL CLASS OF QUINOLOXYACETAMIDE CANCER ANTIMYCOBACTERIAL COMPOUNDS

Jorge Peiro, Mads H. Clausen Eleni Pitta (1,2), Olga Balabon (1,2), Maciej Rogacki (1,2), Fraser Cunningham (2), Jurgen Joossens (1), Koen Augustyns (1), Pieter Van der Veken (1), Robert Bates (2) Department of Chemistry & Center for Nanomedicine and Theranostics, Technical University of Denmark, Kemitorvet 207, DK-2800 Kgs. Lyngby, Denmark 1) Medicinal chemistry laboratory, Department of Pharmaceutical Sciences, University of Antwerp, Universiteitsplein 1, B-2610 Wilrijk, Belgium 2) Diseases of the Developing World (DDW), Tres Cantos Medicines Development Campus (TCMDC), GlaxoSmithKline, Severo Ochoa 2, 28760 Tres Cantos, Spain

Insufficient drug selectivity causes severe side effects and host toxicity even in many of the first-line therapy treatments of diseases. Prodrugs are masked forms of pharmacologically active agents designed to undergo in vivo activation by specific stimuli. The use of prodrugs is generally proposed for the improvement of the ADME Tuberculosis (TB), caused by Mycobacterium tuberculosis, is a disease of antiquity with extraordinary and “drug-like” properties of compounds. However, an additional feature is their application in targeted drug endurance and complex clinical pathology. Within the last few decades, an increasing prevalence of delivery, i.e. the released of the bioactive molecule by disease-specific stimuli.1 Several pathologies, like cancer Multi-Drug-Resistant tuberculosis (MDR-TB) and the often fatal comorbidity between TB and human and chronic inflammatory diseases, are associated with increased levels of reactive oxygen species (ROS), due to immunodeficiency virus (HIV) has been observed. Therefore, the discovery of new chemical entities with novel the generation of inflammation.2 This unique environment at the inflammatory tissue can therefore be used as a modes of action is a high priority in the global health agenda. trigger stimulus. In this work we propose the use of prodrugs for the reduction of the undesired effects of drugs prescribed for inflammation related diseases. Making them inactive until they get activated predominantly or We present a novel series of quinoloxyacetamide derivatives with antimycobacterial activity that was identified exclusively in inflammatory tissue was the strategy suggested (see Fig.1). through a whole cell High Throughput Screening (HTS) performed by GSK. SAR exploration of the primary hit led to micromolar potencies and improved physicochemical profiles. A metabolic stability study of the series In order to achieve this goal, a series of promoieties sensitive to ROS were developed and synthesized. They was performed and an amide bond was found to be responsible for the blood instability. Further medicinal were then coupled to different existing drugs and investigated. Promising results on the stability of the chemistry efforts to replace this labile group, led to a derivative which opened new opportunities for the series. compounds in different physiological conditions, good ADME properties, activation at different ROS This compound did not exhibit any cytotoxic effects or hERG inhibition. concentrations and comparable activity to the parent drug in cell-based assays were obtained. This indicates that the prodrug strategy is a promising tool for the improvement of current therapies for inflammatory diseases Alkylation of 4-hydroxy azaheterocycle precursors was used as a key synthetic step in the preparation of most associated with serious side effects. This project presents a unique site-selective prodrug strategy based on ROS presented compounds. The N- versus O- chemoselectivity of this reaction was strongly dependent on the nature activation suitable for a wide range of diseases and different marketed drugs. of the azaheterocycle and its substitution pattern, highlighting the need for full structural assignment. Further optimization of this promising series of novel antimycobacterial compounds together with target identification,

could provide a strong lead compound in the drug discovery arena.

References 1) Nat. Rev. Drug Discov. 2008, 7, 255-270. 2) Antioxid. Redox Signal. 2014, 20, 1126–1167

102 P044 P045 PRODRUGS SENSITIVE TO REACTIVE OXYGEN SPECIES FOR THE EXTENDED SAR EXPLORATION AND STRUCTURAL TREATMENT OF CHRONIC INFLAMMATORY DISEASES AND OPTIMIZATION OF A NOVEL CLASS OF QUINOLOXYACETAMIDE CANCER ANTIMYCOBACTERIAL COMPOUNDS

Jorge Peiro, Mads H. Clausen Eleni Pitta (1,2), Olga Balabon (1,2), Maciej Rogacki (1,2), Fraser Cunningham (2), Jurgen Joossens (1), Koen Augustyns (1), Pieter Van der Veken (1), Robert Bates (2) Department of Chemistry & Center for Nanomedicine and Theranostics, Technical University of Denmark, Kemitorvet 207, DK-2800 Kgs. Lyngby, Denmark 1) Medicinal chemistry laboratory, Department of Pharmaceutical Sciences, University of Antwerp, Universiteitsplein 1, B-2610 Wilrijk, Belgium 2) Diseases of the Developing World (DDW), Tres Cantos Medicines Development Campus (TCMDC), GlaxoSmithKline, Severo Ochoa 2, 28760 Tres Cantos, Spain

Insufficient drug selectivity causes severe side effects and host toxicity even in many of the first-line therapy treatments of diseases. Prodrugs are masked forms of pharmacologically active agents designed to undergo in vivo activation by specific stimuli. The use of prodrugs is generally proposed for the improvement of the ADME Tuberculosis (TB), caused by Mycobacterium tuberculosis, is a disease of antiquity with extraordinary and “drug-like” properties of compounds. However, an additional feature is their application in targeted drug endurance and complex clinical pathology. Within the last few decades, an increasing prevalence of delivery, i.e. the released of the bioactive molecule by disease-specific stimuli.1 Several pathologies, like cancer Multi-Drug-Resistant tuberculosis (MDR-TB) and the often fatal comorbidity between TB and human and chronic inflammatory diseases, are associated with increased levels of reactive oxygen species (ROS), due to immunodeficiency virus (HIV) has been observed. Therefore, the discovery of new chemical entities with novel the generation of inflammation.2 This unique environment at the inflammatory tissue can therefore be used as a modes of action is a high priority in the global health agenda. trigger stimulus. In this work we propose the use of prodrugs for the reduction of the undesired effects of drugs prescribed for inflammation related diseases. Making them inactive until they get activated predominantly or We present a novel series of quinoloxyacetamide derivatives with antimycobacterial activity that was identified exclusively in inflammatory tissue was the strategy suggested (see Fig.1). through a whole cell High Throughput Screening (HTS) performed by GSK. SAR exploration of the primary hit led to micromolar potencies and improved physicochemical profiles. A metabolic stability study of the series In order to achieve this goal, a series of promoieties sensitive to ROS were developed and synthesized. They was performed and an amide bond was found to be responsible for the blood instability. Further medicinal were then coupled to different existing drugs and investigated. Promising results on the stability of the chemistry efforts to replace this labile group, led to a derivative which opened new opportunities for the series. compounds in different physiological conditions, good ADME properties, activation at different ROS This compound did not exhibit any cytotoxic effects or hERG inhibition. concentrations and comparable activity to the parent drug in cell-based assays were obtained. This indicates that the prodrug strategy is a promising tool for the improvement of current therapies for inflammatory diseases Alkylation of 4-hydroxy azaheterocycle precursors was used as a key synthetic step in the preparation of most associated with serious side effects. This project presents a unique site-selective prodrug strategy based on ROS presented compounds. The N- versus O- chemoselectivity of this reaction was strongly dependent on the nature activation suitable for a wide range of diseases and different marketed drugs. of the azaheterocycle and its substitution pattern, highlighting the need for full structural assignment. Further optimization of this promising series of novel antimycobacterial compounds together with target identification,

could provide a strong lead compound in the drug discovery arena.

References 1) Nat. Rev. Drug Discov. 2008, 7, 255-270. 2) Antioxid. Redox Signal. 2014, 20, 1126–1167

103 P046 P047 COMBINING ANTIOXIDANT, ANTIINFLAMMATORY & INVESTIGATION OF THE BINDING MODE OF A NOVEL CLASS OF HYPOLIPIDEMIC ACTIVITY BY DESIGN: NEW MULTI-POTENT ANTIMYCOBACTERIAL DPRE1 INHIBITORS MORPHOLINE DERIVATIVES FOR ATHEROSCLEROSIS Maciej K. Rogacki (1,2), Olga Balabon (1,2), Eleni Pitta (1,2), Fraser Cunningham (2), Dries Van Rompaey (1), Jurgen Joossens (1), Koen Augustyns (1), Hans De Winter (1), Robert Bates (2), Pieter Van Maria Giasafaki, Maria Potsi, Alexios Matralis, Angeliki Kourounakis der Veken (1) Department of Medicinal Chemistry, School of Pharmacy, University of Athens 15771 Athens, Greece. *[email protected] 1) Laboratory of Medicinal Chemistry, Department of Pharmaceutical Sciences, University of Antwerp, Universiteitsplein 1, 2610 Antwerp, Belgium 2) Diseases of the Developing World (DDW), Tres Cantos Medicines Development Campus (TCMDC), GlaxoSmithKline, Severo Ochoa 2, 28760 Tres Cantos, Spain

Multifunctional agents that address two or more targets of a multifactorial disease, can be expected to produce superior in vivo effects as compared to higher-affinity single-targeted compounds.1,2,3 Based on the pathophysiological mechanisms involved in atherosclerosis (inflammation/hyperlipidemia/oxidative stress), we Tuberculosis is an infectious disease caused by Mycobacterium tuberculosis. With almost 9 million new cases designed new aromatic thio/morpholine derivatives (1-4) by combining several different anti-inflammatory and diagnosed each year and a TB-related death occurring every 20 seconds, the disease is considered a significant antioxidant pharmacophores. threat to global health. In spite of decades of research, the first-line antitubercular therapy still consists of medicines developed more than 40 years ago. With the increasing prevalence of the drug resistant strains of M. tuberculosis, there is a growing and acute need for new, improved antitubercular agents with novel modes of action. Over the past few years increasing interest has been drawn to a new mycobacterial protein - decaprenylphospho-beta-D-ribofuranose 2-oxidase (DprE1). This periplasmic enzyme catalyzes a key step in the biosynthesis of the cell wall components and is potentially an attractive target for future antitubercular drugs. In search of DprE1 inhibitors, a high-throughput screening campaign was performed by GlaxoSmithKline, leading to identification of a novel compound family, based on the imidazolidine-2,4-dione scaffold. Within the framework of the OpenMedChem project – a collaboration between GlaxoSmithKline and the University of Antwerp, structure-activity relationship (SAR) exploration and compound profiling of the discovered hits was initiated. Based on the initial SAR information, the binding mode of the studied compounds These novel compounds, combining antioxidant, antihyperlipidemic and anti-inflammatory activities by design, to DprE1 was investigated by molecular docking, with the use of the MOE and the Autodock/Vina software. exhibited increased in vitro activities against oxidative stress (protection against lipid peroxidation) and Herein we present the results of this analysis and its preliminary confirmation through subsequent design, inflammation (lipoxygenase inhibition). This activity profile was shown to be extended in vivo a) in a synthesis and biological evaluation of new analogues of the active hits. hyperlipidemic mouse model where cholesterol/triglyceride levels were reduced up to 70% and plasma total antioxidant capacity (TAC) increased 2-fold and b) in an inflammatory mouse model (carrageenan-induced mouse paw edema) where compounds exhibited more potent anti-inflammatory action than parent or reference molecules. Based on these results, it appears that the incorporation of the specific antioxidant/anti-inflammatory pharmacophores, into the aromatic thio/morpholine lead structure, enhanced and broadened the pharmacological profile of the new compounds that may find successful therapeutic applications in atherosclerosis and metabolic syndrome disorders.

References 1) M. Katselou, A. Matralis and A. Kourounakis* Curr. Med. Chem., 2014 2) A. Matralis and A. Kourounakis* J. Med. Chem., 2014. 3) E. Ladopoulou and A. Kourounakis* Bioorg.Med.Chem., 2015.

104 P046 P047 COMBINING ANTIOXIDANT, ANTIINFLAMMATORY & INVESTIGATION OF THE BINDING MODE OF A NOVEL CLASS OF HYPOLIPIDEMIC ACTIVITY BY DESIGN: NEW MULTI-POTENT ANTIMYCOBACTERIAL DPRE1 INHIBITORS MORPHOLINE DERIVATIVES FOR ATHEROSCLEROSIS Maciej K. Rogacki (1,2), Olga Balabon (1,2), Eleni Pitta (1,2), Fraser Cunningham (2), Dries Van Rompaey (1), Jurgen Joossens (1), Koen Augustyns (1), Hans De Winter (1), Robert Bates (2), Pieter Van Maria Giasafaki, Maria Potsi, Alexios Matralis, Angeliki Kourounakis der Veken (1) Department of Medicinal Chemistry, School of Pharmacy, University of Athens 15771 Athens, Greece. *[email protected] 1) Laboratory of Medicinal Chemistry, Department of Pharmaceutical Sciences, University of Antwerp, Universiteitsplein 1, 2610 Antwerp, Belgium 2) Diseases of the Developing World (DDW), Tres Cantos Medicines Development Campus (TCMDC), GlaxoSmithKline, Severo Ochoa 2, 28760 Tres Cantos, Spain

Multifunctional agents that address two or more targets of a multifactorial disease, can be expected to produce superior in vivo effects as compared to higher-affinity single-targeted compounds.1,2,3 Based on the pathophysiological mechanisms involved in atherosclerosis (inflammation/hyperlipidemia/oxidative stress), we Tuberculosis is an infectious disease caused by Mycobacterium tuberculosis. With almost 9 million new cases designed new aromatic thio/morpholine derivatives (1-4) by combining several different anti-inflammatory and diagnosed each year and a TB-related death occurring every 20 seconds, the disease is considered a significant antioxidant pharmacophores. threat to global health. In spite of decades of research, the first-line antitubercular therapy still consists of medicines developed more than 40 years ago. With the increasing prevalence of the drug resistant strains of M. tuberculosis, there is a growing and acute need for new, improved antitubercular agents with novel modes of action. Over the past few years increasing interest has been drawn to a new mycobacterial protein - decaprenylphospho-beta-D-ribofuranose 2-oxidase (DprE1). This periplasmic enzyme catalyzes a key step in the biosynthesis of the cell wall components and is potentially an attractive target for future antitubercular drugs. In search of DprE1 inhibitors, a high-throughput screening campaign was performed by GlaxoSmithKline, leading to identification of a novel compound family, based on the imidazolidine-2,4-dione scaffold. Within the framework of the OpenMedChem project – a collaboration between GlaxoSmithKline and the University of Antwerp, structure-activity relationship (SAR) exploration and compound profiling of the discovered hits was initiated. Based on the initial SAR information, the binding mode of the studied compounds These novel compounds, combining antioxidant, antihyperlipidemic and anti-inflammatory activities by design, to DprE1 was investigated by molecular docking, with the use of the MOE and the Autodock/Vina software. exhibited increased in vitro activities against oxidative stress (protection against lipid peroxidation) and Herein we present the results of this analysis and its preliminary confirmation through subsequent design, inflammation (lipoxygenase inhibition). This activity profile was shown to be extended in vivo a) in a synthesis and biological evaluation of new analogues of the active hits. hyperlipidemic mouse model where cholesterol/triglyceride levels were reduced up to 70% and plasma total antioxidant capacity (TAC) increased 2-fold and b) in an inflammatory mouse model (carrageenan-induced mouse paw edema) where compounds exhibited more potent anti-inflammatory action than parent or reference molecules. Based on these results, it appears that the incorporation of the specific antioxidant/anti-inflammatory pharmacophores, into the aromatic thio/morpholine lead structure, enhanced and broadened the pharmacological profile of the new compounds that may find successful therapeutic applications in atherosclerosis and metabolic syndrome disorders.

References 1) M. Katselou, A. Matralis and A. Kourounakis* Curr. Med. Chem., 2014 2) A. Matralis and A. Kourounakis* J. Med. Chem., 2014. 3) E. Ladopoulou and A. Kourounakis* Bioorg.Med.Chem., 2015.

105 P049 P050 HIGH-THROUGHPUT SCREENING FOR INHIBITORS OF POTENTIAL SYNTHETIC APPROACHES FOR PSEUDOMONAS AERUGINOSA ADP-RIBOSYLATING EXOENZYME AZOLO[5,1-b]PURINES S Konstantin Savateev, Victor Fedotov, Evgeny Ulomsky, Vladimir Rusinov Michael Saleeb, Sara Spjut, Mahsa Ebrahimi, Charlotta Sundin, Åke Forsberg, Herwig Schüler, Mikael Elofsson Ural Federal University named after the first President of Russia Boris Yeltsin, department of Organic chemistry, Ekaterinburg, street Mira, 19, Ekaterinburg, ZIP620002, Russian Federation Е-mail: [email protected]

Pseudomonas aeruginosa is a gram-negative bacterium and one of the leading pathogens behind Triazolo[5,1-b]purines are least widespread structural analogues of natural nucleosides and nucleic bases of hospital-acquired infections. It causes a wide variety of life-threating infections including blood, pneumonia, and purine series. At the same time, known representatives of azolopurines exhibit a wide spectrum of antiviral wounds infections, especially in immunocompromised patients such as cystic fibrosis and cancer patients. Due to activity, potency against rheumatoid arthritis, psoriasis, Alzheimer’s and Parkinson’s diseases, etc [1]. Despite the remarkable antibiotic resistance of P. aeruginosa there is a need to develop novel antipseudomonal agents the practical value, azolo[b]purines are very stingily represented in the chemical references due to their synthesis that can be used in combination therapy to ensure treatment of resistant strains. Targeting bacterial virulence, the complexity. ability of the bacteria to cause disease, is an attractive approach to combat antibiotic resistance by ‘disarming’ the bacteria instead of killing it. The type III secretion system (T3SS) is an essential virulence factor and a valid We have proposed a synthesis strategy of purine precursors (2) based on the simple reagents such as therapeutic target in many gram-negative bacteria including P. aeruginosa. It is a syringe-like apparatus located amino-azoles (1) according to the following retroscheme. on the bacterial surface and responsible for transporting effectors proteins into eukaryotic cells. The effector proteins subvert the host cell and create a niche that allows bacterial growth. Exotoxins S and T (ExoS and ExoT) are toxins that are secreted by the P. aeruginosa T3SS. They are ADP-ribosyltransferase (ADPRT) enzymes that modify various eukaryotic proteins, such as small GTPase proteins e.g. Ras, Rho and Rac, which leads to signal-transduction malfunction and eventually cell death. Mutation of exoS at the ADPRT domain attenuates virulence [1, 2] and makes ExoS-ADPRT a putative Firstly, we tried to synthesise diaminoderivatives throught 5,6-dihalogenheterocycles. Synthesis of therapeutic target. aminohaloazolopyrimidines (5) was carried out with good yields, but further halogen substitution could not be realized even in rigorous conditions.

Reduction of arylazo-, nitro- or nitrozo-group is also used to produce amino fragment in 1,2,4-triazolo[1,5-a]pyrimidines. Chlorodeoxygenation of triazolopyrimidines (6) was used to obtain chloroderivatives (7) which were highly unstable and were converted into nitroaminotriazolopyrimidines (8) without isolation with poor yields.

Reaction of 6-arylazotriazolopyrimidines (9) with phosphoryl chloride in acetonitrile and subsequent treatment of intermediates (10) with primary amines gave desired products (11) in satisfactory yields.

For that, we developed an in vitro enzymatic assay (Figure 1) and identified an inhibitor of ExoS-ADPRT activity [3]. Recently, we optimized and employed the assay for a high-throughput screen of a library of 30,000 Our first attempts of azo-group reduction into amino-group in substrate (11b) were unsuccessful. However, we diverse small-molecules at 10 µM. The screen identified 80 potential ExoS ADPRT inhibitors. Hit validation anticipate that the strategies described herein will pave the way for the diaminotriazolo[1,5-a]pyrimidines as furnished 6 inhibitors of ExoS-ADPRT with an IC50 of 3 – 27 µM. Ongoing enzyme kinetics and medicinal intermediates in the synthesis of biologically active triazolo[5,1-b]purines. chemistry programs with the goal to reach compounds with efficacy in vivo will eventually allow us to support the scientific community with a novel chemical probe(s), which can be used to study bacterial virulence in vitro We thank the Russian Federation Ministry of Education (project 2458) and the Russian Foundation for Basic and in vivo. Research grant № 16-33-00159 mol_a.

References References 1) Rangel, S.M., et al. mBio, 2014. 1) WO 2005/011609 A2, 10.02.2005. 2) Sun, Y., et al., The Journal of Immunology, 2012. 188(4): p. 1884-1895 3) Pinto, A.F., et al., Journal of Biomolecular Screening, 2016

106 P049 P050 HIGH-THROUGHPUT SCREENING FOR INHIBITORS OF POTENTIAL SYNTHETIC APPROACHES FOR PSEUDOMONAS AERUGINOSA ADP-RIBOSYLATING EXOENZYME AZOLO[5,1-b]PURINES S Konstantin Savateev, Victor Fedotov, Evgeny Ulomsky, Vladimir Rusinov Michael Saleeb, Sara Spjut, Mahsa Ebrahimi, Charlotta Sundin, Åke Forsberg, Herwig Schüler, Mikael Elofsson Ural Federal University named after the first President of Russia Boris Yeltsin, department of Organic chemistry, Ekaterinburg, street Mira, 19, Ekaterinburg, ZIP620002, Russian Federation Е-mail: [email protected]

Pseudomonas aeruginosa is a gram-negative bacterium and one of the leading pathogens behind Triazolo[5,1-b]purines are least widespread structural analogues of natural nucleosides and nucleic bases of hospital-acquired infections. It causes a wide variety of life-threating infections including blood, pneumonia, and purine series. At the same time, known representatives of azolopurines exhibit a wide spectrum of antiviral wounds infections, especially in immunocompromised patients such as cystic fibrosis and cancer patients. Due to activity, potency against rheumatoid arthritis, psoriasis, Alzheimer’s and Parkinson’s diseases, etc [1]. Despite the remarkable antibiotic resistance of P. aeruginosa there is a need to develop novel antipseudomonal agents the practical value, azolo[b]purines are very stingily represented in the chemical references due to their synthesis that can be used in combination therapy to ensure treatment of resistant strains. Targeting bacterial virulence, the complexity. ability of the bacteria to cause disease, is an attractive approach to combat antibiotic resistance by ‘disarming’ the bacteria instead of killing it. The type III secretion system (T3SS) is an essential virulence factor and a valid We have proposed a synthesis strategy of purine precursors (2) based on the simple reagents such as therapeutic target in many gram-negative bacteria including P. aeruginosa. It is a syringe-like apparatus located amino-azoles (1) according to the following retroscheme. on the bacterial surface and responsible for transporting effectors proteins into eukaryotic cells. The effector proteins subvert the host cell and create a niche that allows bacterial growth. Exotoxins S and T (ExoS and ExoT) are toxins that are secreted by the P. aeruginosa T3SS. They are ADP-ribosyltransferase (ADPRT) enzymes that modify various eukaryotic proteins, such as small GTPase proteins e.g. Ras, Rho and Rac, which leads to signal-transduction malfunction and eventually cell death. Mutation of exoS at the ADPRT domain attenuates virulence [1, 2] and makes ExoS-ADPRT a putative Firstly, we tried to synthesise diaminoderivatives throught 5,6-dihalogenheterocycles. Synthesis of therapeutic target. aminohaloazolopyrimidines (5) was carried out with good yields, but further halogen substitution could not be realized even in rigorous conditions.

Reduction of arylazo-, nitro- or nitrozo-group is also used to produce amino fragment in 1,2,4-triazolo[1,5-a]pyrimidines. Chlorodeoxygenation of triazolopyrimidines (6) was used to obtain chloroderivatives (7) which were highly unstable and were converted into nitroaminotriazolopyrimidines (8) without isolation with poor yields.

Reaction of 6-arylazotriazolopyrimidines (9) with phosphoryl chloride in acetonitrile and subsequent treatment of intermediates (10) with primary amines gave desired products (11) in satisfactory yields.

For that, we developed an in vitro enzymatic assay (Figure 1) and identified an inhibitor of ExoS-ADPRT activity [3]. Recently, we optimized and employed the assay for a high-throughput screen of a library of 30,000 Our first attempts of azo-group reduction into amino-group in substrate (11b) were unsuccessful. However, we diverse small-molecules at 10 µM. The screen identified 80 potential ExoS ADPRT inhibitors. Hit validation anticipate that the strategies described herein will pave the way for the diaminotriazolo[1,5-a]pyrimidines as furnished 6 inhibitors of ExoS-ADPRT with an IC50 of 3 – 27 µM. Ongoing enzyme kinetics and medicinal intermediates in the synthesis of biologically active triazolo[5,1-b]purines. chemistry programs with the goal to reach compounds with efficacy in vivo will eventually allow us to support the scientific community with a novel chemical probe(s), which can be used to study bacterial virulence in vitro We thank the Russian Federation Ministry of Education (project 2458) and the Russian Foundation for Basic and in vivo. Research grant № 16-33-00159 mol_a.

References References 1) Rangel, S.M., et al. mBio, 2014. 1) WO 2005/011609 A2, 10.02.2005. 2) Sun, Y., et al., The Journal of Immunology, 2012. 188(4): p. 1884-1895 3) Pinto, A.F., et al., Journal of Biomolecular Screening, 2016

107 P051 P052 SYNTHESIS OF NOVEL TOWARDS NONNATURAL CARBOHYDRATE-BASED INHIBITORS TETRAHYDRO-4-ARYL-PYRIDO[1,2-C]PYRIMIDINE DERIVATIVES OF PSEUDOMONAS AERUGINOSA VIRULENCE FACTOR LECTIN WITH DUAL SERT AND 5-HT1A ACTIVITY LECB

Grzegorz Ślifirski, Marek Król, Franciszek Herold, Jadwiga Turło Roman Sommer (1,2,3), Annabelle Varrot (4), Anna Hofmann (3,5), Dirk Hauck (1,2,3), Julia Stife (3), Inigo Göttker-Schnetmann (3), Stefanie Wagner (1,2), Aymeric Audfray (4), Andreas Prestel (3,6), Heiko Department of Drug Technology and Pharmaceutical Biotechnology, M. Möller (3,6), Thomas E. Exner (3), Anne Imberty (4), Alexander Titz (1,2,3) Faculty of Pharmacy, Medical University of Warsaw 1, Banacha Str., 02-097 Warsaw, Poland 1) Chemical Biology of Carbohydrates, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), 66123 Saarbrücken, Germany 2) Deutsches Zentrum für Infektionsforschung (DZIF), Standort Hannover-Braunschweig, Germany 3) Department of Chemistry and Graduate School Chemical Biology University of Konstanz, 78457 Konstanz, Germany 4) Centre de Recherche sur les Macromolécules Végétales (CERMAV-UPR5301) CNRS and Université Grenoble Alpes, Our earlier research resulted in the synthesis of a series of pyrido[1,2-c]pyrimidine derivatives, characterized by BP53, 38041 Grenoble cedex 9, France a double mechanism of action: the inhibition of 5-HT reuptake and agonistic activity towards both pre- and 5) Institute of Pharmacy and Food Chemistry, University of Würzburg, 97074 Würzburg, Germany 6) Institute of Chemistry, University of Potsdam, 14476 Potsdam, Germany postsynaptic 5-HT1A receptors1-5. These compounds can be considered a good entry point for novel drug candidates. The aim of our present study was to synthesize and assess the biological properties of new tetrahydro-pyrido[1,2-c]pyrimidine derivatives with dual activity. New ligands were obtained after the modification of previously synthesized pyrido[1,2-c]pyrimidine derivatives with 3-(piperidin-3-yl)-1H-indole residue in the pharmacophore element. Modifications were performed in the terminal portion, where the The rise of resistance against antibiotics in bacteria is a major threat and demands the development of novel pirydo-part of the pyrido[1,2c]pyrimidine ring system was saturated. antibacterial therapies. Infections with Pseudomonas aeruginosa, an opportunistic pathogen, are a severe problem for hospitalized patients and for patients suffering from cystic fibrosis. These bacteria can form biofilms Combining a selective serotonin reuptake inhibitor with the 5-HT1A autoreceptors antagonist was proposed in and thereby increase their resistance towards antibiotics through the physical barrier of the biofilm matrix. The the 1990s as a therapeutic strategy leading to the faster onset of antidepressant action and the greater efficacy of Pseudomonas 6 virulence factor lectin LecB is a carbohydrate-binding adhesin and plays an important role in treatment . Several clinical trials proved that the coadministration of pindolol (5-HT1A autoreceptor antagonist) biofilm formation.[1] The natural ligands for LecB are glycosides of D-mannose (e.g., methyl α-D-mannoside (1 may shorten the latency period of paroxetine and (well-known SSRIs). Concerns about simultaneous, 2 7 )) and L-fucose (e.g., methyl α-L- fucoside ( )), the latter displaying an unusual strong affinity to its lectin unfavourable in the view of antidepressant activity, non-selective blocking of the postsynaptic receptors receptor. This fact was explained by the interaction of the carbohydrate with two calcium ions bound by the generated the concept of combining SSRI activity with the 5-HT1A agonism as another promising strategy for receptor and an additional lipophilic interaction of the terminal methyl group in fucose.[2] Interestingly, although potential new antidepressants development. The FDA registration of vilazodone (Viibryd) in 2011 and 8,9 mannosides are much weaker ligands for LecB, their inhibitory potency was remarkably improved upon vortioxetine (Brintellix) in 2013, as dual-acting antidepressants confirmed the validity of this approach . modification of the equatorial C-6 substituent.[3] The new 4-aryl-pyrido[1,2-c]pyrimidine derivatives were obtained by way of a multi-step chemical synthesis and subjected to analytical studies, using the methods of 1H NMR and 13C NMR spectroscopy as well as HRMS. The pharmacological profile of the obtained compounds was assessed in radioligands binding assays (5-HT1A, SERT). In vivo functional studies will be conducted in the Institute of Pharmacology, the Polish Academy of Sciences, and the Department of Pharmacobiology of the Jagiellonian University Medical College in Kraków; metabolic stability evaluation will be performed in the Department of Pharmaceutical Chemistry, Medical University of Gdańsk. The results of in vitro and in vivo studies will allow us to draw conclusions regarding structure-activity relationship in the tested group of compounds and to select compounds for further pre-clinical evaluation. We acknowledge the financial support of the Polish National Science Center grant, OPUS 6, No. Figure 1: Methyl α-D-mannoside (1) and methyl α-L-fucoside (2) are the carbohydrate epitopes recognized by UMO-2013/11/B/NZ7/01638. the Pseudomonas aeruginosa lectin LecB. Several aspects should be taken into account in development of carbohydrate-derived drugs. These are unspecific References binding to the human pathogen-recognition receptors (PRRs), low oral bioavailability and metabolic instability 1) Herold, F. et al. Novel 4-aryl-pyrido[1,2-c]pyrimidines with dual SSRI and 5-HT1A activity: Part 2. Eur. J. Med. Chem. in human and pathogen. In order to mask the natural carbohydrate character of LecB inhibitors and to increase 44, 4702–4715 (2009). their affinities, a detailed structure‑activity relationship study was performed.[4, 5, 6] Compared to unmodified 2) Herold, F. et al. Novel 4-aryl-pyrido[1,2-c]pyrimidines with dual SSRI and 5-HT1A activity, Part 1. Eur. J. Med. Chem. 1 44, 1710–1717 (2009). carbohydrates, methyl α-D-mannoside ( ) binding properties of LecB ligands were improved regarding their 3) Herold, F. et al. Novel 4-aryl-pyrido[1,2-c]pyrimidines with dual SSRI and 5-HT1A activity. part 3. Eur. J. Med. Chem. potency and complex stability. Herein discovered LecB inhibitors are supposed to overcome drawbacks of the 46, 142–149 (2011). natural carbohydrates as a drug and are a good base for future drug development in the anti-virulence approach. 4) Chodkowski, A. et al. Novel 4-aryl-pyrido[1,2-c]pyrimidines with dual SSRI and 5-HT1A activity. Part 4. Eur. J. Med. Chem. 90, 21–32 (2015). 5) Gomolka, A. et al. Novel 4-aryl-pyrido[1,2-c]pyrimidines with dual SSRI and 5-HT1A activity. part 5. Eur. J. Med. Chem. References 98, 221–236 (2015). 6) Artigas, F. Serotonin receptors involved in antidepressant effects. Pharmacol. Ther. 137, 119–131 (2013). 1) Tielker D, Hacker S, Loris R, Strathmann M, Wingender J, Wilhelm S, Rosenau F, Jaeger K. Microbiology. 2005, 151(Pt 7) Artigas, F., Adell, A. & Celada, P. Pindolol Augmentation of Antidepressant Response. Curr. Drug Targets 7, 139–147 5), 1313-23. (2006). 2) Mitchell E, Houles C, Sudakevitz D, Wimmerova M, Gautier C, Pérez S, Wu AM, Gilboa-Garber N, Imberty A. Nat 8) Dawson, L. A. & Watson, J. M. Vilazodone: A 5-HT1A receptor agonist/serotonin transporter inhibitor for the treatment Struct Biol 2002, 9(12), 918-21. of affective disorders. CNS Neurosci. Ther. 15, 107–117 (2009). 3) Hauck D, Joachim J, Frommeyer B, Varrot A, Philipp B, Möller H, Imberty A, Exner T, Titz A. ACS Chem. Biol. 2013, 8, 9) Sanchez, C., Asin, K. E. & Artigas, F. Vortioxetine, a novel antidepressant with multimodal activity: Review of preclinical 1775-84. and clinical data. Pharmacol. Ther. 145, 43–57 (2015). 4) Sommer R, Exner T E, Titz A. PLoS One 2014, 9, e112822. 5) Hofmann A, Sommer R, Hauck D, Stifel J, Gottker-Schnetmann I, Titz A. Carbohydrate Research 2015, 412, 34-42. 6) Sommer R, Hauck D, Varrot A, Wagner S, Audfray A, Prestel A, Möller H M, Imberty A, Titz A. ChemistryOpen 2015, 4, 756-767.

108 P051 P052 SYNTHESIS OF NOVEL TOWARDS NONNATURAL CARBOHYDRATE-BASED INHIBITORS TETRAHYDRO-4-ARYL-PYRIDO[1,2-C]PYRIMIDINE DERIVATIVES OF PSEUDOMONAS AERUGINOSA VIRULENCE FACTOR LECTIN WITH DUAL SERT AND 5-HT1A ACTIVITY LECB

Grzegorz Ślifirski, Marek Król, Franciszek Herold, Jadwiga Turło Roman Sommer (1,2,3), Annabelle Varrot (4), Anna Hofmann (3,5), Dirk Hauck (1,2,3), Julia Stife (3), Inigo Göttker-Schnetmann (3), Stefanie Wagner (1,2), Aymeric Audfray (4), Andreas Prestel (3,6), Heiko Department of Drug Technology and Pharmaceutical Biotechnology, M. Möller (3,6), Thomas E. Exner (3), Anne Imberty (4), Alexander Titz (1,2,3) Faculty of Pharmacy, Medical University of Warsaw 1, Banacha Str., 02-097 Warsaw, Poland 1) Chemical Biology of Carbohydrates, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), 66123 Saarbrücken, Germany 2) Deutsches Zentrum für Infektionsforschung (DZIF), Standort Hannover-Braunschweig, Germany 3) Department of Chemistry and Graduate School Chemical Biology University of Konstanz, 78457 Konstanz, Germany 4) Centre de Recherche sur les Macromolécules Végétales (CERMAV-UPR5301) CNRS and Université Grenoble Alpes, Our earlier research resulted in the synthesis of a series of pyrido[1,2-c]pyrimidine derivatives, characterized by BP53, 38041 Grenoble cedex 9, France a double mechanism of action: the inhibition of 5-HT reuptake and agonistic activity towards both pre- and 5) Institute of Pharmacy and Food Chemistry, University of Würzburg, 97074 Würzburg, Germany 6) Institute of Chemistry, University of Potsdam, 14476 Potsdam, Germany postsynaptic 5-HT1A receptors1-5. These compounds can be considered a good entry point for novel drug candidates. The aim of our present study was to synthesize and assess the biological properties of new tetrahydro-pyrido[1,2-c]pyrimidine derivatives with dual activity. New ligands were obtained after the modification of previously synthesized pyrido[1,2-c]pyrimidine derivatives with 3-(piperidin-3-yl)-1H-indole residue in the pharmacophore element. Modifications were performed in the terminal portion, where the The rise of resistance against antibiotics in bacteria is a major threat and demands the development of novel pirydo-part of the pyrido[1,2c]pyrimidine ring system was saturated. antibacterial therapies. Infections with Pseudomonas aeruginosa, an opportunistic pathogen, are a severe problem for hospitalized patients and for patients suffering from cystic fibrosis. These bacteria can form biofilms Combining a selective serotonin reuptake inhibitor with the 5-HT1A autoreceptors antagonist was proposed in and thereby increase their resistance towards antibiotics through the physical barrier of the biofilm matrix. The the 1990s as a therapeutic strategy leading to the faster onset of antidepressant action and the greater efficacy of Pseudomonas 6 virulence factor lectin LecB is a carbohydrate-binding adhesin and plays an important role in treatment . Several clinical trials proved that the coadministration of pindolol (5-HT1A autoreceptor antagonist) biofilm formation.[1] The natural ligands for LecB are glycosides of D-mannose (e.g., methyl α-D-mannoside (1 may shorten the latency period of paroxetine and fluoxetine (well-known SSRIs). Concerns about simultaneous, 2 7 )) and L-fucose (e.g., methyl α-L- fucoside ( )), the latter displaying an unusual strong affinity to its lectin unfavourable in the view of antidepressant activity, non-selective blocking of the postsynaptic receptors receptor. This fact was explained by the interaction of the carbohydrate with two calcium ions bound by the generated the concept of combining SSRI activity with the 5-HT1A agonism as another promising strategy for receptor and an additional lipophilic interaction of the terminal methyl group in fucose.[2] Interestingly, although potential new antidepressants development. The FDA registration of vilazodone (Viibryd) in 2011 and 8,9 mannosides are much weaker ligands for LecB, their inhibitory potency was remarkably improved upon vortioxetine (Brintellix) in 2013, as dual-acting antidepressants confirmed the validity of this approach . modification of the equatorial C-6 substituent.[3] The new 4-aryl-pyrido[1,2-c]pyrimidine derivatives were obtained by way of a multi-step chemical synthesis and subjected to analytical studies, using the methods of 1H NMR and 13C NMR spectroscopy as well as HRMS. The pharmacological profile of the obtained compounds was assessed in radioligands binding assays (5-HT1A, SERT). In vivo functional studies will be conducted in the Institute of Pharmacology, the Polish Academy of Sciences, and the Department of Pharmacobiology of the Jagiellonian University Medical College in Kraków; metabolic stability evaluation will be performed in the Department of Pharmaceutical Chemistry, Medical University of Gdańsk. The results of in vitro and in vivo studies will allow us to draw conclusions regarding structure-activity relationship in the tested group of compounds and to select compounds for further pre-clinical evaluation. We acknowledge the financial support of the Polish National Science Center grant, OPUS 6, No. Figure 1: Methyl α-D-mannoside (1) and methyl α-L-fucoside (2) are the carbohydrate epitopes recognized by UMO-2013/11/B/NZ7/01638. the Pseudomonas aeruginosa lectin LecB. Several aspects should be taken into account in development of carbohydrate-derived drugs. These are unspecific References binding to the human pathogen-recognition receptors (PRRs), low oral bioavailability and metabolic instability 1) Herold, F. et al. Novel 4-aryl-pyrido[1,2-c]pyrimidines with dual SSRI and 5-HT1A activity: Part 2. Eur. J. Med. Chem. in human and pathogen. In order to mask the natural carbohydrate character of LecB inhibitors and to increase 44, 4702–4715 (2009). their affinities, a detailed structure‑activity relationship study was performed.[4, 5, 6] Compared to unmodified 2) Herold, F. et al. Novel 4-aryl-pyrido[1,2-c]pyrimidines with dual SSRI and 5-HT1A activity, Part 1. Eur. J. Med. Chem. 1 44, 1710–1717 (2009). carbohydrates, methyl α-D-mannoside ( ) binding properties of LecB ligands were improved regarding their 3) Herold, F. et al. Novel 4-aryl-pyrido[1,2-c]pyrimidines with dual SSRI and 5-HT1A activity. part 3. Eur. J. Med. Chem. potency and complex stability. Herein discovered LecB inhibitors are supposed to overcome drawbacks of the 46, 142–149 (2011). natural carbohydrates as a drug and are a good base for future drug development in the anti-virulence approach. 4) Chodkowski, A. et al. Novel 4-aryl-pyrido[1,2-c]pyrimidines with dual SSRI and 5-HT1A activity. Part 4. Eur. J. Med. Chem. 90, 21–32 (2015). 5) Gomolka, A. et al. Novel 4-aryl-pyrido[1,2-c]pyrimidines with dual SSRI and 5-HT1A activity. part 5. Eur. J. Med. Chem. References 98, 221–236 (2015). 6) Artigas, F. Serotonin receptors involved in antidepressant effects. Pharmacol. Ther. 137, 119–131 (2013). 1) Tielker D, Hacker S, Loris R, Strathmann M, Wingender J, Wilhelm S, Rosenau F, Jaeger K. Microbiology. 2005, 151(Pt 7) Artigas, F., Adell, A. & Celada, P. Pindolol Augmentation of Antidepressant Response. Curr. Drug Targets 7, 139–147 5), 1313-23. (2006). 2) Mitchell E, Houles C, Sudakevitz D, Wimmerova M, Gautier C, Pérez S, Wu AM, Gilboa-Garber N, Imberty A. Nat 8) Dawson, L. A. & Watson, J. M. Vilazodone: A 5-HT1A receptor agonist/serotonin transporter inhibitor for the treatment Struct Biol 2002, 9(12), 918-21. of affective disorders. CNS Neurosci. Ther. 15, 107–117 (2009). 3) Hauck D, Joachim J, Frommeyer B, Varrot A, Philipp B, Möller H, Imberty A, Exner T, Titz A. ACS Chem. Biol. 2013, 8, 9) Sanchez, C., Asin, K. E. & Artigas, F. Vortioxetine, a novel antidepressant with multimodal activity: Review of preclinical 1775-84. and clinical data. Pharmacol. Ther. 145, 43–57 (2015). 4) Sommer R, Exner T E, Titz A. PLoS One 2014, 9, e112822. 5) Hofmann A, Sommer R, Hauck D, Stifel J, Gottker-Schnetmann I, Titz A. Carbohydrate Research 2015, 412, 34-42. 6) Sommer R, Hauck D, Varrot A, Wagner S, Audfray A, Prestel A, Möller H M, Imberty A, Titz A. ChemistryOpen 2015, 4, 756-767.

109 P053 P054 EXTRACTING ACTIONABLE KNOWLEDGE FROM LARGE SCALE A NON-BASIC 5-HT6R ANTAGONISTS. IN VITRO PHARMACOLOGY DATA Jakub Staron, Grzegorz Satała, Dawid Warszycki, Andrzej J. Bojarski Alexander Dossetter (1), Edward Griffen (1), Andrew Leach (1,2), Jessica Stacey (1), Lauren Reid (1), Shane Montague (1) Department of Medicinal Chemistry, Institute of Pharmacology Polish Academy of Sciences 12 Smętna Street, 31-343 Cracow, Poland 1) Medchemica Ltd, Macclesfield, United Kingdom 2) Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, Merseyside, United Kingdom

The subtype six serotonin receptor (5-HT6R) belongs to a rhodopsin-like G-protein coupled receptor family and is located exclusively in the central nervous system, in the regions associated with memory functions and In vitro pharmacology data contains vital information on the probability of binding to a recpetor and species cognition.1,2 In vivo studies have confirmed the antidepressant and procognitive properties of compounds acting selectivity issues, which can have a considerable impact on drug hunting project progress. Data from the at this target.3–5 literature and public databases are variable due to factors like assay conditions and diversity of structural types. Up to date, several thousands of 5-HT6R ligands have been obtained, from which 94% possess protonable Both structural alerts and potential chemical modifications are extracted from the data using matched molecular nitrogen atom, whereas the remaining ones are devoid of this pharmacophore feature yet still exhibit very high pair (MMP) and fragment frequency analysis. The volumne and variety of data requires the development of affinity. The advantage of non-basic ligands lies in their excellent selectivity, especially in the lack of affinity specific methods to deliver knowledge that can be usedto make decisions in a relevant timeframe. Results from 8 towards hERG.6 high priorit drug safety assays will be discussed. This knowledge has the potential benefit of reducing avoidable late stage safety failures and the methodology equally can aid the generation of potent compounds. Here we present the design of a novel, indole-based heterocyclic scaffold through a bioisostere substitution approach, that allowed to obtain a highly active non-basic 5-HT6R ligand with a Ki = 4 nM. Acknowledgements The study was partly supported by the grant OPUS 2014/13/B/NZ7/02210 financed by the Polish National Science Centre.

References 1) R. Kohen, M. A. Metcalf, N. Khan, T. Druck, K. Huebner, J. E. Lachowicz, H. Y. Meltzer, D. R. Sibley, B. L. Roth and M. W. Hamblin, J. Neurochem., 1996, 66, 47–56. 2) A. J. Sleight, F. G. Boess, M. Bös and A. Bourson, Ann. NY. Acad. Sci., 1998, 861, 91–6. 3) A. Nikiforuk, Revi. neurosci., 2014, 25, 367–82. 4) G. V Carr, L. E. Schechter and I. Lucki, Psychopharmacology (Berl)., 2011, 213, 499–507. Figure – General outline of pharmacophore finding process. Examples are VEGFR pharmacophores mapped 5) A. Quiedeville, M. Boulouard, V. Da Silva Costa-Aze, F. Dauphin, V. Bouet and T. Freret, Rev. Neurosci., 2014, 25, 417–27. onto known inhibitors. 6) A. V Ivachtchenko, D. E. Dmitriev, E. S. Golovina, E. S. Dubrovskaya, M. G. Kadieva, A. G. Koryakova, V. M. Kysil, O. D. Mitkin, S. E. Tkachenko, I. M. Okun and A. A. Vorobiov, Bioorgan. Med. Chem. Lett., 2010, 20, 2133–2136.

110 P053 P054 EXTRACTING ACTIONABLE KNOWLEDGE FROM LARGE SCALE A NON-BASIC 5-HT6R ANTAGONISTS. IN VITRO PHARMACOLOGY DATA Jakub Staron, Grzegorz Satała, Dawid Warszycki, Andrzej J. Bojarski Alexander Dossetter (1), Edward Griffen (1), Andrew Leach (1,2), Jessica Stacey (1), Lauren Reid (1), Shane Montague (1) Department of Medicinal Chemistry, Institute of Pharmacology Polish Academy of Sciences 12 Smętna Street, 31-343 Cracow, Poland 1) Medchemica Ltd, Macclesfield, United Kingdom 2) Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, Merseyside, United Kingdom

The subtype six serotonin receptor (5-HT6R) belongs to a rhodopsin-like G-protein coupled receptor family and is located exclusively in the central nervous system, in the regions associated with memory functions and In vitro pharmacology data contains vital information on the probability of binding to a recpetor and species cognition.1,2 In vivo studies have confirmed the antidepressant and procognitive properties of compounds acting selectivity issues, which can have a considerable impact on drug hunting project progress. Data from the at this target.3–5 literature and public databases are variable due to factors like assay conditions and diversity of structural types. Up to date, several thousands of 5-HT6R ligands have been obtained, from which 94% possess protonable Both structural alerts and potential chemical modifications are extracted from the data using matched molecular nitrogen atom, whereas the remaining ones are devoid of this pharmacophore feature yet still exhibit very high pair (MMP) and fragment frequency analysis. The volumne and variety of data requires the development of affinity. The advantage of non-basic ligands lies in their excellent selectivity, especially in the lack of affinity specific methods to deliver knowledge that can be usedto make decisions in a relevant timeframe. Results from 8 towards hERG.6 high priorit drug safety assays will be discussed. This knowledge has the potential benefit of reducing avoidable late stage safety failures and the methodology equally can aid the generation of potent compounds. Here we present the design of a novel, indole-based heterocyclic scaffold through a bioisostere substitution approach, that allowed to obtain a highly active non-basic 5-HT6R ligand with a Ki = 4 nM. Acknowledgements The study was partly supported by the grant OPUS 2014/13/B/NZ7/02210 financed by the Polish National Science Centre.

References 1) R. Kohen, M. A. Metcalf, N. Khan, T. Druck, K. Huebner, J. E. Lachowicz, H. Y. Meltzer, D. R. Sibley, B. L. Roth and M. W. Hamblin, J. Neurochem., 1996, 66, 47–56. 2) A. J. Sleight, F. G. Boess, M. Bös and A. Bourson, Ann. NY. Acad. Sci., 1998, 861, 91–6. 3) A. Nikiforuk, Revi. neurosci., 2014, 25, 367–82. 4) G. V Carr, L. E. Schechter and I. Lucki, Psychopharmacology (Berl)., 2011, 213, 499–507. Figure – General outline of pharmacophore finding process. Examples are VEGFR pharmacophores mapped 5) A. Quiedeville, M. Boulouard, V. Da Silva Costa-Aze, F. Dauphin, V. Bouet and T. Freret, Rev. Neurosci., 2014, 25, 417–27. onto known inhibitors. 6) A. V Ivachtchenko, D. E. Dmitriev, E. S. Golovina, E. S. Dubrovskaya, M. G. Kadieva, A. G. Koryakova, V. M. Kysil, O. D. Mitkin, S. E. Tkachenko, I. M. Okun and A. A. Vorobiov, Bioorgan. Med. Chem. Lett., 2010, 20, 2133–2136.

111 P055 P056 SILIBININ INDUCES THE ACCUMULATION OF PHOSPHOLIPIDS IN SYNTHESIS OF NOVEL BENZOTHIAZOLE-PIPERAZINE HEPATOCYTES AND MOUSE LIVER AND INCREASES THE DERIVATIVES AND THEIR BIOLOGICAL EVALUATION AS HEPATIC BIOTRANSFORMATION CAPACITY ACETYLCHOLINESTERASE INHIBITORS AND CYTOTOXIC AGENTS Maria Thürmer (1), Markus Werner (1), Oliver Werz (1), Amelie Lupp (2), Andreas Koeberle (1) Bengisu Turgutalp (1), Enise Ece Gurdal (1), Hayrettin Ozan Gulcan (2), Tugba Ercetin (2), Mustafa 1) Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Germany Fethi Sahin (2), Irem Durmaz (3), Rengul Cetin-Atalay (4), Wolfgang Sippl (5), Mine Yarim (1) 2) Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University Jena, Germany 1) Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Yeditepe University, 34755, Kayisdagi, Istanbul, Turkey 2) Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Eastern Mediterranean University, Famagusta, Northern Cyprus The silbinin is considered as major active component in extracts of Silybum marianum (milk thistle) 3) Department of Molecular Biology and Genetics, BilGen, Genetics and Biotechnology Research Center, Faculty of Science, Bilkent University, 06800, Bilkent, Ankara, Turkey and is indicated for the treatment of toxic liver damage, in particular for intoxication with Amanita phalloides. 4) Bioinformatics Department, Graduate School of Informatics, Middle East Technical University, Ankara, Turkey Pre-clinical and clinical studies have shown that the standardised extract silymarin induces the regeneration of 5) Martin-Luther-Universität Halle-Wittenberg, Institute of Pharmacy, Medicinal Chemistry Department, Halle (Saale), hepatocytes, impairs inflammatory reactions, suppresses tumorigenesis, improves insulin resistance and exerts Germany anti-atherosclerotic properties. The molecular mechanisms of silymarin are only partially understood. Membrane-stabilizing properties of silymarin, modulation of the activity/function of membrane proteins as well as effects on hepatic lipid metabolism have been discussed since decades. Here, we show that silibinin and even more pronounced the milk thistle-containing phytomarmaceutical Silimarit® significantly increase the content of Background - Chemotherapeutic agents that target cancerous cells selectively are still under investigation as major phospholipid classes (phosphatidylcholines, phosphatidylethanolamines, phosphatidylserines, drug resistance remains as a major problem [1]. In our previous study, benzothiazole-piperazine derivatives are phosphatidylinositols, phosphatidylglycerols or sphingomyelins) in human monocytes and hepatocellular shown to be cytotoxic against breast, hepatocellular, colon cancer cell lines [2]. In neurodegenerative disorders carcinoma HepG2 cells in vitro as well as in murine liver in vivo. The accumulation of hepatic phospholipids is such as Alzheimer’s disease (AD), AChEIs are prescribed primarily for the treatment of cognitive symptoms [3]. accompanied by significant changes in their relative fatty acid composition and an increase of the endoplasmic Recently compounds bearing benzothiazole and piperazine rings are reported for their potent inhibitory action on reticulum-dependent biotransformation capacity of mouse liver. We hypothesize that the apparent expansion of AChE [4]. intracellular membranes, the locus of diverse biotransforming and lipid metabolizing enzymes, might account for the liver-protective properties of silibinin related to intoxication as well as metabolic diseases such as steatosis Aims and Methods - In this study, we aimed to synthesize 11 novel and cirrhosis. N-(6-ethoxybenzothiazole-2-yl)-2-(4-substituted-piperazinyl)acetamide derivatives and investigate their cytotoxic, AChE/BuChE inhibitory activities. The compounds were tested for their cytotoxic activities against several cancer cell lines by sulforhodamine B assay and results were compared with 5-Fluorouracil. AChE/BuChE inhibitory activities of the compounds were determined by the modified Ellman method. In positive control experiments, donepezil was employed as the reference inhibitor. Synthesized compounds were identified with IR, 1H-NMR, 13C-NMR, LC-MS and UV spectra. Their purities were confirmed by elemental analysis. Docking of the inhibitor 2j was carried out using the program Gold 5.1 to predict the interaction mode of these derivatives for AChE.

Results - Ellman study results show that all compunds are selective inhibitors on AChE rather than BuChE. Most active derivative against AChE is (1-methylpiperidine-4-yl)piperazine derivative 2j (77.2%). In addition, 2j has better selectivity over AChE in comparison with reference compound Donepezil (2j; AChE:BuChE, 77.2:18.6, Donepezil; AChE:BuChE, 94.8:70.5).

Conclusion - All compounds have cytotoxic activity against hepatocellular (HUH-7) and colorectal (HCT-116) cancer cell lines. In general, dihalo substituted benzylpiperazine derivatives (2a, 2e) have the highest cytotoxic activities in all tested cell lines. In addition, acetylcholinesterase (AChE) and butyrylcholinesterase (BuChE) inhibitory activities of synthesized compounds are investigated by in vitro Ellman’s method. Compound 2j leads to high and selective inhibition against AChE. Docking study was performed on compound 2j to prove its high affinity to AChE where binding modes of 2j and Donepezil on AChE were found to be similar.

References 1) Deng, X.; Qui, Q.; Yang, B.; Wang, X.; Huang, W.; Qian, H. Design, synthesis and biological evaluation of novel peptides with anti-cancer and drug resistance-reversing activities. Eur. J. Med. Chem. 2015, 89, 540-548. 2) Gurdal, E. E.; Buclulgan, E.; Durmaz, I.; Cetin-Atalay, R.; Yarim, M. Synthesis and Anticancer Activity Evaluation of Some Benzothiazole-Piperazine Derivatives. Anticancer Agents Med. Chem. 2015, 15, 382-389. 3) Pappano, A. J. Basic & Clinical Pharmacology; Katzung B.; G, Trevor A. J. San Francisco, USA, 2014. 4) Ozkay, U. D.; Can, O. D.; Ozkay, Y.; Ozturk, Y.; Effect of benzothiazole/piperazine derivatives on intracerebroventricular streptozotocin-induced cognitive deficits. Pharmacol. Rep. 2012, 64, 834-847.

112 P055 P056 SILIBININ INDUCES THE ACCUMULATION OF PHOSPHOLIPIDS IN SYNTHESIS OF NOVEL BENZOTHIAZOLE-PIPERAZINE HEPATOCYTES AND MOUSE LIVER AND INCREASES THE DERIVATIVES AND THEIR BIOLOGICAL EVALUATION AS HEPATIC BIOTRANSFORMATION CAPACITY ACETYLCHOLINESTERASE INHIBITORS AND CYTOTOXIC AGENTS Maria Thürmer (1), Markus Werner (1), Oliver Werz (1), Amelie Lupp (2), Andreas Koeberle (1) Bengisu Turgutalp (1), Enise Ece Gurdal (1), Hayrettin Ozan Gulcan (2), Tugba Ercetin (2), Mustafa 1) Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Germany Fethi Sahin (2), Irem Durmaz (3), Rengul Cetin-Atalay (4), Wolfgang Sippl (5), Mine Yarim (1) 2) Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University Jena, Germany 1) Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Yeditepe University, 34755, Kayisdagi, Istanbul, Turkey 2) Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Eastern Mediterranean University, Famagusta, Northern Cyprus The flavonoid silbinin is considered as major active component in extracts of Silybum marianum (milk thistle) 3) Department of Molecular Biology and Genetics, BilGen, Genetics and Biotechnology Research Center, Faculty of Science, Bilkent University, 06800, Bilkent, Ankara, Turkey and is indicated for the treatment of toxic liver damage, in particular for intoxication with Amanita phalloides. 4) Bioinformatics Department, Graduate School of Informatics, Middle East Technical University, Ankara, Turkey Pre-clinical and clinical studies have shown that the standardised extract silymarin induces the regeneration of 5) Martin-Luther-Universität Halle-Wittenberg, Institute of Pharmacy, Medicinal Chemistry Department, Halle (Saale), hepatocytes, impairs inflammatory reactions, suppresses tumorigenesis, improves insulin resistance and exerts Germany anti-atherosclerotic properties. The molecular mechanisms of silymarin are only partially understood. Membrane-stabilizing properties of silymarin, modulation of the activity/function of membrane proteins as well as effects on hepatic lipid metabolism have been discussed since decades. Here, we show that silibinin and even more pronounced the milk thistle-containing phytomarmaceutical Silimarit® significantly increase the content of Background - Chemotherapeutic agents that target cancerous cells selectively are still under investigation as major phospholipid classes (phosphatidylcholines, phosphatidylethanolamines, phosphatidylserines, drug resistance remains as a major problem [1]. In our previous study, benzothiazole-piperazine derivatives are phosphatidylinositols, phosphatidylglycerols or sphingomyelins) in human monocytes and hepatocellular shown to be cytotoxic against breast, hepatocellular, colon cancer cell lines [2]. In neurodegenerative disorders carcinoma HepG2 cells in vitro as well as in murine liver in vivo. The accumulation of hepatic phospholipids is such as Alzheimer’s disease (AD), AChEIs are prescribed primarily for the treatment of cognitive symptoms [3]. accompanied by significant changes in their relative fatty acid composition and an increase of the endoplasmic Recently compounds bearing benzothiazole and piperazine rings are reported for their potent inhibitory action on reticulum-dependent biotransformation capacity of mouse liver. We hypothesize that the apparent expansion of AChE [4]. intracellular membranes, the locus of diverse biotransforming and lipid metabolizing enzymes, might account for the liver-protective properties of silibinin related to intoxication as well as metabolic diseases such as steatosis Aims and Methods - In this study, we aimed to synthesize 11 novel and cirrhosis. N-(6-ethoxybenzothiazole-2-yl)-2-(4-substituted-piperazinyl)acetamide derivatives and investigate their cytotoxic, AChE/BuChE inhibitory activities. The compounds were tested for their cytotoxic activities against several cancer cell lines by sulforhodamine B assay and results were compared with 5-Fluorouracil. AChE/BuChE inhibitory activities of the compounds were determined by the modified Ellman method. In positive control experiments, donepezil was employed as the reference inhibitor. Synthesized compounds were identified with IR, 1H-NMR, 13C-NMR, LC-MS and UV spectra. Their purities were confirmed by elemental analysis. Docking of the inhibitor 2j was carried out using the program Gold 5.1 to predict the interaction mode of these derivatives for AChE.

Results - Ellman study results show that all compunds are selective inhibitors on AChE rather than BuChE. Most active derivative against AChE is (1-methylpiperidine-4-yl)piperazine derivative 2j (77.2%). In addition, 2j has better selectivity over AChE in comparison with reference compound Donepezil (2j; AChE:BuChE, 77.2:18.6, Donepezil; AChE:BuChE, 94.8:70.5).

Conclusion - All compounds have cytotoxic activity against hepatocellular (HUH-7) and colorectal (HCT-116) cancer cell lines. In general, dihalo substituted benzylpiperazine derivatives (2a, 2e) have the highest cytotoxic activities in all tested cell lines. In addition, acetylcholinesterase (AChE) and butyrylcholinesterase (BuChE) inhibitory activities of synthesized compounds are investigated by in vitro Ellman’s method. Compound 2j leads to high and selective inhibition against AChE. Docking study was performed on compound 2j to prove its high affinity to AChE where binding modes of 2j and Donepezil on AChE were found to be similar.

References 1) Deng, X.; Qui, Q.; Yang, B.; Wang, X.; Huang, W.; Qian, H. Design, synthesis and biological evaluation of novel peptides with anti-cancer and drug resistance-reversing activities. Eur. J. Med. Chem. 2015, 89, 540-548. 2) Gurdal, E. E.; Buclulgan, E.; Durmaz, I.; Cetin-Atalay, R.; Yarim, M. Synthesis and Anticancer Activity Evaluation of Some Benzothiazole-Piperazine Derivatives. Anticancer Agents Med. Chem. 2015, 15, 382-389. 3) Pappano, A. J. Basic & Clinical Pharmacology; Katzung B.; G, Trevor A. J. San Francisco, USA, 2014. 4) Ozkay, U. D.; Can, O. D.; Ozkay, Y.; Ozturk, Y.; Effect of benzothiazole/piperazine derivatives on intracerebroventricular streptozotocin-induced cognitive deficits. Pharmacol. Rep. 2012, 64, 834-847.

113 P057 P058 NOVEL 11β-HSD1 INHIBITORS FOR AGE-RELATED COGNITIVE ANTIPROLIFERATIVE ACTIVITY OF HIGHLY SELECTIVE DAPK-1 DISORDERS AND ALZHEIMER’S DISEASE INHIBITORS IN HUMAN LEUKEMIC T LYMPHOCYTES

Rosana Leiva (1), Christian Griñan (2), Constantí Seira (3), Elena Valverde (1), Andrew McBride (4), Álvaro Lorente-Macías (1), María J. Pineda de las Infantas (1), Sara Torres-Rusillo (2), Juan J. Molina Margaret Binnie (4), Belén Pérez (5), Axel Bidon-Chanal (3), Mercè Pallàs (2), F. Javier Luque (3), Scott (2), Asier Unciti-Broceta (3), Juan J. Díaz-Mochón (4) P. Webster (4), Santiago Vázquez (1) 1) Departamento de Química Farmacéutica y Orgánica, Facultad de Farmacia, Universidad de Granada, Campus de 1) Laboratori de Química Farmacèutica (Unitat Associada al CSIC), Facultat de Farmàcia i Institut de Biomedicina (IBUB), Cartuja s/n, 18071 Granada, Spain. Universitat de Barcelona, Av. Joan XXIII, 27-31, 08028 Barcelona, Spain. 2) Instituto de Biopatología y Medicina Regenerativa, Centro de Investigación Biomédica, Universidad de Granada, Parque 2) Unitat de Farmacologia i Farmacognòsia, Facultat de Farmàcia i IBUB, Universitat de Barcelona, Av. Joan XXIII, Tecnológico de Ciencias de la Salud (PTS), Avda. del Conocimiento s/n, 18016 Armilla, Granada, Spain. 27-31, 08028 Barcelona, Spain. 3) Edinburgh Cancer Research UK Centre, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, 3) Departament de Fisicoquímica, Facultat de Farmàcia i IBUB, Universitat de Barcelona, Av. Prat de la Riba, 171, 08921 Crewe Road South, Edinburgh EH4 2XR, UK. Santa Coloma de Gramenet, Spain. 4) Centro Pfizer – Universidad de Granada – Junta de Andalucía de Genómica e Investigación Oncológica (GENYO), 4) Endocrinology Unit, Centre for Cardiovascular Science, University of Edinburgh, Queen’s Medical Research Institute, Parque Tecnológico de Ciencias de la Salud (PTS), 18016 Granda, Spain. EH16 4TJ, United Kingdom. 5) Departament de Farmacologia, de Terapèutica i de Toxicologia, Institut de Neurociències, Universitat Autònoma de Barcelona, E-08193, Bellaterra, Barcelona, Spain.

A 18-member library of 6,8,9-poly-substituted purines was prepared from pyrimidines1, primary and N,N-dimethylamides under basic conditions via a novel one-pot synthetic pathways2controlled by amide sizes and the novel analogues were tested against two leukemia cell lines: Jurkat (acute T cell leukemia) and K562 Growing evidence suggests that excessive glucocorticoid activity may contribute to age-associated memory (chronic erythroleukemia) cells. Compounds having a benzoxy group at C6 position of the aromatic ring impairment and Alzheimer’s disease (AD).[1] It is known that glucocorticoids locally generated by exhibited antiproliferative activity in Jurkat cells whereas all compounds induced a lower effect on K562 cells. 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1) play a major role in age-related cognitive impairments, Analysis of cell cycle, Annexin-V3 staining and cleavage of initiator caspases assays showed that the active and that 11β-HSD1 deficiency prevents spatial memory impairments and cognitive decline with aging.[2] purine analogues induce cell death by apoptosis. Based on these results, a new purine derivative was synthesized, Several groups and investigations have reaffirmed these findings with different in vivo experiments in rodent 6-benzyloxy-9-tert-butyl-8-phenyl-9H-purine (6d), which displayed the highest activity of the series against models. Acute and short-term treatments with 11β-HSD1 inhibitors have showed memory consolidation and Jurkat cell lines. Finally, P33-radiolabeled kinase assays using 96 recombinant human kinases known to be improvements in cognitive function in aged mice and AD models.[3] Overall, these data suggest that 11β-HSD1 involved in apoptotic events were performed. Just one of the kinases tested, DAPK-1, was inhibited 50% or inhibitors provide a novel approach through a non-cholinergic mechanism to deal with these cognitive disorders. more by the phenotypic hits at 10 µM, suggesting that the inhibition of this target could be responsible for the induction of cell death by apoptosis. In agreement with the phenotypic results, the most active antiproliferative Given that the enzyme active site includes a hydrophobic pocket to accommodate bulky lipophilic scaffolds, our agent, 6d, displayed also the lowest IC50 value against recombinant DAPK1 (2.5 µM), further supporting the group is currently focused on the design and synthesis of new 11β-HSD1 inhibitors taking advantage of our potential role of this protein on the observed functional response. DAPK-1 inhibition4 led by 6d together with its expertise in polycyclic compounds. pro-apoptotic properties against Jurkat line makes it an interesting candidate for further investigate the role of Our starting point was the hit compound 1, which exhibited a submicromolar potency against 11β-HSD1. Based DAPK1 kinase in triggering apoptosis in cancer cells, a role which is attracting recent interest. on this result, a medicinal chemistry program focused on the polycyclic scaffold optimization delivered more potent inhibitors that were characterized in terms of metabolic stability and brain penetration.[4] Our best candidate 2 was administered to twelve-moth SAMP8 mice during four weeks in their drinking water. Treated References mice performed significantly better than control ones in the behavioral tests demonstrating an improvement in 1) Maria J. Pineda de las Infantas y Villatoro, Juan D. Unciti-Broceta, Rafael Contreras-Montoya, Jose A. Garcia-Salcedo, the cognitive decline of already aged mice. These findings also correlate with the results of the biochemical and Miguel A. Gallo Mezo, Asier Unciti-Broceta & Juan J. Diaz-Mochon, 2015, 5, 9139. 2) Maria J. Pineda de las Infantas, Sara Torres-Rusillo, Juan Diego Unciti-Broceta, Pablo Fernandez-Rubio, Maria Angelica molecular experiments carried out. Overall, these data contribute to confirming 11β-HSD1 as a plausible target Luque-Gonzalez, Miguel A. Gallo, Asier Unciti-Broceta Ignacio J. Molina and Juan J. Díaz-Mochón, Organic and for cognitive disorders including AD. Biomolecular Chemistry, 2015, 13, 5224-5234. 3) G. Koopman, C. P. Reutelingsperger, G. A. Kuijten, R. M. Keehnen, S. T. Pals, and M. H. van Oers, Blood, 1994, 84, 1415-1420. 4) O. Cohen, B. Inbal, J. L. Kissil, T. Raveh, H. Berissi, T. Spivak-Kroizaman, E. Feinstein, and A. Kimchi, J. Cell Biol., 1999, 146, 141-148.

References 1) a) Moisan, M. P. et al Endocrinology 1990, 127, 1450-1455. b) Pelletier, G. et al Neuroscience 2007, 145, 110-115. c) Bisschop, P. H. et al J. Neuroendocrinol. 2013, 25, 425-432. 2) a) MacLullich, A. M. J. et al Neurobiol. Aging 2012, 33, 207.e1-207.e8. b) Yau, J. L. W. et al J. Neurosci. 2011, 31, 4188-4193. c) Yau, J. L. W. et al Frontiers Aging Neurosci. 2012, 4, 24. 3) a) Sooy, K. et al J. Neurosci. 2010, 30, 13867-13872. b) Wheelan, N. et al Neuropharmacol. 2015, 91, 71-76. c) Sooy, K. et al Endocrinology 2015, 156, 4592-4603. d) Mohler, E. G. et al . J. Neurosci. 2011, 31, 5406-5413. 4) Vázquez, S.; Leiva, R.; Valverde, E. EP16382175.4, Universitat de Barcelona and CIDQO 2012.

114 P057 P058 NOVEL 11β-HSD1 INHIBITORS FOR AGE-RELATED COGNITIVE ANTIPROLIFERATIVE ACTIVITY OF HIGHLY SELECTIVE DAPK-1 DISORDERS AND ALZHEIMER’S DISEASE INHIBITORS IN HUMAN LEUKEMIC T LYMPHOCYTES

Rosana Leiva (1), Christian Griñan (2), Constantí Seira (3), Elena Valverde (1), Andrew McBride (4), Álvaro Lorente-Macías (1), María J. Pineda de las Infantas (1), Sara Torres-Rusillo (2), Juan J. Molina Margaret Binnie (4), Belén Pérez (5), Axel Bidon-Chanal (3), Mercè Pallàs (2), F. Javier Luque (3), Scott (2), Asier Unciti-Broceta (3), Juan J. Díaz-Mochón (4) P. Webster (4), Santiago Vázquez (1) 1) Departamento de Química Farmacéutica y Orgánica, Facultad de Farmacia, Universidad de Granada, Campus de 1) Laboratori de Química Farmacèutica (Unitat Associada al CSIC), Facultat de Farmàcia i Institut de Biomedicina (IBUB), Cartuja s/n, 18071 Granada, Spain. Universitat de Barcelona, Av. Joan XXIII, 27-31, 08028 Barcelona, Spain. 2) Instituto de Biopatología y Medicina Regenerativa, Centro de Investigación Biomédica, Universidad de Granada, Parque 2) Unitat de Farmacologia i Farmacognòsia, Facultat de Farmàcia i IBUB, Universitat de Barcelona, Av. Joan XXIII, Tecnológico de Ciencias de la Salud (PTS), Avda. del Conocimiento s/n, 18016 Armilla, Granada, Spain. 27-31, 08028 Barcelona, Spain. 3) Edinburgh Cancer Research UK Centre, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, 3) Departament de Fisicoquímica, Facultat de Farmàcia i IBUB, Universitat de Barcelona, Av. Prat de la Riba, 171, 08921 Crewe Road South, Edinburgh EH4 2XR, UK. Santa Coloma de Gramenet, Spain. 4) Centro Pfizer – Universidad de Granada – Junta de Andalucía de Genómica e Investigación Oncológica (GENYO), 4) Endocrinology Unit, Centre for Cardiovascular Science, University of Edinburgh, Queen’s Medical Research Institute, Parque Tecnológico de Ciencias de la Salud (PTS), 18016 Granda, Spain. EH16 4TJ, United Kingdom. 5) Departament de Farmacologia, de Terapèutica i de Toxicologia, Institut de Neurociències, Universitat Autònoma de Barcelona, E-08193, Bellaterra, Barcelona, Spain.

A 18-member library of 6,8,9-poly-substituted purines was prepared from pyrimidines1, primary alcohols and N,N-dimethylamides under basic conditions via a novel one-pot synthetic pathways2controlled by amide sizes and the novel analogues were tested against two leukemia cell lines: Jurkat (acute T cell leukemia) and K562 Growing evidence suggests that excessive glucocorticoid activity may contribute to age-associated memory (chronic erythroleukemia) cells. Compounds having a benzoxy group at C6 position of the aromatic ring impairment and Alzheimer’s disease (AD).[1] It is known that glucocorticoids locally generated by exhibited antiproliferative activity in Jurkat cells whereas all compounds induced a lower effect on K562 cells. 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1) play a major role in age-related cognitive impairments, Analysis of cell cycle, Annexin-V3 staining and cleavage of initiator caspases assays showed that the active and that 11β-HSD1 deficiency prevents spatial memory impairments and cognitive decline with aging.[2] purine analogues induce cell death by apoptosis. Based on these results, a new purine derivative was synthesized, Several groups and investigations have reaffirmed these findings with different in vivo experiments in rodent 6-benzyloxy-9-tert-butyl-8-phenyl-9H-purine (6d), which displayed the highest activity of the series against models. Acute and short-term treatments with 11β-HSD1 inhibitors have showed memory consolidation and Jurkat cell lines. Finally, P33-radiolabeled kinase assays using 96 recombinant human kinases known to be improvements in cognitive function in aged mice and AD models.[3] Overall, these data suggest that 11β-HSD1 involved in apoptotic events were performed. Just one of the kinases tested, DAPK-1, was inhibited 50% or inhibitors provide a novel approach through a non-cholinergic mechanism to deal with these cognitive disorders. more by the phenotypic hits at 10 µM, suggesting that the inhibition of this target could be responsible for the induction of cell death by apoptosis. In agreement with the phenotypic results, the most active antiproliferative Given that the enzyme active site includes a hydrophobic pocket to accommodate bulky lipophilic scaffolds, our agent, 6d, displayed also the lowest IC50 value against recombinant DAPK1 (2.5 µM), further supporting the group is currently focused on the design and synthesis of new 11β-HSD1 inhibitors taking advantage of our potential role of this protein on the observed functional response. DAPK-1 inhibition4 led by 6d together with its expertise in polycyclic compounds. pro-apoptotic properties against Jurkat line makes it an interesting candidate for further investigate the role of Our starting point was the hit compound 1, which exhibited a submicromolar potency against 11β-HSD1. Based DAPK1 kinase in triggering apoptosis in cancer cells, a role which is attracting recent interest. on this result, a medicinal chemistry program focused on the polycyclic scaffold optimization delivered more potent inhibitors that were characterized in terms of metabolic stability and brain penetration.[4] Our best candidate 2 was administered to twelve-moth SAMP8 mice during four weeks in their drinking water. Treated References mice performed significantly better than control ones in the behavioral tests demonstrating an improvement in 1) Maria J. Pineda de las Infantas y Villatoro, Juan D. Unciti-Broceta, Rafael Contreras-Montoya, Jose A. Garcia-Salcedo, the cognitive decline of already aged mice. These findings also correlate with the results of the biochemical and Miguel A. Gallo Mezo, Asier Unciti-Broceta & Juan J. Diaz-Mochon, 2015, 5, 9139. 2) Maria J. Pineda de las Infantas, Sara Torres-Rusillo, Juan Diego Unciti-Broceta, Pablo Fernandez-Rubio, Maria Angelica molecular experiments carried out. Overall, these data contribute to confirming 11β-HSD1 as a plausible target Luque-Gonzalez, Miguel A. Gallo, Asier Unciti-Broceta Ignacio J. Molina and Juan J. Díaz-Mochón, Organic and for cognitive disorders including AD. Biomolecular Chemistry, 2015, 13, 5224-5234. 3) G. Koopman, C. P. Reutelingsperger, G. A. Kuijten, R. M. Keehnen, S. T. Pals, and M. H. van Oers, Blood, 1994, 84, 1415-1420. 4) O. Cohen, B. Inbal, J. L. Kissil, T. Raveh, H. Berissi, T. Spivak-Kroizaman, E. Feinstein, and A. Kimchi, J. Cell Biol., 1999, 146, 141-148.

References 1) a) Moisan, M. P. et al Endocrinology 1990, 127, 1450-1455. b) Pelletier, G. et al Neuroscience 2007, 145, 110-115. c) Bisschop, P. H. et al J. Neuroendocrinol. 2013, 25, 425-432. 2) a) MacLullich, A. M. J. et al Neurobiol. Aging 2012, 33, 207.e1-207.e8. b) Yau, J. L. W. et al J. Neurosci. 2011, 31, 4188-4193. c) Yau, J. L. W. et al Frontiers Aging Neurosci. 2012, 4, 24. 3) a) Sooy, K. et al J. Neurosci. 2010, 30, 13867-13872. b) Wheelan, N. et al Neuropharmacol. 2015, 91, 71-76. c) Sooy, K. et al Endocrinology 2015, 156, 4592-4603. d) Mohler, E. G. et al . J. Neurosci. 2011, 31, 5406-5413. 4) Vázquez, S.; Leiva, R.; Valverde, E. EP16382175.4, Universitat de Barcelona and CIDQO 2012.

115 P059 P060 DEVELOPMENT OF BISPECIFIC BICYCLIC PEPTIDES SYNTHESIS AND EVALUATION OF AURONE INTERACTIONS WITH TAU AGGREGATES IN ALZHEIMER'S DISEASE Christina Lamers, Christian Heinis Laurent Lunven (1,2), Sabine Chierici (1), Ahcène Boumendjel (2), Marine Peuchmaur (2) Laboratory of Therapeutic Proteins and Peptides (LPPT), Institute of Chemical Sciences and Engineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), Avenue Forel 2, CH-1015 Lausanne, Switzerland 1) Centre National de la Recherche Scientifique (CNRS), UMR 5250, Département de Chimie Moléculaire, Equipe I2BM, 570 rue de la Chimie, Université Grenoble Alpes, BP 53 38041 GRENOBLE CEDEX 9 2) Centre National de la Recherche Scientifique (CNRS), UMR 5063, Département de Pharmacochimie Moléculaire, Equipe MedChem, 470 rue de la Chimie, Université Grenoble Alpes, BP 53 38041 GRENOBLE CEDEX 9 Bicyclic peptides (figure A) are constrained peptides that show higher binding affinity and selectivity for a target as compared to a linear or monocyclic peptide.1 The constrained structure reduces the entropic penalty upon binding and therefore increases affinity, and the rigidity and chemical conjugation render the peptides more resistant to proteases. Several bicyclic peptides previously developed in our lab demonstrated a high stability in Alzheimer’s disease (AD) is the most common neurodegenerative disorder. The two hallmarks of AD in the plasma, as well as in intestinal extract, greatly increasing their usefulness as potential drug candidates.[1] brain are senile plaques composed of extracellular deposits of amyloid-β (Aβ) peptides and neurofibrillary tangles formed by abnormal filaments of tau protein. Trials focusing on targeting Aβ alone have to date been In this study we develop bispecific bicyclic peptides using phage-display[2] to target two homologous serine unsuccessful at a clinical level. As abnormal tau accumulation in the brain precedes the outbreak of senile proteases, the coagulation factor XIIa and plasma kallikrein. Both serine proteases are involved in the intrinsic plaques, the study of the formation of tau filaments has become an increasingly attractive focus over the past few coagulation cascade, which is activated by coagulation factor XII (FXII, Hageman factor). Activated FXII years. So far, a few compounds such as polyphenols have been shown to interfere with the fibrillation process of cleaves plasma prekallikrein (PPK) to generate active plasma kallikrein (PK), which generates FXIIa in a tau in vitro and in particular with neurotoxic oligomer intermediates.(a,b,c) Aurones are a subclass of naturally reciprocal fashion, and an additional inflammatory mediator. Both PK and FXIIa are important targets in occurring with low toxicity and numerous therapeutic interests.(d) This prompted us to investigated coagulation and inflammatory disorders.[3] By blocking both targets simultaneously the reciprocal activation of the interactions of aurones with tau fibres and aggregates. the intrinsic coagulation cascade will be inhibited, which ideally can be done with the administration of only one drug instead of two. We used short sequences of tau as models of tau fibrillation to assess the in vitro inhibitory effect of our compounds by means of Thioflavin fluorescence assays, circular dichroism and atomic force microscopy (AFM). In order to make a bispecific bicyclic peptide, our previously established libraries are evolved with With a 22 amino acid sequence (R3 fragment), we were able to obtain fibres similar to native tau fibres in AFM phage-display where we alternate panning against each target, FXIIa and PK. Peptides have been found that (Figure). The screening of a small library of aurones using our models highlighted the importance of hydroxyl show consensus sequences within the same molecule, comparable to the consensus of previously developed groups for interactions between aurones and fibres, thus supporting the importance of polyphenols on FXIIa and PK inhibitors. Their inhibition constant for both targets and anti-coagulation properties are currently fibrillation. being tested.

Figure: Inhibition of tau model fibres in presence of the aurone U2.

References a) Taniguchi et al., J. Biol. Chem. 2005, 280, 7614-7623 b) Porat et al., Chem Biol Drug Des 2006; 67: 27–37 c) Ono et al., J Neurochem 2003, 87, 172-181 d) Haudecoeur et al., Current Medicinal Chemistry 2012, 19, 2861-2875

Example of bicyclic peptides. A) chemical structure. B) three dimensional model.

References 1) Baeriswyl V, et al. Protein engineering, design & selection : PEDS. 2013; 26: 81-89. 2) Heinis C, et al. Nature chemical biology. 2009; 5: 502-507. 3) Long AT , et al. Journal of Thrombosis and Haemostasis. 2016; 14: 427-437.

116 P059 P060 DEVELOPMENT OF BISPECIFIC BICYCLIC PEPTIDES SYNTHESIS AND EVALUATION OF AURONE INTERACTIONS WITH TAU AGGREGATES IN ALZHEIMER'S DISEASE Christina Lamers, Christian Heinis Laurent Lunven (1,2), Sabine Chierici (1), Ahcène Boumendjel (2), Marine Peuchmaur (2) Laboratory of Therapeutic Proteins and Peptides (LPPT), Institute of Chemical Sciences and Engineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), Avenue Forel 2, CH-1015 Lausanne, Switzerland 1) Centre National de la Recherche Scientifique (CNRS), UMR 5250, Département de Chimie Moléculaire, Equipe I2BM, 570 rue de la Chimie, Université Grenoble Alpes, BP 53 38041 GRENOBLE CEDEX 9 2) Centre National de la Recherche Scientifique (CNRS), UMR 5063, Département de Pharmacochimie Moléculaire, Equipe MedChem, 470 rue de la Chimie, Université Grenoble Alpes, BP 53 38041 GRENOBLE CEDEX 9 Bicyclic peptides (figure A) are constrained peptides that show higher binding affinity and selectivity for a target as compared to a linear or monocyclic peptide.1 The constrained structure reduces the entropic penalty upon binding and therefore increases affinity, and the rigidity and chemical conjugation render the peptides more resistant to proteases. Several bicyclic peptides previously developed in our lab demonstrated a high stability in Alzheimer’s disease (AD) is the most common neurodegenerative disorder. The two hallmarks of AD in the plasma, as well as in intestinal extract, greatly increasing their usefulness as potential drug candidates.[1] brain are senile plaques composed of extracellular deposits of amyloid-β (Aβ) peptides and neurofibrillary tangles formed by abnormal filaments of tau protein. Trials focusing on targeting Aβ alone have to date been In this study we develop bispecific bicyclic peptides using phage-display[2] to target two homologous serine unsuccessful at a clinical level. As abnormal tau accumulation in the brain precedes the outbreak of senile proteases, the coagulation factor XIIa and plasma kallikrein. Both serine proteases are involved in the intrinsic plaques, the study of the formation of tau filaments has become an increasingly attractive focus over the past few coagulation cascade, which is activated by coagulation factor XII (FXII, Hageman factor). Activated FXII years. So far, a few compounds such as polyphenols have been shown to interfere with the fibrillation process of cleaves plasma prekallikrein (PPK) to generate active plasma kallikrein (PK), which generates FXIIa in a tau in vitro and in particular with neurotoxic oligomer intermediates.(a,b,c) Aurones are a subclass of naturally reciprocal fashion, and an additional inflammatory mediator. Both PK and FXIIa are important targets in occurring flavonoids with low toxicity and numerous therapeutic interests.(d) This prompted us to investigated coagulation and inflammatory disorders.[3] By blocking both targets simultaneously the reciprocal activation of the interactions of aurones with tau fibres and aggregates. the intrinsic coagulation cascade will be inhibited, which ideally can be done with the administration of only one drug instead of two. We used short sequences of tau as models of tau fibrillation to assess the in vitro inhibitory effect of our compounds by means of Thioflavin fluorescence assays, circular dichroism and atomic force microscopy (AFM). In order to make a bispecific bicyclic peptide, our previously established libraries are evolved with With a 22 amino acid sequence (R3 fragment), we were able to obtain fibres similar to native tau fibres in AFM phage-display where we alternate panning against each target, FXIIa and PK. Peptides have been found that (Figure). The screening of a small library of aurones using our models highlighted the importance of hydroxyl show consensus sequences within the same molecule, comparable to the consensus of previously developed groups for interactions between aurones and fibres, thus supporting the importance of polyphenols on FXIIa and PK inhibitors. Their inhibition constant for both targets and anti-coagulation properties are currently fibrillation. being tested.

Figure: Inhibition of tau model fibres in presence of the aurone U2.

References a) Taniguchi et al., J. Biol. Chem. 2005, 280, 7614-7623 b) Porat et al., Chem Biol Drug Des 2006; 67: 27–37 c) Ono et al., J Neurochem 2003, 87, 172-181 d) Haudecoeur et al., Current Medicinal Chemistry 2012, 19, 2861-2875

Example of bicyclic peptides. A) chemical structure. B) three dimensional model.

References 1) Baeriswyl V, et al. Protein engineering, design & selection : PEDS. 2013; 26: 81-89. 2) Heinis C, et al. Nature chemical biology. 2009; 5: 502-507. 3) Long AT , et al. Journal of Thrombosis and Haemostasis. 2016; 14: 427-437.

117 P061 DEVELOPMENT OF NOVEL AMIDES AS NON COVALENT INHIBITORS OF IMMUNOPROTEASOME

Roberta Ettari (1), Santo Previti (1), Santina Maiorana (1), Manuela Guccione (1), Sandrine Ongeri (2), Maria Zappalà (1), Silvana Grasso (1)

1) Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Annunziata, 98168 Messina, Italy 2) Molécules Fluorées et Chimie Médicinale, BioCIS, Univ. Paris-Sud, CNRS, Université Paris Saclay, 5 rue Jean-Baptiste Clément, Chaîtenay-Malabry Cedex, France

The ubiquitin-proteasome pathway is the most important non-lysosomal proteolytic system able to control the levels of several proteins that regulate the cell cycle progression and the apoptosis. The proteolytic sites are located into β1, β2 and β5 subunits of 20S core, which are respectively responsible for the caspase-like (C-L), trypsin-like (T-L) and chymotrypsin-like (ChT-L) proteasome activities. Defects and aberrations of this proteolytic system may lead to different types of hematologic malignancies. In addition to the constitutive proteasome, vertebrates express a specialized form of proteasome, named immunoproteasome (i20S), highly expressed in lynfocytes and monocytes; it is worth noting that i20S is the major form of the proteasome expressed in cells of hematopoietic origin, thus targeting i20S could be a promising strategy for the treatment of hematologic malignancies, including multiple myeloma [1]. In the last years, our research group has been actively involved in the development of novel 20S proteasome inhibitors, in particular we developed a series of amides as non-covalent proteasome inhibitors that, with respect to covalent inhibitors, might be a promising alternative to use in therapy, because of the lack of all drawbacks and side-effects related to irreversible inhibition. The non-covalent binding mode of the most active inhibitors (e.g. 1, Fig.1) was corroborated by docking simulations into the 20S proteasome crystal structure: the ligands did not interact with the active-site β5-T1 Og nucleophile, which is considered a common principle of ChT-L inhibitor binding; the isopentyl group linked to the amide moiety projected into the S1 pocket. The compounds accommodated their aromatic group perfectly in the S3 pocket through a series of van derWaals interactions, thereby profoundly stabilizing these moieties in the ChT-L site [2]. Starting from these considerations, we designed novel amides (2, Fig.1 ) as inhibitors of immunoproteasome core particles, characterized by different monocyclic or bicyclic scaffolds linked through a spacer to an amide functionality. The amide group is N-alkyl or N-aryl substituted, according to the strong preference of the immunoproteasome for bulky hydrophobic substrates at the P1 site. The saturated or unsaturated spacer of different length (1-3 carbon atoms) has been selected to evaluate the optimal distance between the amide portion and the scaffold to better allow the accommodation of these moieties into S1 and S3 pocket, respectively. Each novel amide has been tested against all the constitutive and immuno-core particles, to evaluate their selectivity profile, thus allowing us to identify several immuno-core particles selective inhibitors.

Figure 1. Development from the lead compound 1 of novel amides 2

References 1) R. Ettari, S. Previti, A. Bitto, S. Grasso, M. Zappalà, Immunoproteasome-selective inhibitors: a promising strategy to treat hematologic malignancies, autoimmune and inflammatory diseases, Curr. Med. Chem. 2016, 23, 1217-1238. 2) K. Scarbaci, V. Troiano, N. Micale, R. Ettari, L. Tamborini, C. Di Giovanni, C. Cerchia, S. Grasso, E. Novellino, T. Schirmeister, A. Lavecchia, M. Zappalà, Identification of a new series of amides as non-covalent proteasome inhibitors, Eur. J. Med. Chem. 2014, 76, 1-9. 118 P061 DEVELOPMENT OF NOVEL AMIDES AS NON COVALENT INHIBITORS OF IMMUNOPROTEASOME

Roberta Ettari (1), Santo Previti (1), Santina Maiorana (1), Manuela Guccione (1), Sandrine Ongeri (2), Maria Zappalà (1), Silvana Grasso (1)

1) Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Annunziata, 98168 Messina, Italy 2) Molécules Fluorées et Chimie Médicinale, BioCIS, Univ. Paris-Sud, CNRS, Université Paris Saclay, 5 rue Jean-Baptiste Clément, Chaîtenay-Malabry Cedex, France

The ubiquitin-proteasome pathway is the most important non-lysosomal proteolytic system able to control the levels of several proteins that regulate the cell cycle progression and the apoptosis. The proteolytic sites are located into β1, β2 and β5 subunits of 20S core, which are respectively responsible for the caspase-like (C-L), trypsin-like (T-L) and chymotrypsin-like (ChT-L) proteasome activities. Defects and aberrations of this proteolytic system may lead to different types of hematologic malignancies. In addition to the constitutive proteasome, vertebrates express a specialized form of proteasome, named immunoproteasome (i20S), highly expressed in lynfocytes and monocytes; it is worth noting that i20S is the major form of the proteasome expressed in cells of hematopoietic origin, thus targeting i20S could be a promising strategy for the treatment of hematologic malignancies, including multiple myeloma [1]. In the last years, our research group has been actively involved in the development of novel 20S proteasome inhibitors, in particular we developed a series of amides as non-covalent proteasome inhibitors that, with respect to covalent inhibitors, might be a promising alternative to use in therapy, because of the lack of all drawbacks and side-effects related to irreversible inhibition. The non-covalent binding mode of the most active inhibitors (e.g. 1, Fig.1) was corroborated by docking simulations into the 20S proteasome crystal structure: the ligands did not interact with the active-site β5-T1 Og nucleophile, which is considered a common principle of ChT-L inhibitor binding; the isopentyl group linked to the amide moiety projected into the S1 pocket. The compounds accommodated their aromatic group perfectly in the S3 pocket through a series of van derWaals interactions, thereby profoundly stabilizing these moieties in the ChT-L site [2]. Starting from these considerations, we designed novel amides (2, Fig.1 ) as inhibitors of immunoproteasome core particles, characterized by different List of abstracts monocyclic or bicyclic scaffolds linked through a spacer to an amide functionality. The amide group is N-alkyl or N-aryl substituted, according to the strong preference of the immunoproteasome for bulky hydrophobic substrates at the P1 site. The saturated or unsaturated spacer of different length (1-3 carbon atoms) has been selected to evaluate the optimal distance between the amide portion and the scaffold to better allow the accommodation of these moieties into S1 and S3 pocket, respectively. Each novel amide has been tested against all the constitutive and immuno-core particles, to evaluate their selectivity profile, thus allowing us to identify several immuno-core particles selective inhibitors.

Figure 1. Development from the lead compound 1 of novel amides 2

References 1) R. Ettari, S. Previti, A. Bitto, S. Grasso, M. Zappalà, Immunoproteasome-selective inhibitors: a promising strategy to treat hematologic malignancies, autoimmune and inflammatory diseases, Curr. Med. Chem. 2016, 23, 1217-1238. 2) K. Scarbaci, V. Troiano, N. Micale, R. Ettari, L. Tamborini, C. Di Giovanni, C. Cerchia, S. Grasso, E. Novellino, T. Schirmeister, A. Lavecchia, M. Zappalà, Identification of a new series of amides as non-covalent proteasome inhibitors, Eur. J. Med. Chem. 2014, 76, 1-9. 119 ABD ELRAHEEM Eman TOWARD LIBRARIES OF ARTIFICIAL MACROCYCLES FP01 DRACZKOWSKI Piotr EXPERIMENTAL PLATFORM FOR CHARACTERIZATION AND HIGH- CP14 THROUGHPUT SCREENING OF ACETYLCHOLINESTERASE ADORNATO Ilenia IDENTIFICATION OF NOVEL 5-ARYLIDENE-3-PHENYL-2- P001 INHIBITORS. THIOXOIMIDAZOLIDIN-4-ONES AS INHIBITORS OF THE CHYMOTRYPSIN-LIKE ACTIVITIES OF THE CONSTITUTIVE AND DUMA Bogdan TARGETING ASPRS IN THE FIGHT AGAINST M. TUBERCULOSIS P016 IMMUNO-PROTEASOMES FAMILGLINI Valeria DRUG DESIGN, SYNTHESIS AND BIOLOGICAL EVALUATION OF CP02 ADORNATO Ilenia DESIGN AND SYNTHESIS OF DUAL INHIBITORS OF ALDOSE P002 ANTIRETROVIRAL AND ANTICANCER AGENTS REDUCTASE AND PROTEIN TYROSINE PHOPSHATASE 1B FEDOROWICZ Joanna SYNTHESIS AND ANTIBACTERIAL ACTIVITY OF FLUORESCENT P017 AL HARTHY Thuraya DESIGN AND SYNTHESIS OF SOME 2-ARYL-5-FLUORO-6-(4- P003 QUINOLONES METHYL-1-PIPERAZINYL) BENZOTHIAZOLES OF POTENTIAL ANTITUMOR ACTIVITIES FIORILLO Gaetano RECENT ADVANCES IN ANTITUMOUR BERBERINES P018

ALEKSANDROVA Galina CERTAIN COMMON REGULARITIES IN THE CHEMICAL STRUCTURE P004 FREEK Janssen DEVELOPMENT OF HIGHLY SELECTIVE AND REVERSIBLE CP05 OF ANTIHISTAMINES, AND THEIR INFLUENCE ON PROPERTIES DIACYLGLYCEROL LIPASE INHIBITORS

ALEKSANDROVA Galina MODIFICATION OF STRUCTURE OF A P005 GALDEANO Carles DRUGGING THE UNDRUGGABLE: TARGETING CHALLENGING E3 CP04 HYDROXYTRIFLUOROMETHYLINDOLE, FOR THE PURPOSE OF LIGASES INCREASE IN ITS WATER SOLUBILITY GIGANTE Alba IDENTIFICATION OF NOVEL NON-NATURAL SUPRAMOLECULAR FP08 AMIN Mohammad DEVELOPMENT OF STEREOSELECTIVE SYNTHSES AND CHEMICAL P006 LIGANDS AS STABILIZERS OF 14-3-3? PROTEIN-PROTEIN BIOLOGY STUDIES FOR BROMODOMAIN INHIBITORS AS ANTI INTERACTIONS CANCER DRUG GIRAUDO Alessandro SYNTHESIS AND PHARMACOLOGICAL EVALUATION OF NOVEL P019 BALABON Olga DISCOVERY, STRUCTURAL EXPLORATION AND BIOLOGICAL FP02 AMINO BIOISOSTERES FOR THE GABAa RECEPTOR PROFILING OF A NOVEL CLASS OF ANTIMYCOBACTERIAL DPRE1 INHIBITORS GRACE Jake HIGH THROUGHPUT LIBRARY SYNTHESIS IN MEDICINAL P020 CHEMISTRY BOSCA Federica DESIGN OF PORPHYRIN-LOADED NANOBUBBLES FOR P007 THERANOSTIC APPLICATIONS GRINBERGA Signe SYNTHESIS AND PHARMACOLOGICAL EVALUATION OF AMIDINE P021 CONTAINING GABAA RECEPTOR AGONISTS BROCKMEYER Fabian ORALLY BIOAVAILABLE ANTIMALARIAL 4(1H)-QUINOLONE FP03 PRODRUGS WITH SINGLE-DOSE CURES GUR Zehra Tugce NOVEL BRP-7 DERIVATIVES TARGETING FLAP: POTENT P022 INHIBITORS OF LEUKOTRIENE BIOSYNTHESIS BUGNO Ryszard DESIGN, SYNTHESIS AND STRUCTURE-ACTIVITY RELATIONSHIPS P008 OF 1-(ARYLSULFONYL)-1H-INDOLE DERIVATIVES AS NON-BASIC 5- GURINOVA Jana TARGETS IN ORPHAN DISEASES AND THEIR REPURPOSING FP09 HT6 RECEPTOR LIGANDS CANDIDATES - ANALYSIS WITH A KNIME WORKFLOW USING OPEN PHACTS CACHEUX Fanny NOVEL DERIVATIVES OF THE TSPO LIGAND DPA-714 FOR IN VIVO OC04 PET IMAGING OF NEUROINFLAMMATION HEIMHALT Maren BACTERIAL GROWTH INHIBITION BY TARGETING DAHP SYNTHASE P023 WITH A RATIONALLY DESIGNED OXIME-BASED INHIBITOR CAMI-KOBECI Gerta IMPROVED ANALGESICS: BU08028 A NOVEL, BIFUNCTIONAL P009 NOP/MOP LIGAND HELLSBERG Eva SELECTIVITY PROFILING OF THE HUMAN MONOAMINE FP10 TRANSPORTERS: A CASE STUDY ON CATHINONE ANALOGS CARVALHO Luís OXO-β-SULTAMS AS ACTIVITY-BASED PROBES P010 HIETANEN Ari INNOVATIVE MEDICINES INITIATIVE CHEM21: IMPLEMENTATION P024 CHATRON Nolan HUMAN VITAMIN K EPOXIDE REDUCTASE (HVKORC1): P011 OF KEY TECHNOLOGIES AT ORION PHARMA MEDICINAL SEARCH FOR THE FUNCTIONAL CONFORMATIONAL STATES CHEMISTRY

CHATZOPOULOU Maria FISHING FOR AN OFF-TARGET": DECONVOLUTING THE FP05 HIGASHINO Masato LEAD OPTIMIZATION OF GABAAa5 RECEPTOR NEGATIVE P025 MOLECULAR MECHANISM OF GPCR-INDUCED MACROPHAGE ALLOSTERIC MODULATORS CHEMOTAXIS" HOGENDORF Adam DETERMINING THE BINDING MODE OF 3-(1-ALKYL-1H-IMIDAZOL-5- FP16 CHIODARELLI Giacomo DEVELOPMENT OF NEW SYNTHETIC INHIBITORS OF P012 YL)-1H-INDOLES MYCOBACTERIUM TUBERCULOSIS DPRE2 HULPIA Fabian PURINE MIMICKING 3'-ETHYNYLRIBOFURANOSE NUCLEOSIDES: CP13 CLUNIE-O'CONNOR DEVELOPING HIGH THROUGHPUT SCREENS TO IDENTIFY SMALL P013 NEW OPPORTUNITIES FOR AN OLD SUGAR MODIFICATION Caitlin MOLECULE ACTIVATORS OF THE ADULT EPICARDIUM IDEMUDIA Omoruyi NEW METAL CHELATING PYRAZOLONE BASED LIGANDS AND P027 COCCO Mattia ACRYLAMIDES AND INFLAMMASOMES: ANOTHER STEP TOWARDS P014 THEIR Co/Cu COMPLEXES OF THERAPEUTIC PROPERTIES DIRECT NLRP3 INHIBITION KAVANAGH Madeline DEVELOPMENT OF POTENT, CELL-ACTIVE INHIBITORS OF THE OC01 COOPER Anna DESIGN, SYNTHESIS AND PHARMACOLOGICAL EVALUATION OF FP06 MYCOBACTERIUM TUBERCULOSIS CHOLESTEROL OXIDASE FLUORESCENT TOOLS FOR STUDYING CANNABINOID TYPE 2 CYP125 RECEPTOR KICKINGER Stefanie MOLECULAR DOCKING OF NOVEL NON-COMPETITIVE hBGT1 P028 DALTON Sam DEVELOPMENT OF NOVEL, SELECTIVE AND IRREVERSIBLE PI3Kὁ FP07 INHIBITORS TO EXPLORE THEIR STRUCTURE-ACTIVITY INHIBITORS RELATIONSHIP

DE LAS HERAS RUIZ SYNTHESIS OF NR ANALOGUES TO INHIBIT GLUCOSE P015 KIHL Henriette PHOTOSWITCHABLE 4-(PIPERIDIN-4-YL)-1-HYDROXYPYRAZOLE FP11 Edgar METABOLISM IN CANCER CELLS ANALOGUES FOR THE GABA TYPE A RECEPTORS

KLIKA SKOPIC Mateja DEVELOPMENT OF SYNTHESIS METHODOLOGY FOR DNA- FP04 ENCODED SMALL MOLECULE SCREENING LIBRARIES 120 ABD ELRAHEEM Eman TOWARD LIBRARIES OF ARTIFICIAL MACROCYCLES FP01 DRACZKOWSKI Piotr EXPERIMENTAL PLATFORM FOR CHARACTERIZATION AND HIGH- CP14 THROUGHPUT SCREENING OF ACETYLCHOLINESTERASE ADORNATO Ilenia IDENTIFICATION OF NOVEL 5-ARYLIDENE-3-PHENYL-2- P001 INHIBITORS. THIOXOIMIDAZOLIDIN-4-ONES AS INHIBITORS OF THE CHYMOTRYPSIN-LIKE ACTIVITIES OF THE CONSTITUTIVE AND DUMA Bogdan TARGETING ASPRS IN THE FIGHT AGAINST M. TUBERCULOSIS P016 IMMUNO-PROTEASOMES FAMILGLINI Valeria DRUG DESIGN, SYNTHESIS AND BIOLOGICAL EVALUATION OF CP02 ADORNATO Ilenia DESIGN AND SYNTHESIS OF DUAL INHIBITORS OF ALDOSE P002 ANTIRETROVIRAL AND ANTICANCER AGENTS REDUCTASE AND PROTEIN TYROSINE PHOPSHATASE 1B FEDOROWICZ Joanna SYNTHESIS AND ANTIBACTERIAL ACTIVITY OF FLUORESCENT P017 AL HARTHY Thuraya DESIGN AND SYNTHESIS OF SOME 2-ARYL-5-FLUORO-6-(4- P003 QUINOLONES METHYL-1-PIPERAZINYL) BENZOTHIAZOLES OF POTENTIAL ANTITUMOR ACTIVITIES FIORILLO Gaetano RECENT ADVANCES IN ANTITUMOUR BERBERINES P018

ALEKSANDROVA Galina CERTAIN COMMON REGULARITIES IN THE CHEMICAL STRUCTURE P004 FREEK Janssen DEVELOPMENT OF HIGHLY SELECTIVE AND REVERSIBLE CP05 OF ANTIHISTAMINES, AND THEIR INFLUENCE ON PROPERTIES DIACYLGLYCEROL LIPASE INHIBITORS

ALEKSANDROVA Galina MODIFICATION OF STRUCTURE OF A P005 GALDEANO Carles DRUGGING THE UNDRUGGABLE: TARGETING CHALLENGING E3 CP04 HYDROXYTRIFLUOROMETHYLINDOLE, FOR THE PURPOSE OF LIGASES INCREASE IN ITS WATER SOLUBILITY GIGANTE Alba IDENTIFICATION OF NOVEL NON-NATURAL SUPRAMOLECULAR FP08 AMIN Mohammad DEVELOPMENT OF STEREOSELECTIVE SYNTHSES AND CHEMICAL P006 LIGANDS AS STABILIZERS OF 14-3-3? PROTEIN-PROTEIN BIOLOGY STUDIES FOR BROMODOMAIN INHIBITORS AS ANTI INTERACTIONS CANCER DRUG GIRAUDO Alessandro SYNTHESIS AND PHARMACOLOGICAL EVALUATION OF NOVEL P019 BALABON Olga DISCOVERY, STRUCTURAL EXPLORATION AND BIOLOGICAL FP02 AMINO BIOISOSTERES FOR THE GABAa RECEPTOR PROFILING OF A NOVEL CLASS OF ANTIMYCOBACTERIAL DPRE1 INHIBITORS GRACE Jake HIGH THROUGHPUT LIBRARY SYNTHESIS IN MEDICINAL P020 CHEMISTRY BOSCA Federica DESIGN OF PORPHYRIN-LOADED NANOBUBBLES FOR P007 THERANOSTIC APPLICATIONS GRINBERGA Signe SYNTHESIS AND PHARMACOLOGICAL EVALUATION OF AMIDINE P021 CONTAINING GABAA RECEPTOR AGONISTS BROCKMEYER Fabian ORALLY BIOAVAILABLE ANTIMALARIAL 4(1H)-QUINOLONE FP03 PRODRUGS WITH SINGLE-DOSE CURES GUR Zehra Tugce NOVEL BRP-7 DERIVATIVES TARGETING FLAP: POTENT P022 INHIBITORS OF LEUKOTRIENE BIOSYNTHESIS BUGNO Ryszard DESIGN, SYNTHESIS AND STRUCTURE-ACTIVITY RELATIONSHIPS P008 OF 1-(ARYLSULFONYL)-1H-INDOLE DERIVATIVES AS NON-BASIC 5- GURINOVA Jana TARGETS IN ORPHAN DISEASES AND THEIR REPURPOSING FP09 HT6 RECEPTOR LIGANDS CANDIDATES - ANALYSIS WITH A KNIME WORKFLOW USING OPEN PHACTS CACHEUX Fanny NOVEL DERIVATIVES OF THE TSPO LIGAND DPA-714 FOR IN VIVO OC04 PET IMAGING OF NEUROINFLAMMATION HEIMHALT Maren BACTERIAL GROWTH INHIBITION BY TARGETING DAHP SYNTHASE P023 WITH A RATIONALLY DESIGNED OXIME-BASED INHIBITOR CAMI-KOBECI Gerta IMPROVED ANALGESICS: BU08028 A NOVEL, BIFUNCTIONAL P009 NOP/MOP LIGAND HELLSBERG Eva SELECTIVITY PROFILING OF THE HUMAN MONOAMINE FP10 TRANSPORTERS: A CASE STUDY ON CATHINONE ANALOGS CARVALHO Luís OXO-β-SULTAMS AS ACTIVITY-BASED PROBES P010 HIETANEN Ari INNOVATIVE MEDICINES INITIATIVE CHEM21: IMPLEMENTATION P024 CHATRON Nolan HUMAN VITAMIN K EPOXIDE REDUCTASE (HVKORC1): P011 OF KEY TECHNOLOGIES AT ORION PHARMA MEDICINAL SEARCH FOR THE FUNCTIONAL CONFORMATIONAL STATES CHEMISTRY

CHATZOPOULOU Maria FISHING FOR AN OFF-TARGET": DECONVOLUTING THE FP05 HIGASHINO Masato LEAD OPTIMIZATION OF GABAAa5 RECEPTOR NEGATIVE P025 MOLECULAR MECHANISM OF GPCR-INDUCED MACROPHAGE ALLOSTERIC MODULATORS CHEMOTAXIS" HOGENDORF Adam DETERMINING THE BINDING MODE OF 3-(1-ALKYL-1H-IMIDAZOL-5- FP16 CHIODARELLI Giacomo DEVELOPMENT OF NEW SYNTHETIC INHIBITORS OF P012 YL)-1H-INDOLES MYCOBACTERIUM TUBERCULOSIS DPRE2 HULPIA Fabian PURINE MIMICKING 3'-ETHYNYLRIBOFURANOSE NUCLEOSIDES: CP13 CLUNIE-O'CONNOR DEVELOPING HIGH THROUGHPUT SCREENS TO IDENTIFY SMALL P013 NEW OPPORTUNITIES FOR AN OLD SUGAR MODIFICATION Caitlin MOLECULE ACTIVATORS OF THE ADULT EPICARDIUM IDEMUDIA Omoruyi NEW METAL CHELATING PYRAZOLONE BASED LIGANDS AND P027 COCCO Mattia ACRYLAMIDES AND INFLAMMASOMES: ANOTHER STEP TOWARDS P014 THEIR Co/Cu COMPLEXES OF THERAPEUTIC PROPERTIES DIRECT NLRP3 INHIBITION KAVANAGH Madeline DEVELOPMENT OF POTENT, CELL-ACTIVE INHIBITORS OF THE OC01 COOPER Anna DESIGN, SYNTHESIS AND PHARMACOLOGICAL EVALUATION OF FP06 MYCOBACTERIUM TUBERCULOSIS CHOLESTEROL OXIDASE FLUORESCENT TOOLS FOR STUDYING CANNABINOID TYPE 2 CYP125 RECEPTOR KICKINGER Stefanie MOLECULAR DOCKING OF NOVEL NON-COMPETITIVE hBGT1 P028 DALTON Sam DEVELOPMENT OF NOVEL, SELECTIVE AND IRREVERSIBLE PI3Kὁ FP07 INHIBITORS TO EXPLORE THEIR STRUCTURE-ACTIVITY INHIBITORS RELATIONSHIP

DE LAS HERAS RUIZ SYNTHESIS OF NR ANALOGUES TO INHIBIT GLUCOSE P015 KIHL Henriette PHOTOSWITCHABLE 4-(PIPERIDIN-4-YL)-1-HYDROXYPYRAZOLE FP11 Edgar METABOLISM IN CANCER CELLS ANALOGUES FOR THE GABA TYPE A RECEPTORS

KLIKA SKOPIC Mateja DEVELOPMENT OF SYNTHESIS METHODOLOGY FOR DNA- FP04 ENCODED SMALL MOLECULE SCREENING LIBRARIES 121 KODAL Anne Louise NEW AVENUES IN PROTEIN LABELING- EXPLOITING DNA FOR CP01 MOULD Daniel DESIGN AND DEVELOPMENT OF POTENT SERIES OF REVERSIBLE P042 Bank DIRECTING CHEMICAL REACTIONS INHIBITORS OF LYSINE SPECIFIC METHYLASE 1 NEWSON Harriet LEAD OPTIMISATION OF A NOVEL CLASS OF TRYPANOSOMACIDAL FP13 KOEBERLE Andreas FUNCTIONAL LIPIDOMICS-BASED LEAD DISCOVERY REVEALS CP06 AGENTS NOVEL ROLE OF PHOSPHOLIPIDS IN VITAMIN A SIGNALING OVER Björn HOW TO DESIGN CELL PERMEABLE NON-PEPTIDIC OC03 KÖNIG Stefanie THE MELLEOLIDE DEHYDROARMELLYLORSELLINATE (DAO) P029 MACROCYCLES INHIBITS 5-LIPOXYGENASE AND DECREASES EICOSANOID BIOSYNTHESIS IN HUMAN LEUKOCYTES PEIN Helmut IMPAIRED FATTY ACID BIOSYNTHESIS DURING APOPTOSIS P043 DECREASES AKT PHOSPHORYLATION THROUGH SPECIFIC LANZ Jan SCREENING OF NATURE BANK AND DIVERSE SCAFFOLD LIBRARY P030 CHANGES IN THE CELLULAR PHOSPHOLIPID COMPOSITION FROM COMPOUNDS AUSTRALIA AGAINST AMPK PEIRO Jorge PRODRUGS SENSITIVE TO REACTIVE OXYGEN SPECIES FOR THE P044 LEIVA-MARTÍNEZ NOVEL 11β-HSD1 INHIBITORS FOR AGE-RELATED COGNITIVE P057 TREATMENT OF CHRONIC INFLAMMATORY DISEASES AND Rosana DISORDERS AND ALZHEIMER’S DISEASE CANCER

LIVENDAHL Madeleine DESIGN AND SYNTHESIS OF 2,2'-DIINDOLYLMETHANES TO CP08 PEREIRA Patricia GLYCO-CONJUGATED PHOTOSENSITIZERS: FROM SYNTHESIS TO CP10 SELECTIVELY TARGET CERTAIN G-QUADRUPLEX DNA VALIDATION IN CANCER PHOTODYNAMIC THERAPY STRUCTURES PITTA Eleni EXTENDED SAR EXPLORATION AND STRUCTURAL OPTIMIZATION P045 LOPEZ CARA Luisa ANTICANCER AND STRUCTURE ACTIVITY RELATIONSHIP OF P031 OF A NOVEL CLASS OF QUINOLOXYACETAMIDE Carlota AMINOPHENOLS CHOLINE KINASE INHIBITORS ANTIMYCOBACTERIAL COMPOUNDS

LOPEZ CARA Luisa ANTICANCER AGENTS BASED ON CIS-RESTRICTED P032 POTSI Maria COMBINING ANTIOXIDANT, ANTIINFLAMMATORY & HYPOLIPIDEMIC P046 Carlota COMBRETASTATIN A-4 ANALOGUES ACTIVITY BY DESIGN: NEW MULTI-POTENT MORPHOLINE DERIVATIVES FOR ATHEROSCLEROSIS LUNVEN Laurent SYNTHESIS AND EVALUATION OF AURONE INTERACTIONS WITH P060 TAU AGGREGATES IN ALZHEIMER'S DISEASE PREVET Hugues NEW 3D-SCAFFOLDS FOR FRAGMENT-BASED LEAD DISCOVERY CP11 (FBLD): EXPLORATION OF CHEMICAL SPACE USING LÖSER Konstantin INHIBITION OF AKT BY POLYUNSATURATED P033 FUNCTIONALIZED SPIROHYDANTOINS. PHOSPHATIDYLCHOLINE – A NOVEL APPROACH FOR POTENTIATING LIPOSOMAL ANTI-CANCER THERAPY PREVITALI Viola SYNTHESIS AND BIOLOGICAL EVALUATION OF DIAROMATIC FP14 GUANIDINIUM-LIKE DERIVATIVES AS PROTEIN KINASE INHIBITORS MALLINGER Aurelie DISCOVERY OF CCT251921: A POTENT, SELECTIVE AND ORALLY OC02 FOR THE TREATMENT OF CANCER BIOAVAILABLE SMALL MOLECULE MODULATOR OF THE MEDIATOR COMPLEX-ASSOCIATED KINASES CDK8 AND CDK19 PREVITI Santo DEVELOPMENT OF NOVEL 1,4-BENZODIAZEPINES AS FP15 ANTITRYPANOSOMAL AGENTS MARIA KIMATRAI NEW MOLECULES DERIVED FROM PREVIOUSLY REPORTED P034 SALVADOR ANTICANCER AGENT WITH POTENT ANTILEUKEMIC ACTIVITY ARE PREVITI Santo DEVELOPMENT OF NOVEL AMIDES AS NON COVALENT P061 INHIBITORS OF STAT5 PHOSPHORYLATION INHIBITORS OF IMMUNOPROTEASOME

MARIA KIMATRAI NEW SYMMETRICAL BISCATIONIC CHOLINE KINASE INHIBITORS P035 PRICE Kathryn ANTIMALARIAL DRUG DISCOVERY: EXPLORING THE MEP CP03 SALVADOR WITH ANTIPLASMODIAL ACTIVITY PATHWAY

MARTINS GOMES Ana A SUSTAINABLE APPROACH TO RECYCLING CLASSIC P036 REES David CASE HISTORIES OF FRAGMENT-BASED DRUG DISCOVERY KL01 Sofia ANTIMALARIALS TOWARDS NEW ANTIPROLIFERATIVE DRUGS ROGACKI Maciej INVESTIGATION OF THE BINDING MODE OF A NOVEL CLASS OF P047 ANTIMYCOBACTERIAL DPRE1 INHIBITORS MATHUR Sunil DESIGN AND EVALUATION OF NOVEL THERANOSTIC P037 FLUOROGENIC DUAL PROBE-PRODRUG IN CANCER RUBIO RUIZ Belen NEAR-INFRARED (NIR) PHOTOTHERMAL THERAPY USING FP20 BIOCOMPATIBLE PALLADIUM NANOPARTICLES MEKER Sigalit STABLE, EFFECTIVE, AND SELECTIVE ANTICANCER PHENOLATO CP09 TITANIUM(IV) COMPLEXES SAINAS Stefano TARGETING THE HUMAN DIHYDROOROTATE DEHYDROGENASE FP17 (hDHODH) BY A SCAFFOLD HOPPING BIOISOSTERIC APPROACH MEXIA Nikitia DESIGN, SYNTHESIS AND BIOLOGICAL EVALUATION OF CP07 USING HYDROXYLATED PENTAATOMIC HETEROCYCLES SELECTIVE CDK INHIBITORS BASED ON FLAVOPIRIDOL STRUCTURE SALEEB Michael HIGH-THROUGHPUT SCREENING FOR INHIBITORS OF P049 PSEUDOMONAS AERUGINOSA ADP-RIBOSYLATING EXOENZYME S MLYNARCZYK Dariusz DENDRIMER-SUBSTITUTED SULFANYL PORPHYRAZINES AS P038 PROMISING PHOTOSENSITIZERS FOR PDT SAVATEEV Konstantin POTENTIAL SYNTHETIC APPROACHES FOR AZOLO[5,1-B]PURINES P050

MLYNARCZYK Dariusz DIAZEPINOPORPHYRAZINES WITH HYPERBRANCHED P039 SLIFIRSKI Grzegorz SYNTHESIS OF NOVEL TETRAHYDRO-4-ARYL-PYRIDO[1,2- P051 SUBSTITUENTS AS POTENTIAL PHOTOSENSITIZERS FOR C]PYRIMIDINE DERIVATIVES WITH DUAL SERT AND 5-HT1A PHOTODYNAMIC THERAPY (PDT) ACTIVITY

MLYNARCZYK Dariusz SYNTHESIS AND PHYSICO-CHEMICAL PROPERTIES OF DENDRITIC P040 SOMMER Roman TOWARDS NONNATURAL CARBOHYDRATE-BASED INHIBITORS OF P052 SULFANYL PORPHYRAZINES OF POTENTIAL APPLICATIONS IN PSEUDOMONAS AERUGINOSA VIRULENCE FACTOR LECTIN LECB NANOMEDICINE STACEY Jessica EXTRACTING ACTIONABLE KNOWLEDGE FROM LARGE SCALE IN P053 MLYNARCZYK Dariusz DENDRIMERIC PHTHALOCYANINE AND PYRAZINOPORPHYRAZINE P041 VITRO PHARMACOLOGY DATA DERIVATIVES AS POTENTIAL PHOTOSENSITIZERS FOR PHOTODYNAMIC THERAPY (PDT) STARON Jakub A NON-BASIC 5-HT6R ANTAGONISTS. P054

MOSTARDA Serena EXTENDING SAR OF BILE ACIDS AS FXR/TGR5 MODULATORS: FP12 THÜRMER Maria SILIBININ INDUCES THE ACCUMULATION OF PHOSPHOLIPIDS IN P055 SYNTHESIS AND BIOLOGICAL ACTIVITY OF RING C MODIFIED HEPATOCYTES AND MOUSE LIVER AND INCREASES THE HEPATIC CDCA DERIVATIVES BIOTRANSFORMATION CAPACITY 122 KODAL Anne Louise NEW AVENUES IN PROTEIN LABELING- EXPLOITING DNA FOR CP01 MOULD Daniel DESIGN AND DEVELOPMENT OF POTENT SERIES OF REVERSIBLE P042 Bank DIRECTING CHEMICAL REACTIONS INHIBITORS OF LYSINE SPECIFIC METHYLASE 1 NEWSON Harriet LEAD OPTIMISATION OF A NOVEL CLASS OF TRYPANOSOMACIDAL FP13 KOEBERLE Andreas FUNCTIONAL LIPIDOMICS-BASED LEAD DISCOVERY REVEALS CP06 AGENTS NOVEL ROLE OF PHOSPHOLIPIDS IN VITAMIN A SIGNALING OVER Björn HOW TO DESIGN CELL PERMEABLE NON-PEPTIDIC OC03 KÖNIG Stefanie THE MELLEOLIDE DEHYDROARMELLYLORSELLINATE (DAO) P029 MACROCYCLES INHIBITS 5-LIPOXYGENASE AND DECREASES EICOSANOID BIOSYNTHESIS IN HUMAN LEUKOCYTES PEIN Helmut IMPAIRED FATTY ACID BIOSYNTHESIS DURING APOPTOSIS P043 DECREASES AKT PHOSPHORYLATION THROUGH SPECIFIC LANZ Jan SCREENING OF NATURE BANK AND DIVERSE SCAFFOLD LIBRARY P030 CHANGES IN THE CELLULAR PHOSPHOLIPID COMPOSITION FROM COMPOUNDS AUSTRALIA AGAINST AMPK PEIRO Jorge PRODRUGS SENSITIVE TO REACTIVE OXYGEN SPECIES FOR THE P044 LEIVA-MARTÍNEZ NOVEL 11β-HSD1 INHIBITORS FOR AGE-RELATED COGNITIVE P057 TREATMENT OF CHRONIC INFLAMMATORY DISEASES AND Rosana DISORDERS AND ALZHEIMER’S DISEASE CANCER

LIVENDAHL Madeleine DESIGN AND SYNTHESIS OF 2,2'-DIINDOLYLMETHANES TO CP08 PEREIRA Patricia GLYCO-CONJUGATED PHOTOSENSITIZERS: FROM SYNTHESIS TO CP10 SELECTIVELY TARGET CERTAIN G-QUADRUPLEX DNA VALIDATION IN CANCER PHOTODYNAMIC THERAPY STRUCTURES PITTA Eleni EXTENDED SAR EXPLORATION AND STRUCTURAL OPTIMIZATION P045 LOPEZ CARA Luisa ANTICANCER AND STRUCTURE ACTIVITY RELATIONSHIP OF P031 OF A NOVEL CLASS OF QUINOLOXYACETAMIDE Carlota AMINOPHENOLS CHOLINE KINASE INHIBITORS ANTIMYCOBACTERIAL COMPOUNDS

LOPEZ CARA Luisa ANTICANCER AGENTS BASED ON CIS-RESTRICTED P032 POTSI Maria COMBINING ANTIOXIDANT, ANTIINFLAMMATORY & HYPOLIPIDEMIC P046 Carlota COMBRETASTATIN A-4 ANALOGUES ACTIVITY BY DESIGN: NEW MULTI-POTENT MORPHOLINE DERIVATIVES FOR ATHEROSCLEROSIS LUNVEN Laurent SYNTHESIS AND EVALUATION OF AURONE INTERACTIONS WITH P060 TAU AGGREGATES IN ALZHEIMER'S DISEASE PREVET Hugues NEW 3D-SCAFFOLDS FOR FRAGMENT-BASED LEAD DISCOVERY CP11 (FBLD): EXPLORATION OF CHEMICAL SPACE USING LÖSER Konstantin INHIBITION OF AKT BY POLYUNSATURATED P033 FUNCTIONALIZED SPIROHYDANTOINS. PHOSPHATIDYLCHOLINE – A NOVEL APPROACH FOR POTENTIATING LIPOSOMAL ANTI-CANCER THERAPY PREVITALI Viola SYNTHESIS AND BIOLOGICAL EVALUATION OF DIAROMATIC FP14 GUANIDINIUM-LIKE DERIVATIVES AS PROTEIN KINASE INHIBITORS MALLINGER Aurelie DISCOVERY OF CCT251921: A POTENT, SELECTIVE AND ORALLY OC02 FOR THE TREATMENT OF CANCER BIOAVAILABLE SMALL MOLECULE MODULATOR OF THE MEDIATOR COMPLEX-ASSOCIATED KINASES CDK8 AND CDK19 PREVITI Santo DEVELOPMENT OF NOVEL 1,4-BENZODIAZEPINES AS FP15 ANTITRYPANOSOMAL AGENTS MARIA KIMATRAI NEW MOLECULES DERIVED FROM PREVIOUSLY REPORTED P034 SALVADOR ANTICANCER AGENT WITH POTENT ANTILEUKEMIC ACTIVITY ARE PREVITI Santo DEVELOPMENT OF NOVEL AMIDES AS NON COVALENT P061 INHIBITORS OF STAT5 PHOSPHORYLATION INHIBITORS OF IMMUNOPROTEASOME

MARIA KIMATRAI NEW SYMMETRICAL BISCATIONIC CHOLINE KINASE INHIBITORS P035 PRICE Kathryn ANTIMALARIAL DRUG DISCOVERY: EXPLORING THE MEP CP03 SALVADOR WITH ANTIPLASMODIAL ACTIVITY PATHWAY

MARTINS GOMES Ana A SUSTAINABLE APPROACH TO RECYCLING CLASSIC P036 REES David CASE HISTORIES OF FRAGMENT-BASED DRUG DISCOVERY KL01 Sofia ANTIMALARIALS TOWARDS NEW ANTIPROLIFERATIVE DRUGS ROGACKI Maciej INVESTIGATION OF THE BINDING MODE OF A NOVEL CLASS OF P047 ANTIMYCOBACTERIAL DPRE1 INHIBITORS MATHUR Sunil DESIGN AND EVALUATION OF NOVEL THERANOSTIC P037 FLUOROGENIC DUAL PROBE-PRODRUG IN CANCER RUBIO RUIZ Belen NEAR-INFRARED (NIR) PHOTOTHERMAL THERAPY USING FP20 BIOCOMPATIBLE PALLADIUM NANOPARTICLES MEKER Sigalit STABLE, EFFECTIVE, AND SELECTIVE ANTICANCER PHENOLATO CP09 TITANIUM(IV) COMPLEXES SAINAS Stefano TARGETING THE HUMAN DIHYDROOROTATE DEHYDROGENASE FP17 (hDHODH) BY A SCAFFOLD HOPPING BIOISOSTERIC APPROACH MEXIA Nikitia DESIGN, SYNTHESIS AND BIOLOGICAL EVALUATION OF CP07 USING HYDROXYLATED PENTAATOMIC HETEROCYCLES SELECTIVE CDK INHIBITORS BASED ON FLAVOPIRIDOL STRUCTURE SALEEB Michael HIGH-THROUGHPUT SCREENING FOR INHIBITORS OF P049 PSEUDOMONAS AERUGINOSA ADP-RIBOSYLATING EXOENZYME S MLYNARCZYK Dariusz DENDRIMER-SUBSTITUTED SULFANYL PORPHYRAZINES AS P038 PROMISING PHOTOSENSITIZERS FOR PDT SAVATEEV Konstantin POTENTIAL SYNTHETIC APPROACHES FOR AZOLO[5,1-B]PURINES P050

MLYNARCZYK Dariusz DIAZEPINOPORPHYRAZINES WITH HYPERBRANCHED P039 SLIFIRSKI Grzegorz SYNTHESIS OF NOVEL TETRAHYDRO-4-ARYL-PYRIDO[1,2- P051 SUBSTITUENTS AS POTENTIAL PHOTOSENSITIZERS FOR C]PYRIMIDINE DERIVATIVES WITH DUAL SERT AND 5-HT1A PHOTODYNAMIC THERAPY (PDT) ACTIVITY

MLYNARCZYK Dariusz SYNTHESIS AND PHYSICO-CHEMICAL PROPERTIES OF DENDRITIC P040 SOMMER Roman TOWARDS NONNATURAL CARBOHYDRATE-BASED INHIBITORS OF P052 SULFANYL PORPHYRAZINES OF POTENTIAL APPLICATIONS IN PSEUDOMONAS AERUGINOSA VIRULENCE FACTOR LECTIN LECB NANOMEDICINE STACEY Jessica EXTRACTING ACTIONABLE KNOWLEDGE FROM LARGE SCALE IN P053 MLYNARCZYK Dariusz DENDRIMERIC PHTHALOCYANINE AND PYRAZINOPORPHYRAZINE P041 VITRO PHARMACOLOGY DATA DERIVATIVES AS POTENTIAL PHOTOSENSITIZERS FOR PHOTODYNAMIC THERAPY (PDT) STARON Jakub A NON-BASIC 5-HT6R ANTAGONISTS. P054

MOSTARDA Serena EXTENDING SAR OF BILE ACIDS AS FXR/TGR5 MODULATORS: FP12 THÜRMER Maria SILIBININ INDUCES THE ACCUMULATION OF PHOSPHOLIPIDS IN P055 SYNTHESIS AND BIOLOGICAL ACTIVITY OF RING C MODIFIED HEPATOCYTES AND MOUSE LIVER AND INCREASES THE HEPATIC CDCA DERIVATIVES BIOTRANSFORMATION CAPACITY 123 TOMASIC Tihomir DISCOVERY OF NOVEL ATPase INHIBITORS OF DNA GYRASE AND CP12 TOPOISOMERASE IV STARTING FROM MARINE SPONGE ALKALOID OROIDIN

TURGUTALP Bengisu SYNTHESIS OF NOVEL BENZOTHIAZOLE-PIPERAZINE P056 DERIVATIVES AND THEIR BIOLOGICAL EVALUATION AS ACETYLCHOLINESTERASE INHIBITORS AND CYTOTOXIC AGENTS

VUORINEN Aini TOWARDS THE ELUCIDATION OF THE MECHANISM OF ACTION OF FP18 SMALL MOLECULE UPREGULATORS OF UTROPHIN USING CHEMICAL PROTEOMICS

WOOD Anthony HETEROCYCLES AND MEDICINAL CHEMISTRY: THE IMPORTANCE KL02 OF INNOVATIVE SYNTHESIS

ZORZI Alessandro DEVELOPMENT OF AN ALBUMIN-BINDING LIGAND FOR FP19 PROLONGING THE PLASMA HALF-LIFE OF PEPTIDE THERAPEUTICS

124 TOMASIC Tihomir DISCOVERY OF NOVEL ATPase INHIBITORS OF DNA GYRASE AND CP12 TOPOISOMERASE IV STARTING FROM MARINE SPONGE ALKALOID OROIDIN

TURGUTALP Bengisu SYNTHESIS OF NOVEL BENZOTHIAZOLE-PIPERAZINE P056 DERIVATIVES AND THEIR BIOLOGICAL EVALUATION AS ACETYLCHOLINESTERASE INHIBITORS AND CYTOTOXIC AGENTS

VUORINEN Aini TOWARDS THE ELUCIDATION OF THE MECHANISM OF ACTION OF FP18 SMALL MOLECULE UPREGULATORS OF UTROPHIN USING CHEMICAL PROTEOMICS

WOOD Anthony HETEROCYCLES AND MEDICINAL CHEMISTRY: THE IMPORTANCE KL02 OF INNOVATIVE SYNTHESIS

ZORZI Alessandro DEVELOPMENT OF AN ALBUMIN-BINDING LIGAND FOR FP19 PROLONGING THE PLASMA HALF-LIFE OF PEPTIDE THERAPEUTICS

List of participants

125

ABD ELRAHEEM Eman GRONINGEN UNIVERSITY, GRONINGEN, NL CHIODARELLI Giacomo UNIVERSITY OF BIRMINGHAM / GLAXOSMITHKLINE, TRES [email protected] CANTOS, ES

[email protected]

ADORNATO Ilenia UNIVERSITY OF MESSINA, MESSINA, IT [email protected] CLOHESSY Thomas GLAXOSMITHKLINE, STEVENAGE, GB

[email protected]

AL HARTHY Thuraya SULTAN QABOOS UNIVERSITY, MUSCAT, OM [email protected] CLUNIE-O'CONNOR Caitlin UNIVERSITY OF OXFORD, OXFORD, GB

[email protected] ALEKSANDROVA Galina STERLITAMAK BRANCH OF THE BASHKIR STATE UNIVERSITY, STERLITAMAK, RUSSIA, STERLITAMAK, RU COCCO Mattia UNIVERSITY OF TURIN, TURIN, IT

[email protected] [email protected]

ALKER David DAVID ALKER ASSOCIATES, BIRCHINGTON, GB COOPER Anna UNIVERSITY OF OTAGO, DUNEDIN, NZ

[email protected] [email protected]

AMIN Mohammad MONASH INSTITUTE OF PHARMACEUTICAL SCIENCES, DALTON Sam UNIVERSITY OF STRATHCLYDE, GLASGOW, GB

MELBOURNE, AU [email protected]

[email protected]

DE LAS HERAS RUIZ Edgar UNIVERSITY OF BIRMINGHAM, BIRMINGHAM, GB

ANDERSEN Nikolaj TECHNICAL UNIVERSITY OF DENMARK, KGS. LYNGBY, DK [email protected]

[email protected] DEYLE Kaycie EPFL, CRISSIER, CH

ATKINSON Ben UNIVERSITY COLLEGE LONDON, LONDON, GB [email protected] [email protected]

DOLLINGER Horst BOEHRINGER INGELHEIM PHARMA, BIBERACH, DE

AUGUSTYNS Koen UNIVERSITY OF ANTWERP, ANTWERP, BE [email protected]

[email protected] DRACZKOWSKI Piotr UNIVERSITY OF LUBLIN, LUBLIN, PL

BALABON Olga UNIVERSITY OF ANTWERP, WILRIJK, BE [email protected]

[email protected] DUMA Bogdan GLAXOSMITHKLINE PLC., TRES CANTOS, ES

BARNIOL-XICOTA Marta UNIVERSITAT DE BARCELONA, BARCELONA, ES [email protected]

[email protected] EDYVANE Holley-Ann MERCK KGAA, DARMSTADT, DE

BOSCA Federica UNIVERSITY OF TORINO, TORINO, IT [email protected] [email protected]

FAMILGLINI Valeria UNIVERSITY OF ROME, ROME, IT

BRAGA MARTINS-TEIXEIRA UNIVERSITY OF SÃO PAULO, RIBEIRÃO PRETO, BR [email protected] Maristela [email protected]

FEDOROWICZ Joanna MEDICAL UNIVERSITY OF GDANSK, GDANSK, PL

BROCKMEYER Fabian NORTHEASTERN UNIVERSITY, BOSTON, US [email protected]

[email protected] FIORILLO Gaetano NAXOSPHARMA SRL, NOVATE MILANESE, IT

BRUNSCHWEIGER Andreas TU DORTMUND, DORTMUND, DE [email protected] [email protected]

FREEK Janssen LEIDEN UNIVERSITY, LEIDEN, NL BUGNO Ryszard POLISH ACADEMY OF SCIENCES, KRAKÓW, PL [email protected]

[email protected] GALDEANO Carles UNIVERSITAT DE BARCELONA, BARCELONA, ES CACHEUX Fanny CEA, ORSAY, FR [email protected]

[email protected] GAMEIRO ROS Isabel Maria UNIVERSIDAD AUTONOMA DE MADRID, MADRID, ES

CAMI-KOBECI Gerta UNIVERSITY OF BATH, BATH, GB [email protected] [email protected]

GIGANTE Alba DUISBURG ESSEN UNIVERSITY, ESSEN, DE

CARVALHO Luís IMED.ULISBOA, SANTARÉM, PT [email protected] [email protected]

GIRAUDO Alessandro UNIVERSITY OF TURIN, TURIN, IT CHATRON Nolan ENS CACHAN, CACHAN, FR [email protected]

[email protected]

GÓMEZ-SANTACANA Xavier VRIJE UNIVERSITEIT AMSTERDAM, AMSTERDAM, NL CHATZOPOULOU Maria UNIVERSITY OF OXFORD, OXFORD, GB [email protected]

[email protected]

126

ABD ELRAHEEM Eman GRONINGEN UNIVERSITY, GRONINGEN, NL CHIODARELLI Giacomo UNIVERSITY OF BIRMINGHAM / GLAXOSMITHKLINE, TRES [email protected] CANTOS, ES

[email protected]

ADORNATO Ilenia UNIVERSITY OF MESSINA, MESSINA, IT [email protected] CLOHESSY Thomas GLAXOSMITHKLINE, STEVENAGE, GB

[email protected]

AL HARTHY Thuraya SULTAN QABOOS UNIVERSITY, MUSCAT, OM [email protected] CLUNIE-O'CONNOR Caitlin UNIVERSITY OF OXFORD, OXFORD, GB

[email protected] ALEKSANDROVA Galina STERLITAMAK BRANCH OF THE BASHKIR STATE UNIVERSITY, STERLITAMAK, RUSSIA, STERLITAMAK, RU COCCO Mattia UNIVERSITY OF TURIN, TURIN, IT

[email protected] [email protected]

ALKER David DAVID ALKER ASSOCIATES, BIRCHINGTON, GB COOPER Anna UNIVERSITY OF OTAGO, DUNEDIN, NZ

[email protected] [email protected]

AMIN Mohammad MONASH INSTITUTE OF PHARMACEUTICAL SCIENCES, DALTON Sam UNIVERSITY OF STRATHCLYDE, GLASGOW, GB

MELBOURNE, AU [email protected]

[email protected]

DE LAS HERAS RUIZ Edgar UNIVERSITY OF BIRMINGHAM, BIRMINGHAM, GB

ANDERSEN Nikolaj TECHNICAL UNIVERSITY OF DENMARK, KGS. LYNGBY, DK [email protected]

[email protected] DEYLE Kaycie EPFL, CRISSIER, CH

ATKINSON Ben UNIVERSITY COLLEGE LONDON, LONDON, GB [email protected] [email protected]

DOLLINGER Horst BOEHRINGER INGELHEIM PHARMA, BIBERACH, DE

AUGUSTYNS Koen UNIVERSITY OF ANTWERP, ANTWERP, BE [email protected]

[email protected] DRACZKOWSKI Piotr UNIVERSITY OF LUBLIN, LUBLIN, PL

BALABON Olga UNIVERSITY OF ANTWERP, WILRIJK, BE [email protected]

[email protected] DUMA Bogdan GLAXOSMITHKLINE PLC., TRES CANTOS, ES

BARNIOL-XICOTA Marta UNIVERSITAT DE BARCELONA, BARCELONA, ES [email protected]

[email protected] EDYVANE Holley-Ann MERCK KGAA, DARMSTADT, DE

BOSCA Federica UNIVERSITY OF TORINO, TORINO, IT [email protected] [email protected]

FAMILGLINI Valeria UNIVERSITY OF ROME, ROME, IT

BRAGA MARTINS-TEIXEIRA UNIVERSITY OF SÃO PAULO, RIBEIRÃO PRETO, BR [email protected] Maristela [email protected]

FEDOROWICZ Joanna MEDICAL UNIVERSITY OF GDANSK, GDANSK, PL

BROCKMEYER Fabian NORTHEASTERN UNIVERSITY, BOSTON, US [email protected]

[email protected] FIORILLO Gaetano NAXOSPHARMA SRL, NOVATE MILANESE, IT

BRUNSCHWEIGER Andreas TU DORTMUND, DORTMUND, DE [email protected] [email protected]

FREEK Janssen LEIDEN UNIVERSITY, LEIDEN, NL BUGNO Ryszard POLISH ACADEMY OF SCIENCES, KRAKÓW, PL [email protected]

[email protected] GALDEANO Carles UNIVERSITAT DE BARCELONA, BARCELONA, ES CACHEUX Fanny CEA, ORSAY, FR [email protected]

[email protected] GAMEIRO ROS Isabel Maria UNIVERSIDAD AUTONOMA DE MADRID, MADRID, ES

CAMI-KOBECI Gerta UNIVERSITY OF BATH, BATH, GB [email protected] [email protected]

GIGANTE Alba DUISBURG ESSEN UNIVERSITY, ESSEN, DE

CARVALHO Luís IMED.ULISBOA, SANTARÉM, PT [email protected] [email protected]

GIRAUDO Alessandro UNIVERSITY OF TURIN, TURIN, IT CHATRON Nolan ENS CACHAN, CACHAN, FR [email protected]

[email protected]

GÓMEZ-SANTACANA Xavier VRIJE UNIVERSITEIT AMSTERDAM, AMSTERDAM, NL CHATZOPOULOU Maria UNIVERSITY OF OXFORD, OXFORD, GB [email protected]

[email protected]

127

GRACE Jake CHEMSPEED TECHNOLOGIES, AUGST, CH KOEBERLE Andreas FRIEDRICH SCHILLER UNIVERSITY JENA, JENA, DE

[email protected] [email protected]

GRACIA Jordi ALMIRALL, BARCELONA, ES KÖNIG Stefanie FRIEDRICH SCHILLER UNIVERSITY JENA, JENA, DE [email protected] [email protected]

GRINBERGA Signe UNIVERSITY OF COPENHAGEN, COPENHAGEN, DK LAMERS Christina EPFL, LAUSANNE, CH

[email protected] [email protected]

GUR Zehra Tugce GAZI UNIVERSITY, ANKARA, TR LANZ Jan UNIVERSITY OF SOUTHERN DENMARK, ODENSE M, DK [email protected] [email protected]

GURINOVA Jana UNIVERSITY OF VIENNA, VIENNA, AT LEIVA-MARTÍNEZ Rosana UNIVERSITY OF BARCELONA, BARCELONA, ES

[email protected] [email protected]

HAUGAARD Anne Stæhr UNIVERSITY OF COPENHAGEN, COPENHAGEN, DK LELONG Bruno MERCK, SAINT QUENTIN FALLAVIER CEDEX, FR

[email protected] [email protected]

HEIMHALT Maren MCMASTER UNIVERSITY, HAMILTON, CA LIVENDAHL Madeleine UMEA UNIVERSITY, UMEÅ, SE

[email protected] [email protected]

HELLSBERG Eva UNIVERSITY OF VIENNA, VIENNA, AT LOPEZ CARA Luisa Carlota UNIVERSITY OF GRANADA, GRANADA, ES

[email protected] [email protected]

HIETANEN Ari ORIONCORPORATION ORION PHARMA, TURKU, FI LORENTE MACIAS Alvaro UNIVERSIDAD DE GRANADA, GRANADA, ES

[email protected] [email protected]

HIGASHINO Masato ONO PHARMACEUTICAL CO.,LTD, OSAKA, JP LUNVEN Laurent UNIVERSITÉ GRENOBLE ALPES, GRENOBLE, FR

[email protected] [email protected]

HOGENDORF Adam INSTITUTE OF PHARMACOLOGY, KRAKÓW, PL LÖSER Konstantin FRIEDRICH SCHILLER UNIVERSITY JENA, JENA, DE

[email protected] [email protected]

HOLLAND Matthew MERCK, MANCHESTER, GB MAHY William UNIVERSITY COLLEGE LONDON, LONDON, GB

[email protected] [email protected]

HULPIA Fabian UGENT, GHENT, BE MALLINGER Aurelie INSTITUTE OF CANCER RESEARCH, SUTTON, LONDON, GB

[email protected] [email protected]

IDEMUDIA Omoruyi UNIVERSITY OF FORT HARE, ALICE, ZA MARIA Kimatrai Salvador UNIVERSIDAD DE GRANADA, GRANADA, ES [email protected] [email protected]

JIMENEZ Niamh F. HOFFMANN-LA ROCHE, BASEL, CH MARTINS GOMES Ana Sofia ICETA - INSTITUTO DE CIÊNCIAS, TECNOLOGIAS E

[email protected] AGROAMBIENTE DA UNIVERSIDADE DO PORTO, PORTO, PT

[email protected] JONES Phil UNIVERSITY OF DUNDEE, DUNDEE, GB

[email protected] MATHUR Sunil EDINBURGH NAPIER UNIVERSITY, EDINBURGH, GB

[email protected] JOSA CULLERE Laia UNIVERSITY OF OXFORD, OXFORD, GB [email protected] MEKER Sigalit THE HEBREW UNIVERSITY OF JERUSALEM, JERUSALEM, IL

[email protected]

JOZWIAK Krzysztof MEDICAL UNIVERSITY OF LUBLIN, LUBLIN, PL

[email protected] MEXIA Nikitia UNIVERSITY OF ATHENS, ATHENS, GR [email protected]

KAVANAGH Madeline UNIVERSITY OF CAMBRIDGE, CAMBRIDGE, GB [email protected] MLYNARCZYK Dariusz POZNAN UNIVERSITY OF MEDICAL SCIENCES, POZNAN;, PL

[email protected] KICKINGER Stefanie UNIVERSITY OF VIENNA, VIENNA, AT

[email protected] MOSTARDA Serena UNIVERSITÀ DEGLI STUDI DI PERUGIA, PERUGIA, IT

[email protected] KIHL Henriette UNIVERSITY OF COPENHAGEN, COPENHAGEN, DK

[email protected] MOULD Daniel CANCER RESEARCH UK MANCHESTER INSTITUTE, MANCHESTER, GB

KODAL Anne Louise Bank NOVO NORDISK, BAGSVÆRD, DK [email protected]

[email protected]

128

GRACE Jake CHEMSPEED TECHNOLOGIES, AUGST, CH KOEBERLE Andreas FRIEDRICH SCHILLER UNIVERSITY JENA, JENA, DE

[email protected] [email protected]

GRACIA Jordi ALMIRALL, BARCELONA, ES KÖNIG Stefanie FRIEDRICH SCHILLER UNIVERSITY JENA, JENA, DE [email protected] [email protected]

GRINBERGA Signe UNIVERSITY OF COPENHAGEN, COPENHAGEN, DK LAMERS Christina EPFL, LAUSANNE, CH

[email protected] [email protected]

GUR Zehra Tugce GAZI UNIVERSITY, ANKARA, TR LANZ Jan UNIVERSITY OF SOUTHERN DENMARK, ODENSE M, DK [email protected] [email protected]

GURINOVA Jana UNIVERSITY OF VIENNA, VIENNA, AT LEIVA-MARTÍNEZ Rosana UNIVERSITY OF BARCELONA, BARCELONA, ES

[email protected] [email protected]

HAUGAARD Anne Stæhr UNIVERSITY OF COPENHAGEN, COPENHAGEN, DK LELONG Bruno MERCK, SAINT QUENTIN FALLAVIER CEDEX, FR

[email protected] [email protected]

HEIMHALT Maren MCMASTER UNIVERSITY, HAMILTON, CA LIVENDAHL Madeleine UMEA UNIVERSITY, UMEÅ, SE

[email protected] [email protected]

HELLSBERG Eva UNIVERSITY OF VIENNA, VIENNA, AT LOPEZ CARA Luisa Carlota UNIVERSITY OF GRANADA, GRANADA, ES

[email protected] [email protected]

HIETANEN Ari ORIONCORPORATION ORION PHARMA, TURKU, FI LORENTE MACIAS Alvaro UNIVERSIDAD DE GRANADA, GRANADA, ES

[email protected] [email protected]

HIGASHINO Masato ONO PHARMACEUTICAL CO.,LTD, OSAKA, JP LUNVEN Laurent UNIVERSITÉ GRENOBLE ALPES, GRENOBLE, FR

[email protected] [email protected]

HOGENDORF Adam INSTITUTE OF PHARMACOLOGY, KRAKÓW, PL LÖSER Konstantin FRIEDRICH SCHILLER UNIVERSITY JENA, JENA, DE

[email protected] [email protected]

HOLLAND Matthew MERCK, MANCHESTER, GB MAHY William UNIVERSITY COLLEGE LONDON, LONDON, GB

[email protected] [email protected]

HULPIA Fabian UGENT, GHENT, BE MALLINGER Aurelie INSTITUTE OF CANCER RESEARCH, SUTTON, LONDON, GB

[email protected] [email protected]

IDEMUDIA Omoruyi UNIVERSITY OF FORT HARE, ALICE, ZA MARIA Kimatrai Salvador UNIVERSIDAD DE GRANADA, GRANADA, ES [email protected] [email protected]

JIMENEZ Niamh F. HOFFMANN-LA ROCHE, BASEL, CH MARTINS GOMES Ana Sofia ICETA - INSTITUTO DE CIÊNCIAS, TECNOLOGIAS E

[email protected] AGROAMBIENTE DA UNIVERSIDADE DO PORTO, PORTO, PT

[email protected] JONES Phil UNIVERSITY OF DUNDEE, DUNDEE, GB

[email protected] MATHUR Sunil EDINBURGH NAPIER UNIVERSITY, EDINBURGH, GB

[email protected] JOSA CULLERE Laia UNIVERSITY OF OXFORD, OXFORD, GB [email protected] MEKER Sigalit THE HEBREW UNIVERSITY OF JERUSALEM, JERUSALEM, IL

[email protected]

JOZWIAK Krzysztof MEDICAL UNIVERSITY OF LUBLIN, LUBLIN, PL

[email protected] MEXIA Nikitia UNIVERSITY OF ATHENS, ATHENS, GR [email protected]

KAVANAGH Madeline UNIVERSITY OF CAMBRIDGE, CAMBRIDGE, GB [email protected] MLYNARCZYK Dariusz POZNAN UNIVERSITY OF MEDICAL SCIENCES, POZNAN;, PL

[email protected] KICKINGER Stefanie UNIVERSITY OF VIENNA, VIENNA, AT

[email protected] MOSTARDA Serena UNIVERSITÀ DEGLI STUDI DI PERUGIA, PERUGIA, IT

[email protected] KIHL Henriette UNIVERSITY OF COPENHAGEN, COPENHAGEN, DK

[email protected] MOULD Daniel CANCER RESEARCH UK MANCHESTER INSTITUTE, MANCHESTER, GB

KODAL Anne Louise Bank NOVO NORDISK, BAGSVÆRD, DK [email protected]

[email protected]

129

NEMEC Vaclav MASARYK UNIVERSITY, BRNO, CZ SOMMER Roman HELMHOLTZ CENTRE FOR INFECTION RESEARCH,

[email protected] SAARBRÜCKEN, DE

[email protected] NEWSON Harriet UNIVERSITY OF WESTERN AUSTRALIA, CRAWLEY, AU

[email protected] STACEY Jessica MEDCHEMICA LIMITED, CHESHIRE, GB

[email protected] OLIVEIRA ALMEIDA Ana UNIVERSITY OF BIRMINGHAM, BIRMINGHAM, GB

Isabel [email protected] STARON Jakub INSTITUTE OF PHARMACOLOGY PAS, KRAKOW, PL

[email protected] OVER Björn MACROCYCLE ADME, BEYOND RO5 AND NATURAL PRODUCT-FRAGMENTS, LANDVETTER, SE THÜRMER Maria UNIVERSITY OF JENA, JENA, DE

[email protected] [email protected]

PEIN Helmut FRIEDRICH-SCHILLER-UNIVERSITY, JENA, DE TOMASIC Tihomir UNIVERSITY OF LJUBLJANA, LJUBLJANA, SI

[email protected] [email protected]

PEIRO Jorge TECHNICAL UNIVERSITY OF DENMARK, KONGENS LYNGBY, TRAORE Tenin DOMAINEX LTD, CAMBRIDGE, GB DK [email protected]

[email protected] TURGUTALP Bengisu YEDITEPE UNIVERSITY, ISTANBUL, TR PEREIRA Patricia UNIVERSIDADE DE AVEIRO, AVEIRO, PT [email protected]

[email protected] UZREK Burhan YEDITEPE UNIVERSITY, ISTANBUL, TR

PITTA Eleni UNIVERSITY OF ANTWERP, ANTWERP, BE [email protected]

[email protected] VALEMBOIS Sophie COPENHAGEN UNIVERSITY, COPENHAGEN, DK POTSI Maria UNIVERSITY OF ATHENS, ATHENS, GR [email protected]

[email protected] VUORINEN Aini UNIVERSITY OF OXFORD, OXFORD, GB

PRASHANTKUMAR Khirsariya MASARYK UNIVERSITY, BRNO, CZ [email protected]

[email protected] WARING Mike UNIVERSITY OF NEWCASTLE, NEWCASTLE UPON TYNE, GB PREVET Hugues LABORATORY U1177 - UNIVERSITY OF LILLE, LILLE, FR [email protected]

[email protected]

WHELDON Mary UNIVERSITY COLLEGE LONDON, LONDON, GB

PREVITALI Viola TRINITY COLLEGE DUBLIN, DUBLIN, IE [email protected] [email protected]

WILDING Birgit THE INSTITUTE OF CANCER RESEARCH, LONDON, GB PREVITI Santo UNIVERSITY OF MESSINA, MESSINA, IT [email protected]

[email protected]

WOOD Anthony PFIZER, CAMBRIDGE, US

PRICE Kathryn UNIVERSITY OF LIVERPOOL, LIVERPOOL, GB [email protected]

[email protected] ZORZI Alessandro ECOLE POLYTECHNIQUE FÉDÉRALE DE LAUSANNE, REES David ASTEX PHARMACEUTICALS, CAMBRIDGE, GB LAUSANNE, CH

[email protected] [email protected]

ROGACKI Maciej UNIVERSITY OF ANTWERP, ANTWERP, BE [email protected]

RUBIO RUIZ Belen UNIVERSITY OF EDINBURGH, EDINBURGH, GB

[email protected]

SAINAS Stefano UNIVERSITA DEGLI STUDI DI TORINO, TORINO, IT [email protected]

SALEEB Michael UMEA UNIVERSITY, UMEA, SE

[email protected]

SAVATEEV Konstantin URFU, EKATERINBURG, RU

[email protected]

SLIFIRSKI Grzegorz MEDICAL UNIVERSITY OF WARSAW, WARSAW, PL

[email protected]

130

NEMEC Vaclav MASARYK UNIVERSITY, BRNO, CZ SOMMER Roman HELMHOLTZ CENTRE FOR INFECTION RESEARCH,

[email protected] SAARBRÜCKEN, DE

[email protected] NEWSON Harriet UNIVERSITY OF WESTERN AUSTRALIA, CRAWLEY, AU

[email protected] STACEY Jessica MEDCHEMICA LIMITED, CHESHIRE, GB

[email protected] OLIVEIRA ALMEIDA Ana UNIVERSITY OF BIRMINGHAM, BIRMINGHAM, GB

Isabel [email protected] STARON Jakub INSTITUTE OF PHARMACOLOGY PAS, KRAKOW, PL

[email protected] OVER Björn MACROCYCLE ADME, BEYOND RO5 AND NATURAL PRODUCT-FRAGMENTS, LANDVETTER, SE THÜRMER Maria UNIVERSITY OF JENA, JENA, DE

[email protected] [email protected]

PEIN Helmut FRIEDRICH-SCHILLER-UNIVERSITY, JENA, DE TOMASIC Tihomir UNIVERSITY OF LJUBLJANA, LJUBLJANA, SI

[email protected] [email protected]

PEIRO Jorge TECHNICAL UNIVERSITY OF DENMARK, KONGENS LYNGBY, TRAORE Tenin DOMAINEX LTD, CAMBRIDGE, GB DK [email protected]

[email protected] TURGUTALP Bengisu YEDITEPE UNIVERSITY, ISTANBUL, TR PEREIRA Patricia UNIVERSIDADE DE AVEIRO, AVEIRO, PT [email protected]

[email protected] UZREK Burhan YEDITEPE UNIVERSITY, ISTANBUL, TR

PITTA Eleni UNIVERSITY OF ANTWERP, ANTWERP, BE [email protected]

[email protected] VALEMBOIS Sophie COPENHAGEN UNIVERSITY, COPENHAGEN, DK POTSI Maria UNIVERSITY OF ATHENS, ATHENS, GR [email protected]

[email protected] VUORINEN Aini UNIVERSITY OF OXFORD, OXFORD, GB

PRASHANTKUMAR Khirsariya MASARYK UNIVERSITY, BRNO, CZ [email protected]

[email protected] WARING Mike UNIVERSITY OF NEWCASTLE, NEWCASTLE UPON TYNE, GB PREVET Hugues LABORATORY U1177 - UNIVERSITY OF LILLE, LILLE, FR [email protected]

[email protected]

WHELDON Mary UNIVERSITY COLLEGE LONDON, LONDON, GB

PREVITALI Viola TRINITY COLLEGE DUBLIN, DUBLIN, IE [email protected] [email protected]

WILDING Birgit THE INSTITUTE OF CANCER RESEARCH, LONDON, GB PREVITI Santo UNIVERSITY OF MESSINA, MESSINA, IT [email protected]

[email protected]

WOOD Anthony PFIZER, CAMBRIDGE, US

PRICE Kathryn UNIVERSITY OF LIVERPOOL, LIVERPOOL, GB [email protected]

[email protected] ZORZI Alessandro ECOLE POLYTECHNIQUE FÉDÉRALE DE LAUSANNE, REES David ASTEX PHARMACEUTICALS, CAMBRIDGE, GB LAUSANNE, CH

[email protected] [email protected]

ROGACKI Maciej UNIVERSITY OF ANTWERP, ANTWERP, BE [email protected]

RUBIO RUIZ Belen UNIVERSITY OF EDINBURGH, EDINBURGH, GB

[email protected]

SAINAS Stefano UNIVERSITA DEGLI STUDI DI TORINO, TORINO, IT [email protected]

SALEEB Michael UMEA UNIVERSITY, UMEA, SE

[email protected]

SAVATEEV Konstantin URFU, EKATERINBURG, RU

[email protected]

SLIFIRSKI Grzegorz MEDICAL UNIVERSITY OF WARSAW, WARSAW, PL

[email protected]

131