Conference Book
Transcription
Conference Book
Sponsors of the DPhG Annual Meeting 2015 MEDI Conference Book I3 – Interactions, Integrations and Innovations Annual Meeting of the German Pharmaceutical Society 2015 – DPhG Printed by: Odenkirchener Druck- und Verlags GmbH Oppelner Straße 6 41199 Mönchengladbach Tel. +49 (0) 2166 - 610295-0 Fax +49 (0) 2166 - 610295-10 Internet: www.o-d-v.de Düsseldorf, Germany, September 2015 ISBN 978-3-9816225-2-2 Annual Meeting of the German Pharmaceutical Society – DPhG Conference Book I3 – Interactions, Integrations and Innovations Düsseldorf, Germany September 23 – 25, 2015 at Heinrich Heine University www.2015.dphg.de Institutional Sponsors Förderer der DPhG-Jahrestagung 2015 Hinweis (FSA-Kodex Fachkreise) Daiichi Sankyo Deutschland GmbH: Umfang der Unterstützung: 1.000 EUR Zweck der Unterstützung: Sponsoring. TABLE OF CONTENTS CONFERENCE COMMITTEES ................................................................................................................................... ii WELCOME ADDRESS ............................................................................................................................................... iii GENERAL INFORMATION ........................................................................................................................................ iv LOCATIONS ................................................................................................................................................................. vi CONFERENCE PROGRAM OVERVIEW.................................................................................................................. 1 1 PLENARY LECTURES............................................................................................................................................ 11 2 SCIENTIFIC LECTURES ....................................................................................................................................... 19 2.1 Antiviral Drugs ......................................................................................................................................... 20 2.2 Blood/Brain Barrier ................................................................................................................................. 26 2.3 Regulation of Beta-Cell Function – Implications for Diabetes ...................................................... 29 2.4 GLISTEN – GPCR Medicinal Chemistry ............................................................................................. 33 2.5 Evidence based Medication Management ......................................................................................... 37 2.6 (Bio)Analytics ........................................................................................................................................... 41 2.7 Allosteric Regulation .............................................................................................................................. 46 2.8 Anti-infective Compounds..................................................................................................................... 50 2.9 Poorly Solubles ........................................................................................................................................ 55 2.10 PPP in Drug Development ..................................................................................................................... 59 2.11 Screening Techniques in Pharmacology & Drug Development .................................................... 63 2.12 Signaling in Cell Death........................................................................................................................... 68 2.13 Anticancer and Epigenetic Drugs ........................................................................................................ 72 2.14 Personalized Medicine – Biomarker and Diagnostics ..................................................................... 77 2.15 Focused Pharmaceutical Research....................................................................................................... 80 2.16 Future Molecular Design ....................................................................................................................... 83 2.17 Medication Safety in Special Patient Groups ................................................................................... 87 2.18 Hot Topics in Pharmaceutical Biology – Young Investigators in the Spotlight ........................ 90 3 SHORT POSTER LECTURES ............................................................................................................................... 97 4 POSTERS .............................................................................................................................................................. 111 4.1 Biopharmaceutics (POS.001-POS.006) ........................................................................................... 112 4.2 Pharmacology (POS.007-POS.014).................................................................................................. 115 4.3 Natural compounds (POS.015-POS.022) ....................................................................................... 119 4.4 Pharmaceutical Technology and drug formulations (POS.023-POS.061) .............................. 122 4.5 Analytics (POS.062-POS.077) ........................................................................................................... 138 4.6 GPCR/Ion channels (POS.078-POS.095)......................................................................................... 144 4.7 Cancer/Epigenetics (POS.096-POS.118)......................................................................................... 151 4.8 Drug design/Medicinal chemistry (POS.119-POS.172)............................................................... 159 4.9 Inflammation (POS.173-POS.184)................................................................................................... 178 4.10 Clinical Pharmacy (POS.185-198) .................................................................................................... 183 4.11 Biotechnology (POS.199-POS.204).................................................................................................. 189 4.12 Other topics (POS.205-POS.214) ..................................................................................................... 192 AUTHOR INDEX ..................................................................................................................................................... 195 DPhG Annual Meeting 2015 Conference Book • i CONFERENCE COMMITTEES Scientific committee: Prof. Dr. Stefan Laufer Prof. Dr. Andreas Link Dr. Olaf Queckenberg Prof. Dr. Christoph Friedrich Prof. Dr. Peter Gmeiner Prof. Dr. Jochen Klein Prof. Dr. Peter Langguth Prof. Dr. Kristina Friedland Prof. Dr. Angelika Vollmar Prof. Dr. Hermann Wätzig Prof. Dr. Thomas Efferth Prof. Dr. Ulrike Holzgrabe Prof. Dr. Ulrich Jaehde Prof. Dr. Heyo Kroemer Prof. Dr. Irmgard Merfort Prof. Dr. Klaus Mohr Prof. Dr. Peter Ruth Prof. Dr. Manfred Schubert-Zsilavecz Prof. Dr. Andrea Sinz Prof. Dr. Holger Stark Prof. Dr. Dieter Steinhilber Prof. Dr. Werner Weitschies Prof. Dr. Gerhard Winter Organization committee: Prof. Dr. Jörg Breitkreuz Dr. German Erlenkamp Dr. Barbara Gioffreda Prof. Dr. Holger Gohlke Dr. Alexandra Hamacher Dr. Finn Hansen Prof. Dr. Matthias Kassack Prof. Dr. Dr.h.c. Peter Kleinebudde Prof. Dr. Thomas Kurz Prof. Dr. Stefanie Läer Prof. Dr. Claus Passreiter Dr. Christopher Pfleger Prof. Dr. Peter Proksch Prof. Dr. Holger Stark Dr. Aleksandra Zivkovic Photo: Heinrich-Heine-Universitaet Duesseldorf Jörg Reich ii • DPhG Annual Meeting 2015 Conference Book WELCOME ADDRESS Dear Colleagues, As President of the German Pharmaceutical Society (Deutsche Pharmazeutische Gesellschaft, DPhG) and as congress Chairman of this meeting it is our pleasure to give you a warm welcome in Düsseldorf attending our Annual Meeting 2015. This year for a very special occasion we have a double anniversary. 125 years for pharmaceutical sciences of the DPhG as well as the founding of the Heinrich Heine University Düsseldorf 50 years ago can be celebrated together with this Annual Meeting. Many things have changed in this long time, but still the different aspects on drugs and drug development in life sciences are in the focus. The motto of this meeting with the important “three I`s” – Interactions, Integrations and Innovations (I3) – still meets the highly interdisciplinary fields in life science for the different pharmaceutical disciplines. The fantastic diversity in subjects ranges from anti-infective drugs, to new chemical entities or natural compounds addressing diabetes, cancer or central diseases. Various topics such as medicinal chemistry on GPCRs, drug design, analytics, biochemistry, pharmacology, drug formulation as well as medication management and personalized medicine will be offered as well as some discussion on regulatory affairs. In this respect, many scientific sessions may have an overlay with other pharmaceutical topics handled in other sessions of the compact and exciting programme demonstrating these exciting “I3”. Beside the science, we hope that you will find some time before or after the congress to enjoy the city of Düsseldorf and the Rhine area since we are in the fortunate position of being able to entertain and surprise our visitors with a wide variety of sights to see. Many thanks to the scientific and organization committees as well as the scientific chairs for the strong support and to the Pharmaceutical Society of Japan (JPS) for the long-lasting partnership. We would also like to thank all participants of the meeting for sharing their recent findings and for their contributions. This abstract book provides all necessary information on the programme as well as on the abstracts of the scientific contributions of plenary lectures, scientific lectures, short poster lectures and posters contributing to fruitful scientific exchange and discussions. Enjoy this meeting, talk to friends as well as to co-operation partners and make new ones. Have a good time. Dieter Steinhilber, President Holger Stark, Chairman DPhG Annual Meeting 2015 Conference Book • iii GENERAL INFORMATION The Annual DPhG Meeting 2015 takes place at the campus of the Heinrich-Heine University, Düsseldorf in building 23.01. LANGUAGE The Conference language is English, no simultaneous translation will be provided. INSTRUCTIONS FOR USING CONFERENCE WLAN If your institution is member of the “eduroam” community, you can use the wireless network “eduroam”. The configuration of your device should be the same as instructed by your home institution. Please use your account and the domain of your home institution. If your institution is not member of the “eduroam” community, you can obtain a guest account and a password at the Conference office. CONFFERENCE OFFICE The Conference office is located at the Conference building 23.01. Opening hours: Wednesday, September 23rd, 2015: 10:00 – 18:00; on the left of lecture hall 3A Thursday, September 24th, 2015: 8:00 – 18:00; Building 23.02. Room U1.24 Friday, September 25th, 2015: 8:30 – 12:00; Building 23.02. Room U1.24 LIABILITY The Organizers of the Conference cannot be held responsible for any loss, theft, damage or injury to any person or property during the Conference, whatever the cause may be. The liability of persons and enterprises providing means of transportations or other services remains unaffected. Each congress participant and accompanying person takes part in all tours at his/her own risk. HHU App For more information about the HHU, please also check the HHU App, freely available for Android and iOS. (For Android) iv • DPhG Annual Meeting 2015 Conference Book (For iOS) ABSTRACT AND POSTER NUMBERS Each abstract has a unique identifier, a letter-number combination. Letters refer to the conference topic a contribution was assigned to (i.e. plenary lectures are identified by the letter “P”, scientific lectures by the letters “SL”, and poster presentations by the letters “POS”). Please note that in case of poster presentations the abstract number is identical with the poster number. Poster abstracts, which are selected for a short poster lectures, were assigned by an additionally letter-number combination and are identified by the letters “SPL”. Please refer to the authors index on page 195 for direct access to specific abstracts. POSTER SESSIONS Topics: Natural compounds, Pharmaceutical Technology and drug formulations, Cancer/Epigenetics, Drug design/Medicinal chemistry, Biotechnology, Biopharmaceutics, Clinical Pharmacy, Inflammation, GPCR/Ion channels, Pharmacology, Analytics, other topics Posters with even poster numbers are assigned to poster session I, while odd poster numbers are assigned to poster session II. Presenting authors are asked to be present at their poster during the poster sessions. Poster session I (even poster numbers) Poster session II (odd poster numbers) Session Wednesday, September 23rd, 2015, 18:00 – 22:00 Thursday, September 24th, 2015, 10:00 – 15:30 Set-up Wednesday, September 23rd, 2015, before 15:00 Thursday, September 24th, 2015, before 10:00 Dismantling Wednesday, September 23rd, 2015, after 22:00 Thursday, September 24th, 2015, after 18:00 CONFERENCE DINNER Separate registration necessary (special fee). Please refer to the Conference office for registration and details. The Conference dinner will take place at “Im Goldenen Ring”, Burgplatz 21, 40213 Düsseldorf. BADGES Badges will be issued to all registered participants and enable access to all scientific sessions. DPhG Annual Meeting 2015 Conference Book • v LOCATIONS Heinrich Heine University campus: The Heinrich Heine University (HHU) campus is located in the south of Düsseldorf, approx. 3.5 km from downtown Düsseldorf and the central station. Arriving by plane: The Düsseldorf airport is approx. 7 km in the north of the center of Düsseldorf. The S-Bahn train station is located bellow terminal C. From here take the S11 (destination “Bergisch Gladbach”) to the Düsseldorf central station. At Düsseldorf central station you can directly travel to the university either by tram (line 707) or Stadtbahn (line U79), destination “Uni-Ost /Botanischer Garten”). Arriving by train: Düsseldorf Central station is connected to all international long-distance routes. From here you can directly travel to the University either by tram (line 707) or Stadtbahn (line U79), destination “UniOst /Botanischer Garten”). Arriving by car: From the East: Coming from the interchange “Hildener Kreuz”, follow the A46 to Düsseldorf. Take the exit toward “D-Zentrum/D-Universität” and follow the B8. At the first traffic lights turn left into Universitätsstrasse. From the West: Coming from the interchange “Neuss-Süd”, follow the A46 to Wuppertal/DüsseldorfSüd. After passing the bridge, crossing the river Rhine, take the exit toward “D-Bilk/D-Zentrum/DHafen/D-Benrath”. Stay on the right lane and follow the signs to “D-Benrath” (Münchner Straße). Take the exit D-Himmelgeist and follow the signage to the University. The university has a lot of free parking places (except for the clinics). Please note that all parking offenders on university premises will be towed away. Heinrich Heine University Campus Building 23.01, Universitätsstr. 1 (how to get to the HHU) vi • DPhG Annual Meeting 2015 Conference Book Haus der Universität: The Haus der Universität is located in downtown Düsseldorf (northern end of the Königsallee), approx. 3.5 km from the Heinrich Heine University (HHU) Arriving from the Heinrich Heine University by public transport: • Stadtbahn (line U79): from the station “Uni-Ost /Botanischer Garten” at the University to the Heinrich-Heine-Allee (downtown) Arriving by car: The Haus der Universität is located in the precinct and cannot be reached directly by car. Please take some extra time for arriving, due to several construction work in this area. Regular parking slots in this area are rare and it is advisable to use one of the following car parks on your own costs: • „Schadow Arkaden“; Martin-Luther-Platz 28; daily 7:00 – 1:00 • „Kö-Bogen“; Königsallee 2: daily 6:00 – 1:00 • „Düsseldorfer Schauspielhaus“; Gustaf-Gründgens-Platz 1; daily 6:00 – 24:00 • „Kaufhof an der Kö“; Königsallee 1 – 9; Mon – Sat 9:00 – 20:00 Haus der Universität (how to get to the “Haus der Universität”) DPhG Annual Meeting 2015 Conference Book • vii CONFERENCE PROGRAM OVERVIEW Pre-Meeting Program Tuesday, September 22nd Bürgersymposium: Geschichte der Pharmazie in Düsseldorf Ort: Haus der Universität, Düsseldorf Schadowplatz 14 14:00 – 14:15 Begrüßung durch den Vorsitzenden der FG Geschichte der Pharmazie Prof. Dr. Christoph Friedrich, Marburg 14:15 – 15:00 Zur Entwicklung des Hochschulfaches Pharmazie an der Heinrich-Heine-Universität Düsseldorf Prof. Dr. Horst Weber, HHU Düsseldorf 15:00 – 15:45 Zur Entwicklung des Apothekenwesens in Düsseldorf Dr. Frederik Vongehr, Mörs 15:45 – 16:15 Kaffeepause 16:15 – 17:00 Vesters Archiv – eine besondere pharmaziehistorische Quelle aus Düsseldorf im Spannungsfeld privater, städtischer und universitärer Interessen Prof. Dr. Frank Leimkugel, HHU Düsseldorf 17:00 – 17:45 Zur Geschichte des Düsseldorfer Aqua-Zoos und seines Begründers, Apotheker Theodor Löbbecke (1821–1901) Sandra Honigs, Aqua-Zoo – Löbbecke-Museum Düsseldorf 19:00 – 21:00 Treffen Arbeitsgemeinschaft Katastrophenpharmazie Ort: Haus der Universität, Düsseldorf, Schadowplatz 14 20:00 – 21:00 Notfall- und Katastrophenpharmazie - Ein Beitrag der Apotheker zum Bevölkerungsschutz Daniel Neuser, Düsseldorf DPhG Annual Meeting 2015 Conference Book • 1 CONFERENCE PROGRAM OVERVIEW Wednesday, September 23rd Meetings der DPhG-Fachgruppen 9:00 – 11:00 11:30 – 13:00 Sitzung VdPPHI e.V. Hochschullehrersitzung, B. Clement (HS 3A) Fachgruppe Pharm./Med. Chemie P. Gmeiner (HS 3A) Fachgruppe Pharm. Biologie A. Vollmar (HS 3D) Fachgruppe Pharmakologie J. Klein (HS 3E) Fachgruppe Pharm. Technologie P. Langguth (HS 3B) Fachgruppe Klinische Pharmazie K. Friedland (HS 3F) Fachgruppe Industriepharmazie C. Küster (U1.23) Wednesday, September 23rd Main Symposium (Congress language English) 13:00 – 13:30 Opening of the Annual DPhG Meeting 2015 (HS 3A) I3 – Interactions, Integrations and Innovations 13:30 – 14:15 Plenary lecture, Christa E. Müller, Interactions, Integrations and Innovations in medicinal chemistry of purinergic signaling (HS 3A) 14:15 – 15:00 Plenary lecture, Jörg Breitkreutz, New concepts and products for individual drug dosing (HS 3A) P.2 15:00 – 15:30 Coffee break - Poster viewing (even numbers) P.1 SHORT TALKS 15:30 – 17:00 SL1 (HS 3A) SL2 (HS 3D) SL3 (HS 3E) Antiviral Drugs Chairs: H. Rübsamen-Schaeff, H. Zimmermann Blood/Brain Barrier Chairs: G. Fricker, F. Helm Regulation of beta-Cell Function – Implications for Diabetes Chairs: G. Drews, E. Oetjen 15:30 SL.01 Thomas Pietschmann: Directly acting antivirals against hepatitis C virus – close to “perfectovir” 15:30 SL.07 Robin Tremmel: Liposomal delivery of Trientine for treatment of Wilson’s Disease in the brain 15:30 SL.10 Gisela Drews: Role of bile acids in beta-cell function 15:45 SL.02 Daniela Paulsen: How to deal with HBV? - State of the art and new concepts in HBV therapy 16:00 SL.03 Christoph Stephan: Advances and challenges in diagnostics & therapy for HIV/AIDS 15:50 SL.11 Ingo Rustenbeck: Mobility of submembrane SL.08 insulin granules 16:00 Stefan Liebner: Molecular regulation of endothelial barrier properties in the central nervous system 16:15 SL.04 Helga Rübsamen-Schaeff: The era after nucleosides: Novel therapeutic approaches against herpes viruses 16:15 SL.12 Martina Düfer: Reactive oxygen species: Pharmacological target or physiological prerequisite for beta-cell function? continued on the next page 2 • DPhG Annual Meeting 2015 Conference Book CONFERENCE PROGRAM OVERVIEW 16:30 SL.05 Stephan Ludwig: New avenues in anti-influenza therapy 16:45 SL.06 Johan Neyts: Antivirals, a lot has been achieved, yet a long way to go 16:30 SL.09 Marise Kolter: Toxicity of poly(n-butylcayanacrylate) (PBCA) nanoparticles designed to overcome the blood brain barrier 17:00 – 18:00 Short poster lectures (HS 3A) 17:00 – 18:00 Freunde der DPhG (building 23.02, room U1.23) 18:00 – 22:00 Poster viewing (even numbers) and Welcome Reception 16:35 SL.13 Elke Oetjen: Regulation of beta-cell function and mass by the dual leucine zipper kinase DPhG Annual Meeting 2015 Conference Book • 3 CONFERENCE PROGRAM OVERVIEW Thursday, September 24th 8:30 – 9:15 Plenary lecture, Martin Biel, Ebola and more: Endolysosomal cation channels as novel drug targets (HS 3A) P.3 9:15 – 10:00 Plenary lecture, Robin Thurmond, The pre-clinical and clinical development of histamine H4 receptor antagonists (HS 3A) P.4 10:00 – 10:30 Coffee break - Poster viewing (odd numbers) SHORT TALKS 10:30 – 12:00 SL4 (HS 3A) GLISTEN - GPCR Medicinal Chemistry Chairs: N. Tschammer, P. Kolb SL5 (HS 3D) SL6 (HS 3E) Evidence Based Medication Management Chairs: G. Hempel, S. Läer 10:30 SL.14 10:30 Francine Acher: Georg Hempel: Introduction Selective orthosteric agonists in 10:35 SL.18 class C GPCRs Thilo Bertsche: Medication management in clinical care 11:00 SL.15 Hans Bräuner-Osborne: Identification and pharmacological characterization of endogenous and surrogate ligands for orphan G proteincoupled receptors 10:30 SL.22 Kai Stühler: Protein mass spectrometry – Methods and applications in clinical proteomics 10:50 SL.23 Gerhard Scriba: Capillary electrophoresis-based SL.19 stereospecific enzyme assay for methionine sulfoxide reductase 11:00 Olaf Rose: Medication management in ambulatory Care: Preliminary results of the WestGem-Study 11:10 SL.24 Shigeru Ohta: New approach for drug discovery and development: Prediction of human drug metabolism using chimeric mice transplanted with human SL.20 hepatocytes 11:25 Stefan Derix: 11:30 SL.16 AMTS activities of the Sylwia Gawron: Pharmacists-Chamber of North Structure free optimization of Rhine fragments for the β2-adrenoreceptor 11:45 SL.17 Matthias Hillenbrand: Comprehensive analysis of heterotrimeric G-protein complex diversity and their interactions with GPCRs in solution (Bio)Analytics Chairs: J. Heilmann, M. Lämmerhofer 11:45 SL.21 Judith Hildebrand: Comparison of three lists of Potentially Inadequate Medications in old age (PIMs) in four German long-term care facilities with special regard to Adverse Drug Events (ADEs) (organized by GLISTEN COST Action CM1207) 4 • DPhG Annual Meeting 2015 Conference Book 11:30 SL.25 Ryoichi Fujiwara: Importance of extrahepatic UDPglucuronosyltransferase 1A1 in bilirubin metabolism 11:45 SL.26 Maria K. Parr: Renaissance of supercritical fluid chromatography: Fast and sensitive analysis of polar drugs and their metabolites by hyphenated mass spectrometry CONFERENCE PROGRAM OVERVIEW Thursday, September 24th 12:00 – 13:30 Break for lunch time - Poster viewing (odd numbers) SHORT TALKS 13:30 – 15:00 SL7 (HS 3A) SL8 (HS 3D) Allosteric Regulation Chairs: K. Mohr, M. Bünemann Anti-Infective Compounds Chairs: U. Holzgrabe, P. Proksch Poorly Solubles Chairs: P. Kleinebudde, W. Weitschies 13:30 SL.27 Ursula Storch: Molecular insights into the mechanosensivity of histamine H1 receptors 13:30 SL.31 Tanja Schirmeister: Vinylsulfone-based inhibitors of rhodesain as new antitrypanosomal compounds 13:30 SL.36 Susanne Page: The use of screening tools in the development of amorphous solid dispersions 13:55 SL.28 Nuska Tschammer: 14:00 SL.32 Biased signaling and probe Christian Klein: dependence at the chemokine Discovery of dengue protease receptor CXCR3 inhibitors with nanomolar affinity 14:00 SL.37 Heike Bunjes: Colloidal carrier systems for the formulation of poorly watersoluble drugs 14:15 SL.33 Alexander Titz: 14:20 SL.29 The lectin LecB as target for anti-infectives against chronic Wiebke Seemann: Pseudomonas aeruginosa Muscarinic M2 receptor infections allosterism: Context-sensitive signaling 14:30 SL.34 Rainer Kalscheuer: Chlorflavonin inhibits growth of Mycobacterium tuberculosis by targeting branched-chain amino acid biosynthesis 14:45 SL.30 14:45 SL.35 Susanne Hermans: Georgios Daletos: Detection of unexplored Marine natural products as allosteric pockets using a potential sources for new “dummy” ligand approach antitubercular agents 15:00 – 15:30 SL9 (HS 3E) 14:30 SL.38 Michael Hacker: Oligomeric cross-linkers for hydrogel formulation from ECMderived macromolecules 14:45 SL.39 Dominique Lunter: Contribution of Confocal Raman Microscopy (CRM) to the validation of an ex vivo skin penetration method Coffee break - Poster viewing (odd numbers) DPhG Annual Meeting 2015 Conference Book • 5 CONFERENCE PROGRAM OVERVIEW Thursday, September 24th SHORT TALKS 15:30 – 17:00 SL10 (HS 3A) SL11 (HS 3D) SL12 (HS 3E) PPP in Drug Development Chairs: S. Knapp, F. Bracher Screening Techniques in Pharmacology & Drug Development Chairs: E. Kostenis, H. Wätzig Signaling in Cell Death Chairs: I. Merfort, T. Efferth 15:30 SL.40 Stefan Knapp: A PPP for the development of chemical tool compounds 15:30 SL.44 Hermann Wätzig: Data quality in drug discovery – The role of analytical performance in ligand binding assays 15:30 SL.49 Min Li-Weber: Sensitization of the anti-cancer efficacy of the Bcl-2 family inhibitor ABT-263 by natural compounds 15:50 SL.45 Ramona Schrage: Dynamic mass redistribution to 16:00 SL.41 probe the signaling repertoire of 16:00 SL.50 GPCRs Anke Müller-Fahrnow: Beatrice Bachmeier: Public Private Partnership in miR181b is included by the lead discovery: Overview and 16:10 SL.46 chemopreventive polyphenol case study on binding kinetics curcumin and inhibits breast Manuel Grundmann: cancer metastasis via downLabel-free biosensors help to regulation of the inflammatory reveal a new mechanism of cytokines CXCL1 and 2 GPCRs activation 16:20 SL.42 Conrad Kunick: Development of a selective DYRK1A inhibitor in a PPP framework 16:20 SL.47 Manuel Koch: Assay technologies addressing GPCRs in drug discovery 16:30 SL.51 Qiaoli Zhao: Shikonin and its derivatives inhibit phosphorylation of the epidermal growth factor receptor signaling and synergistically kill glioblastoma cells in combination with SL.43 erlotinib 16:40 Olaf Kelber: The role of adenosine in colonic inflammation – A study in rat colon preparations in vitro 16:45 SL.48 Frank Böckler: Semi-automatic fluorescence anisotropy titrations (saFLAT) enhance screening throughput and data quality 6 • DPhG Annual Meeting 2015 Conference Book 16:45 SL.52 Bertan Bopp: A novel autodisplay based screening assay for the identification of small molecules that inhibit the dimerization of human chaperone Hsp90 CONFERENCE PROGRAM OVERVIEW Thursday, September 24th SHORT TALKS 17:00 – 18:30 SL13 (HS 3A) Anticancer and Epigenetic Drugs Chairs: M. Kassack, M. Jung SL14 (HS 3D) SL15 (HS 3E) Personalized Medicine – Biomarker and Diagnostics Chairs: O. Queckenberg, T. Dingermann 17:00 SL.53 17:00 SL.58 Daniel Rauh: Reinhard Ortmann: Targeted cancer therapies The challenge of leverage genetic biomarkers from research level to routine clinical IVD testing 17:20 SL.54 Sebastian Wesselborg: Role of apoptosis signaling in tumorigenesis and therapy 17:30 SL.59 resistance Frank Kramer: The potential of biomarkers to 17:40 SL.55 support decision making in clinical studies in cardiovascular Stefan Günther: A novel CREBBP/p300 inhibitor indications and its molecular effects in cancer cells 18:00 SL.56 Christian A. Olsen: Macrocyclic inhibitors of human histone deacetylase enzymes 18:20 SL.57 Steffen Lüdeke: Correlation of conformation with cytotoxic activity of Focused Pharmaceutical Research Chairs: S. Laufer, D. Steinhilber 17:00 SL.61 Dieter Steinhilber: Project group translational medicine and pharmacology, Fraunhofer IME 17:25 SL.62 Christa E. Müller: Neuroallianz consortium – Example of an academicindustrial collaboration 17:50 SL.63 Stefan Laufer: The Interfaculty Centre for SL.60 Pharmacogenomics and Pharma Research (ICEPHA) 18:00 Georg Lautscham: From “omics”-technologies to stratified medicine in autoimmune diseases 18:15 General discussion α-aminoxy oligopeptides: A circular dichroism study 17:00 – 18:30 Workshop, T. Hotopp, Tipps und Hinweise für eine erfolgreiche Antragstellung (HS 3B) 19:30 Conference dinner (Im Goldenen Ring, Burgplatz 21, 40213 Düsseldorf) DPhG Annual Meeting 2015 Conference Book • 7 CONFERENCE PROGRAM OVERVIEW Friday, September 25th 8:30 – 9:15 Plenary lecture, Dieter Häussinger, Liver research in Düsseldorf: Bridging basic and clinical science HS 3A) P.5 9:15 – 10:00 Plenary lecture, Karl Broich, Drug approval and regulatory science – Where to go? (HS 3A) P.6 SHORT TALKS 10:30 – 12:00 SL16 (HS 3A) SL17 (HS 3D) SL18 (HS 3E) Future Molecular Design Chairs: G. Schneider, H. Gohlke Medication Safety in Special Patient Groups Chairs: K. Friedland, U. Jaehde Hot Topics in Pharmaceutical Biology – Young Investigators in the Spotlight Chairs: S. Alban, A. Vollmar 10:30 SL.64 Andrew Hopkins: Automated design of bispecific small molecule drugs 10:30 SL.68 Stephan Scherneck: Medication safety of pregnant and lactating women 10:30 SL.71 Kristian Wende: Cold atmospheric plasma – A future therapeutic approach? Bioanalytics as route to fundamental understanding 10:45 SL.72 Jandirk Sendker: About the potential of plant senescence as a new source for drug discovery 11:00 SL.65 Tina Ritschel: KRIPO-Protein binding site similarities for drug design 11:00 SL.69 Stefanie Läer: Medication for neonates, infants and children 11:20 SL.66 Johannes Kirchmair: Predicting the sites and products of drug metabolism 11:30 SL.70 Ulrich Jaehde: Medication safety of elderly 11:40 SL.67 patients in nursing homes Gisbert Schneider: Enforcing drug discovery by computational molecular design 11:00 SL.73 Alexander Kristian Apel: Regulation of the heterologously expressed novobiocin gene cluster by the host strain Streptomyces coelicolor M512 11:15 SL.74 Sonja Keßler: mRNA binding proteins in metaflammation and hepatocarcinogenesis 11:30 SL.75 Timo Niedermeyer: Cyanobacteria in natural product research 11:45 SL.76 Till Schäberle: Analysis of natural product – Biosynthesis in the post-genomic era 12:00 – 13:00 Short lunch break 13:00 – 13:45 Plenary lecture, Piet Herdewijn, From synthetic nucleic acids to artificial genes and genomes: Drugs for the future? (HS 3A) P.7 14:00 – 15:00 Closing ceremony (HS 3A) 15:15 – 16:30 DPhG Jahreshauptversammlung (HS 3A) 8 • DPhG Annual Meeting 2015 Conference Book CONFERENCE PROGRAM OVERVIEW Post-Meeting Program Saturday, September 26th 15:00 – 18:30 Tag der Offizinpharmazie in Kooperation mit der Deutschen Gesellschaft für Schmerzmedizin, zentrale Fortbildung der DPhG Regionalgruppe Nordrhein und FG Allgemeinpharmazie mit LAK NR Ort: Heinrich-Heine-Universität, Universitätsstr. 1, 40225 Düsseldorf, Hörsaal 3D, Gebäude 23.01 PD. Dr. med. Michael Überall, Nürnberg, Präsident der Deutschen Schmerzliga e.V Erster Teil der Schmerzschulung 1. Informationen zur „Deutschen Gesellschaft für Schmerzmedizin“ und der „Deutschen Schmerzliga“ Die Versorgungssituation des Schmerzpatienten in Deutschland 2. Grundlagen Schmerz (1) Definition (akut vs. chronisch, etc.), Strukturen des nozizeptiven Systems, endogene Schmerzkontrollmechanismen, Pathophysiologie acuter vs- chronischer Schmerzen 3. Grundlagen Schmerz (2) Epidemioligie, Klassifikation, Schmerz als Symptopm vs. Schmerz als Krankheit, bio-psycho-soziales Krankheitsmodell, Begleiterscheinungen, Ko-Morbidität, etc. 4. Differentialdiagnostik Anamnese/ Diagnostik, Verlauf, Chronofizierungsrisiken, Grundlagen der Prävention und Therapie akuter/chronischer Schmerzen, Schmerzmessung, Selbstauskunftinstrumente, etc. 5. Ziele der (medikamentösen) Schmerztherapie Schmerzlinderung, Funktionsverbesserung, Teilhabe an den Aktivitäten des (all)täglichen Lebens, Lebensqualität, individuelle Behandlungs-ziele, etc. 6. Schmerztherapie – WHO-Stufe 1 (Nichtopioide) Paracetamol, Ibuprofen, ASS, Diclofenac, Coxibe, Metamizol, etc.; jeweils mit WM, UAW, KI (Schwerpunkt auf apothekenpflichtige Fertigarzneimittel und Beratungs-/ Verkaufssituationen in der Apotheke) ANMELDUNG Anmeldung zum Tag der Offizinpharmazie in Kooperation mit der Deutschen Gesellschaft für Schmerzmedizin Kursart: Vortrag mit TED Prüfung Teilnehmergebühren 1. Teil: • Kostenfrei Bitte melden Sie sich bzw. Ihre Apotheke zum Tag der Offizinapotheke online an: www.dphg.de/apo15 Teilnehmergebühren 2. Teil: Zum Erwerb des Zertifikats: • 190 € (zzgl. MwSt) Anmeldung für den 2. Kurs über die DGS vor Ort oder über: www.dgschmerzmedizin.de/dgs_campus.html Möglichkeit und Kosten der Rezertifizierung: • Jährlich: 38 € (zzgl. MwSt) • DPhG-Mitgliedsapotheken: 28 € (zzgl. MwSt) DPhG Annual Meeting 2015 Conference Book • 9 10 • DPhG Annual Meeting 2015 Conference Book 1 PLENARY LECTURES DPhG Annual Meeting 2015 Conference Book • 11 PLENARY LECTURES Interactions, Integrations and Innovations in medicinal chemistry of purinergic signaling P.1 Müller, C. E.1 1 PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical Chemistry I, University of Bonn, Bonn, Germany Membrane receptors activated by purines are subdivided into three distinct families: (i) nucleotide or P2 receptors, further divided into G protein-coupled P2Y receptors and ATP-gated ion channel P2X receptors; (ii) adenosine or P1 receptors (A1, A2A, A2B, A3), and (iii) P0 receptors activated by the nucleobase adenine [1,2]. Purine receptors are widely distributed in the body. Their important role in signal transduction is increasingly recognized and appreciated, and their potential as drug targets is explored and exploited with growing success. The physiological ligands of the three classes of receptors, ATP/ADP, adenosine, and adenine, are metabolically linked, and enzymes interconverting them, in particular ectonucleotidases, are fine-tuning purinergic signalling. Tool compounds for a wide range of membrane proteins involved in purinergic signalling have been developed in the past decades. Recent successful efforts of our group have focused on the development of novel assays [3-5] and structure-based approaches [6-8] to identify and optimize P2 receptor antagonists and ectonucleotidase inhibitors [9-11]. Potent and selective tool compounds are crucial for elucidating the (patho) physiological roles of purinergic signalling [12-14]. References: 1. Fredholm, B. B.: Pharmacol. Rev. 2011, 63, 1-34. 2. Müller, C. E.: Curr. Med. Chem. 2015, 22, 929-941. 3. Freundlieb, M. et al.: Anal. Biochem. 2014, 446, 53-58. 4. Lee, S. Y.; Müller C.E.: Electrophoresis 2014, 35, 855–863. 5. Fiene, A. et al.: Analyst 2015, 140, 140-148. 6. Zebisch, M. et al. J. Struct. Biol. 2014, 185, 336-341. 7. Zhang, K. et al.: Nature 2014, 509, 115-118. 8. Zhang, J. et al.: Nature 2014, 509, 119-122. 9. Chang, L. et al. J. Med. Chem. 2014, 57, 10080-100100. 10. Lee, S. et al: Biochem. Pharmacol. 2015, 93, 171–181. 11. Bhattarai, S. et al. J. Med. Chem. 2015, 58(13), 6248-6263. 12. Gnad, T. et al.: Nature 2014, 516, 395-399. 13. Sassi, Y. et al. J. Clin. Invest. 2014, 124, 5385-5397. 14. Kaster, M. P. et al.: Proc. Natl. Acad. Sci. 2015, 112(25), 7833-7838. 12 • DPhG Annual Meeting 2015 Conference Book PLENARY LECTURES New concepts and products for individual drug dosing P.2 Breitkreutz, J.1 1 Institute of Pharmaceutics and Biopharmaceutics, Heinrich Heine University Düsseldorf, Universitätsstr. 1, 40225 Düsseldorf, Germany Recent progress in genetic and metabolic profiling, pharmaceutical research and medical knowledge has led to improved drug therapies with precise dosing recommendations for medicinal products. Many experts see so called ‘Personalized Medicine’ as the future drug therapy for all patients. However, there are no appropriate drug dosage forms available which match these requirements, e.g. tablets and capsules are mostly provided with only one or two different dose strengths. Splitting is often no option due to the resulting uneven pieces with incorrect doses, loss of drug by dust forming, or destroying functional polymer films. Especially for the paediatric and geriatric patients many medicinal products are inappropriate when taking the specific requirements of these populations into account [1]. Recent innovations in pharmaceutical technology and medical device industry show exciting advances and promise to overcome the present gap of patient-adapted, individualized dosing options. Small-sized tablets (< 3 mm in diameter), so called mini-tablets, can be designed as orodispersible or dispersible drug formulations with very small dosing steps [2]. Most recently, it has been demonstrated by our group in a series of three clinical studies that these mini-tablets can be taken as good or even better than a sweet syrup by all paediatric patients, including pre-term and term neonates [3]. Orodispersible films are another option for precise and individual dosing. Some prescriptional drug products with this technology has already entered the market, but more innovations are needed for individual dosing. Film preparation can be performed under the conditions of community or hospital pharmacies and show some advantages such as better content uniformity over conventional capsules preparation [4]. Another strategy are innovative medical device enabling the cutting of film strips according to the prescribed dose. Printing technologies using two- or three-dimensional printers and drug-loaded inks are currently under investigations in various labs for enabling individualized doses and personalized medicines [5]. It can be foreseen that printed medicines will replace many conventional dosage forms in industrial processing, but also compounding in hospital or community pharmacies. Furthermore, individualized printed medicines open completely new treatment options in surgery, cancer, dental medicine and many more medical disciplines. Scientific pharmacists should quickly explore these technologies and take the lead regarding drug development, production, quality control and regulatory sciences in this fascinating area of technology. References: 1. Breitkreutz, J.; Boos, J.: Exp. Opin. Drug Deliv. 2007, 4: 37-45. 2. Stoltenberg, I.; Breitkreutz, J.: Eur. J. Pharm. Biopharm. 2011, 78: 462-469. 3. Klingmann, V. et al.: J. Pediatr. 2015, in press. 4. Visser, J. C. et al.: Int. J. Pharm. 2015, 478: 155-163. 5. Preis, M. et al.: Int. J. Pharm. 2015, in press. DPhG Annual Meeting 2015 Conference Book • 13 PLENARY LECTURES Ebola and more: Endolysosomal cation channels as novel drug targets P.3 Biel, M.1 1 Center for Integrated Protein Science Munich CiPSM at the Department of Pharmacy – Center for Drug Research, Ludwig-MaximiliansUniversität München, Butenandtstr. 5-13, 81377 Munich, Germany Ion channels are transmembrane proteins that confer the flux of ions across lipid membranes of the cell. In the past, the major focus of ion channel research has been on channels present in the plasma membrane and several members of these channels have been exploited as drug targets. More recently, ion channels that are localized in the membrane of intracellular organells have attracted interest. Importantly, a growing number of these channels has been linked to the pathologies of human diseases making them interesting candidates for drug development. In my lecture, I will give an overview on our recent work on so-called two-pore channels (TPC1 and TPC2) [1-3]. These channels are exclusively expressed in endolysosomal organells. To facilitate functional characterization of TPCs in their native environment (i.e. the lysosomal membrane) we have applied novel electrophysiological techniques. Moreover, knockout mouse models for TPCs have been generated. We found that TPC2 plays a key role in endolysosomal trafficking and, thereby, is involved in the life cycle control of many cellular receptors including EGF-R, LDL-R, and Tf-R. Deletion of TPC2 in mice leads to cholesterol overload and the development of fatty liver hepatitis [1]. We also found that TPCs are essential for the cellular uptake of Ebola viruses (EBOVs) [3]. Disrupting TPC function by gene knockout, small interfering RNAs or small-molecule inhibitors halted virus trafficking and prevented infection with EBOVs. Tetrandrine, the most potent small molecule that was tested, inhibited infection of human macrophages, the primary targets of EBOVs in vivo, and also showed therapeutic efficacy in mice. Taken together, TPCs represent a novel class of ion channels with significant relevance for future drug development. Compounds acting on these channels may be considered in the treatment of diseased states affecting homeostatic control of cellular receptors and cell metabolites and may be effective for antiviral therapy. References: 1. Grimm, C. et al.: Nature Commun. 2014, 4: 4699. 2. Ruas, M. et al.: EMBO J 2015, 34: 1743-1758. 3. Sakurai, Y. et al.: Science 2015, 347: 995-998. 14 • DPhG Annual Meeting 2015 Conference Book PLENARY LECTURES The pre-clinical and clinical development of histamine H4 receptor antagonists P.4 Thurmond, R. L.1 1 Janssen Research & Development, LLC 3210 Merryfield Row, San Diego, CA 92107 The histamine H4 receptor is a high affinity receptor for histamine and has become an attractive target for the development of drugs for the treatment of inflammation, pruritus, allergy and asthma. The H4 receptor mediates chemotaxis and cytokine release of mast cells, eosinophils, monocytes, dendritic cells and T cells. In addition, histamine released from mast cells or from other cell types can influence T cell polarization via activation of the H4 receptor. The receptor also mediates T cell activity in vivo and has a proinflammatory effect not only in models of the innate immune response, but also in models of asthma and contact dermatitis, where it mainly affects T cell responses. Extensive medicinal chemistry and pharmacology efforts have led to the development of modulators with excellent potency and selectivity for the H4 receptor. This has enabled exploration of the role of the receptor in human disease and provided candidates for clinical investigation. Several compounds have reached the clinic including JNJ 39758979 that has progressed into phase II clinical trials. JNJ-39758979 has shown efficacy in preclinical pruritus and atopic dermatitis models [1,2]. Preclinically it reduces histamine-induced scratching in mice and it reduces inflammation in a FITC-induced dermatitis model. These findings have been validated in the clinic with JNJ-39758979 being shown to reduce histamine-induce itch in humans and have effects in patients with atopic dermatitis [3,4]. This talk will highlight recent clinical and preclinical data supporting a role for the H4 receptor in human disease, as well as some of the obstacles encountered when advancing compounds with a novel mechanism into clinical studies. References: 1. Thurmond, R. L. et al.: J. Pharmacol. Exp. Ther. 2014, 349:176-184. 2. Savall, B. M. et al.: J. Med. Chem. 2014, 57:2429–2439. 3. Kollmeier, A. K. et al.: J. Pharmacol. Exp. Ther. 2014, 350:181-187. 4. Murata, Y. et al.: J. Dermatol. 2015, 42:129-139. DPhG Annual Meeting 2015 Conference Book • 15 PLENARY LECTURES Liver research in Düsseldorf: Bridging basic and clinical science P.5 Häussinger, D.1 1 Department Internal Medicine, Gastroenterology, Hepatology and Infectious Diseases, Heinrich Heine University Düsseldorf Clinical and experimental liver research has a long-standing tradition at the Heinrich Heine University Düsseldorf. Over the last two decades, important discoveries were made in the DFG-funded Collaborative Research Centers SFB 575 “Experimental Hepatology” and SFB 974 “Communication and Systems Relevance of Liver Damage and Regeneration” and the Clinical Research Group 217 “Hepatobiliary Transport in Health and Disease” and a few selected achievements of these research networks will be presented in the following. Liver cell hydration was identified as a dynamic parameter which can change within minutes under the influence of cumulative substrate uptake into the hepatocyte, hormones, oxidative stress, nerve impulses, urea and hydrophobic bile acids. Hepatocytes can sense these hydration (cell volume) changes and activate signalling events which link cell hydration to cell function. ß1 integrins act as osmosensors in response to cell swelling, and trigger a proliferative, anabolic and choleretic state. On the other hand, hepatocyte shrinkage is sensed via intracellular Cl- and endosomes and triggers a proapoptotic, catabolic and cholestatic state. Hepatocyte swelling is also an integral part of insulin signalling and mediates the anti-proteolytic and proliferative effects of the hormone. Bile acid secretion is the result of a coordinated action of sinusoidal bile salt uptake (e.g. Ntcp) and canalicular secretion (e.g. via Bsep) and is regulated not only at the level of gene expression, but also at the level of transporter retrieval and insertion, which trigger cholestasis and choleresis, respectively. Bile acid retention in the hepatocyte induces liver damage through activation of CD95-dependent apoptosis. The different mutations of the bile salt export pump Bsep can trigger a broad spectrum of liver pathologies. One of these is the newly discovered autoimmune-Bsep deficiency, which resembles progressive familial intrahepatic cholestasis (PFIC-2), but cannot be cured by liver transplantation. Ursodesoxycholate (UDC), which is rapidly conjugated in vivo with taurine (TUDC) is frequently used in the treatment of cholestatic liver disease due to its choleretic and cytoprotective effects. Recent data showed that TUDC directly interacts with and activates intracellular ß1 integrins, thereby triggering osmosignalling events resulting in choleresis, proliferation and cytoprotection. Bile acids are also be sensed by the membrane-bound and G-protein-coupled receptor TGR5 (Gpbar). The receptor is expressed in many organs, including the brain and in liver it is found in sinusoidal endothelial cells (SEC), Kupffer cells (KC) and cholangiocytes, but not in liver parenchymal cells (hepatocytes). TGR5 acts as a bile acid sensor in cilia of cholangiocytes and triggers bile flow, cholangiocyte protection and proliferation. TGR5 is overexpressed in cholangiocarcinoma and may in view of its proliferative action be a novel target for anti-tumor therapy. Hepatic stellate cells (HSC) were identified as liverresident mesenchymal stem cells and play an important role in liver regeneration as shown in vitro and in in vivo HSC transplantation experiments. Hepatic differentiation of HSC is not only achieved by growth factors (HGF/FGF4), but surprisingly also by TUDC at low concentrations. In addition to the stem cell function of HSC, the space of Dissé was identified as a stem cell niche in the liver. These findings have considerable impact on our understanding of liver regeneration, extramedullary hematopoiesis and liver metastasis. In liver, glutamine synthetase (GS) is restricted to a small perivenous hepatocyte population and acts here as a high affinity ammonia scavenger before the hepatic blood reaches the systemic circulation. Liver-specific GS knockout triggers hyperammonemia and cerebral alterations characteristic for hepatic encephalopathy (HE), a neuropsychiatric syndrome frequently accompanying liver cirrhosis. This complication is the consequence of a low grade cerebral edema and an oxidative/nitrosative stress response, which can be triggered by ammonia, inflammatory cytokines and sedatives and results in cerebral protein tyrosine nitration, RNA oxidation, HE-specific alterations in gene expression and induction of astrocyte senescence. These phenomena were also identified in the brain from patients dying with liver cirrhosis and HE, but not in cirrhotic patients without HE. Interference with these pathophysiological events may provide new approaches for the treatment of HE. 16 • DPhG Annual Meeting 2015 Conference Book PLENARY LECTURES Drug approval and regulatory science – Where to go? P.6 Broich, K.1 1 Federal Institute of Drugs and Medical Devices, 53175 Bonn, Germany Requirements for marketing authorisation and monitoring of safety of medicinal products is highly harmonized in the European Union (EU), however, patients increasingly demand earlier access to new and innovative products. This leads to new concepts and challenges for regulators, e.g. the concept of adaptive pathways is based on a lifespan approach consisting of an early approval of a medicine on a limited database for a restricted patient population in areas of high unmet medical need. Based on this initial approval the product will be further developed with buyin from multiple stakeholders including HTA/reimbursement bodies, which needs early exchange on study designs, study endpoints and active comparators for clinical trials. A new clinical trial directive has been introduced and national implementation is underway. To fulfil our responsibilities and to adapt to the challenges ahead our own regulatory research activities will be strengthened. Core areas for these activities have been defined (personalized medicine, pharmacoepidemiology, safety of medical devices) and together with partners from academia, learned societies, research organisations and patient representatives we are fostering regulatory science at the BfArM. The key features of these challenges and activities will be presented. DPhG Annual Meeting 2015 Conference Book • 17 PLENARY LECTURES From synthetic nucleic acids to artificial genes and genomes: Drugs for the future? P.7 Herdewijn, P.1 1 Medicinal Chemistry, Rega Institute for Medical Research, KU Leuven, Minderbroedersstraat 10, 3000 Leuven, Belgium With the aim to develop synthetic oligonucleotides, genes and genomes for therapeutic purposes, we have started a selection procedure of chemically modified nucleotides and nucleic acids that could fulfil the requirements for in vivo propagation. Several backbone modified nucleic acids (XNA) have been compared for in vivo transliteration into DNA as well as new base-pair systems to develop an orthogonal genetic alphabet. The implementation of XNA implies the development of dedicated polymerases to be able to select synthetic aptamers with and without catalytic activity. Alternatively, implementation of synthetic genes and genomes can be reached by in vivo evolution technologies and the first unlimited self-reproduction of a microorganism with a chemically modified genome has been realized. 18 • DPhG Annual Meeting 2015 Conference Book 2 SCIENTIFIC LECTURES DPhG Annual Meeting 2015 Conference Book • 19 SCIENTIFIC LECTURES 2.1 Antiviral Drugs Chair: H. Rübsamen-Schaeff, H. Zimmermann SL.01 Directly acting antivirals for hepatitis C virus – close to “perfectovir” Pietschmann, T.1 Division Experimental Virology, TWINCORE - Centre for Experimental and Clinical Infection Research, Feodor-Lynen-Str. 7-9, 30625 Hannover, Germany 1 Chronic infection with hepatitis C virus is associated with severe liver disease including hepatitis, cirrhosis and hepatocellular carcinoma. End stage liver disease due to HCV infection is one of the major indications for liver transplantation. With an estimated number of 160 million infected individuals world-wide, HCV infection is a global challenge for public health. Recent advances in HCV drug development have led to the licensing of three different drug classes including protease and polymerase inhibitors as well as drugs targeting the viral NS5A protein. Combination therapies including these so called directly acting antivirals (DAAs) cure more than 90% of infected patients – at least in clinical study populations. However, these regimens are very expensive limiting access to therapy particularly in countries with highest HCV prevalence. Thus, in the absence of an HCV vaccine, it is unlikely that these efficacious yet expensive novel drugs will rapidly suppress the HCV disease burden globally. Therefore, to identify cost effective therapeutics for treatment of HCV infection we screened a compound library including licensed drugs for treatment of neuronal or heart diseases. Using this approach, we identify a number of structurally related clinically licensed phenothiazines and diphenylmethylpiperazines that inhibit HCV infection in a dose dependent fashion. Among our primary hits, we focused our attention on flunarizine, a diphenylmethylpiperazine and T-type calcium channel inhibitor used to treat migraine. This drug specifically inhibited HCV cell entry (IC50 of 388.2 nM) in Huh-7 cells and in primary human hepatocytes in a genotype-dependent fashion. It also reduced HCV infection in a genetically humanized mouse model for HCV cell entry. Using a comprehensive set of experiments including single particle tracking imaging analyses, time of addition studies, viral resistance selection, and numerous virus chimeras, we provide evidence that this drug specifically inhibits HCV membrane fusion by targeting the functions of E1 and E2. While flunarizine exclusively targets HCV genotype 2 viruses we show that closely related licenced drugs like pimozide have an expanded cross-genotype coverage targeting GTs 3, 5 and 7 viruses with an IC50 in the low micromolar range. Although resistance to these drugs can be selected in cell culture, we show that these mutations involve highly conserved residues and that they enhance viral susceptibility to neutralizing antibodies. Of note, resistance mutations to these drugs map to a subdomain in the viral E1 protein which has previously been implicated as viral fusion peptide. These observations reveal novel details about HCV membrane fusion. Moreover, flunarizine and related compounds represent first-in-class HCV fusion inhibitors that merit consideration for repurposing as cost-effective component of HCV combination therapies. 20 • DPhG Annual Meeting 2015 Conference Book ANTIVIRAL DRUGS SL.02 How to deal with HBV? - State of the art and new concepts in HBV therapy Paulsen, D.1 1 AiCuris GmbH & Co.KG, 42117 Wuppertal, Germany Chronic hepatitis B virus (HBV) infection is a major cause of severe liver disease and premature liver-related mortality, worldwide. Antiviral therapy has made continuous progress over the past 20 years resulting in two strategies to treat chronic hepatitis B: Interferon therapy or nucelos(t)idic inhibitors of the HBV polymerase. These antiviral drugs are effectively suppressing HBV replication, but only a minority of patients is able to mount the necessary immune control to keep HBV without treatment at bay. Therefore, the current state of the art in HBV treatment is long-term and perhaps lifelong therapy. New concepts for HBV therapy are underway – either targeting the virus directly or tackling reconstitution of the immune response via immunomodulatory approaches. Virion assembly and release inhibitors, entry inhibitors, cccDNA inhibitors, TLR agonists, and therapeutic vaccination approaches will be discussed in more detail. DPhG Annual Meeting 2015 Conference Book • 21 SCIENTIFIC LECTURES SL.03 Advances and challenges in diagnostics & therapy for HIV/AIDS Stephan, C.1 1 Zentrum der Inneren Medizin / Schwerpunkt Infektiologie Universitätsklinikum Frankfurt, Goethe-Universität, 60590 Frankfurt HIV-infected patients have reached the daily work of many physicians. Treatment and care of affected individuals remains to be a challenge, despite the co-morbidity and co-infections’ therapy achievements, but as well because of an increasingly aging patient group. However, the guidance and strategic decisions and planning of antiretroviral therapy always belongs to the specialized HIV-treating physician, in order to control combination therapy and review co-therapies. Nowadays establishment of HIV diagnosis is still taking place often in advanced stage of HIV-disease and this requires highly sophisticated treatment standards. Late diagnosis means disadvantages for the individual patient. The missed diagnosis in prior medical history in a late presenting individual is resulting in avoidable morbidity and subsequent HIV transmissions. Current strategies for cure from HIV are fading the efforts to develop a therapeutic vaccination, which had suffered many setbacks in the past. Actual cure strategies include individual induction of broadly neutralizing antibodies on the one hand, and gene therapeutic approaches to manipulate lymphocyte function, i.e. regulating the infectiousness of human cells. 22 • DPhG Annual Meeting 2015 Conference Book ANTIVIRAL DRUGS SL.04 The era after nucleosides: Novel therapeutic approaches against herpes viruses Rübsamen-Schaeff, H.1 1 AiCuris GmbH &Co KG, Friedrich-Ebert-Strasse 475, 42117 Wuppertal , Germany Herpesviruses are a large family of viruses and cause chronic infections in humans and animals. While an immunocompetent individual generally will control these viruses, they can cause severe infections or even be lifethreatening in all conditions of immune-incompetence, e.g. for neonates, transplant recipients or AIDS patients. The treatment of herpesviruses has so far been confined to inhibitors of the viral polymerases using mainly nucleoside analogues. However, this approach has two shortcomings: 1) Polymerase inhibitors may cross-react with human polymerases leading to a number of toxic effects. 2) Polymerase inhibitors are prodrugs and require for their activation a viral enzyme to conduct the first phosphorylation step, so that they are not active in uninfected cells and do not protect these from infection. We, therefore, have attempted to address other targets of herpes viruses and generated a terminase inhibitor, Letermovir, against the Human Cytomegalovirus (HCMV) and a helicase-primase inhibitor, Pritelivir, against Herpes Simplex Viruses (HSV 1 and 2). Both drugs stem from non-nucleosidic classes: Letermovir is a 3,4-dihydroquinazoline-4-yl-acetic acid derivative and Pritelivir is a 2-phenyl-N-thiazole-2-yl-acetamide derivative. Both drugs are active in and protect uninfected cells against infection. Notably, both drugs also have a very steep doseresponse curve for antiviral efficacy. In contrast to polymerase inhibitors they show only small shifts in IC50, when the multiplicity of the viral infection per cell is increased, meaning that they can very well deal with high viral loads. These nonclinical observations have also translated into the clinics: In phase II clinical trials, a high efficacy and good safety profile could be demonstrated for Letermovir (1) as well as for Pritelivir (2). In addition, Pritelivir demonstrated superiority over the nucleoside analogue valacyclovir in a head to head clinical phase II study in patients with genital herpes, again with a good tolerability similar to valacyclovir. Letermovir is presently in phase III clinical testing in patients, who received a stem cell transplantation. Pritelivir is presently on clinical hold due to so far unexplained observations in a 39 week animal study. (http://clinicaltrials.gov/ct2/show/NCT02137772?term=letermovir&rank=1). References: 1. Chemaly, R. F. et al: N Engl. J Med 2014, 370: 1781-1789. 2. Wald, A. L. et al: N Engl. J Med 2014, 370: 201-210. DPhG Annual Meeting 2015 Conference Book • 23 SCIENTIFIC LECTURES SL.05 New avenues in anti-influenza therapy Ludwig, S.1 Institute of Molecular Virology, Westfälische-Wilhelms-University Muenster, Von Esmarch Str. 56, 48149 Muenster, Germany 1 Influenza virus infection results in the activation of a variety of intracellular signaling responses. Influenza viruses have acquired the capability to exploit some of these activities to support efficient replication. This dependence of influenza virus propagation on cellular signaling factors provides opportunities for a novel approach of antiviral interventions that targets essential host factors instead of viral components. We have identified several cell signaling targets that are suitable for antiviral intervention, including the classical mitogenic MAP kinase cascade, that regulates active viral RNP export, or the NF-kappaB pathway, that interferes with the apoptotic response. In previous work we could demonstrate that inhibition of these pathways efficiently blocked virus replication in cells and animals. Several inhibitors of the NF-kappaB pathway or the MAP kinase cascade are now under advanced clinical evaluation or even licensed for clinical use for other diseases. We show that these novel signaling blockers (a) efficiently inhibit influenza virus replication in cell culture and mouse models, (b) are broadly active against all influenza A and B viruses analysed so far, (c) showed no tendency to induce resistant virus variants, (d) are not toxic for cells or animals in the concentration and time line used, and (e) confirm the postulated and fully unravelled mode of action: blocking of the transport of viral genomes from the nucleus. Inhibitors of the classical MAP kinase cascade and blockers of the NF-kappaB pathway are promising compounds against influenza with a broad activity and no tendency to induce resistance. Since there are existing drugs under clinical evaluation or even licensed compounds available, repurposing of those drugs may be a fast and efficient way for the development of a completely novel anti-influenza approach. Accordingly, a phase II clinical trial with a drug that blocks the NF-kappaB pathway is currently ongoing [www.clinicaltrialsregister.eu/ctr-search/trial/2012-004072-19/DE]. This is the first trial with an anti-influenza drug that targets a cellular signalling factor and thus represents a paradigm change in anti influenza therapy. 24 • DPhG Annual Meeting 2015 Conference Book ANTIVIRAL DRUGS SL.06 Antivirals, a lot has been achieved, yet a long way to go Neyts, J.1 1 Laboratory of Virology, Rega Institute for Medical Research, University of Leuven, Belgium Today, small molecule antiviral drugs are available for the treatment of infections with herpesviruses, HIV, HBV and HCV as well as with influenza viruses. Ribavirin, a broad-spectrum (but aspecific) antiviral is being used with very limited success for the treatment of RSV infections. Yet, for many other viruses that cause life-threatening infections [several of which are considered emerging and/or neglected] there are no drugs available. Ideally, potent and broad-spectrum (i.e., pan-genus or pan-family virus activity) antiviral drugs should be developed whereby one drug could be used for the treatment of a number of such viral infections. I will discuss those viral infections with the greatest need for an antiviral as well as the latest developments in the field. DPhG Annual Meeting 2015 Conference Book • 25 SCIENTIFIC LECTURES 2.2 Blood/Brain Barrier Chairs: G. Fricker, F. Helm SL.07 Liposomal delivery of Trientine for treatment of Wilson’s Disease in the brain Tremmel, R. 1; Fricker, G.1; Helm, F.1 1 Institut für Pharmazie und Molekulare Biotechnologie , Ruprecht-Karls Universität Heidelberg, Im Neuenheimer Feld 329, 69210 Heidelberg Wilson disease, a rare, inherited, autosomal recessive disorder, which leads to copper accumulation in several organs including the brain. Beside other symptoms it causes neurological and psychiatric disorders, due to the lack of therapeutics which are able to pass the blood brain barrier (BBB). Therapeutics for a front-line treatment of Wilson disease are D-Penicillamin and Trientine. Both substances are chelators and forming complexes with copper to be excreted in the urine. However, both of them show a poor CNS penetration. One option to overcome the BBB is the encapsulation of API’s in nanosized carriers like nanoparticles or liposomes, which can be surface modified with vectors targeting the BBB. One possible vector is apolipoprotein-E4 (ApoE), which is transported across the BBB via transcytosis starting with binding to the low-density lipoprotein receptor. Other options are the coupling of cationized bovine serum albumin (cBSA) to the liposomal surface in order to utilize adsorptive endocytosis or the coupling of positively charged cell penetrating peptides. Here we describe the manufacture of trientine loaded liposomes, their coupling to the above mentioned vectors and the evaluation of the BBB penetrating properties in vitro in isolated brain capillary endothelial cells (PBCEC) and in vivo in rats after i.v.-administration. We reached encapsulation efficiencies of Trientine in liposomes up to 30%. After incubating PBCEC’s with liposomal formulations, 10-15% of the administered concentration could accumulate inside the cells. In vivo a 12fold accumulation of liposomal Trientin in brain parenchyma could be observed, compared to Trientine in solution. 26 • DPhG Annual Meeting 2015 Conference Book BLOOD/BRAIN BARRIER SL.08 Molecular regulation of endothelial barrier properties in the central nervous system Guerit, S. 1; Czupalla, C. J.1*; Devraj, K.1; Ziegler, N.1; Gerhardt, H. 2; Plate, K. H.1; Liebner, S.1 1 Institute of Neurology (Edinger-Institute), Goethe University, Heinrich-Hoffmann-Str. 7, 60528 Frankfurt, Germany Integrative Vaskular Biology, MDC Berlin, Robert-Rössle-Straße 10, 13125 Berlin-Buch * current address: Stanford University School of Medicine, Department of Pathology, Lane Building, Mailcode 5324, Stanford, CA. 943055324 2 Endothelial Wnt/β-catenin signaling is necessary for developmental angiogenesis of the central nervous system (CNS) and differentiation, maturation and maintenance of the blood-brain barrier (BBB) [1]. In the adult brain, pericytes and astrocytes are the closest cellular neighbors of the barrier endothelium in the neuro-vascular unit (NVU). Although both cell types doubtlessly participate in BBB maintenance and integrity, the contribution of Wnt/βcatenin signaling herein remains obscure. In order to characterize AC-derived Wnts as BBB maintaining factors, we made use of in vitro (Evilox/lox TAT-Cre treated ACs) and in vivo (GFAP-Cre:Evilox/lox mice, AC∆Evi) model systems in which ACs do not express the Evi protein, which is essential for the release of Wnts. Trans-endothelial electrical resistance (TEER) was significantly decreased when murine brain endothelioma cells (MBE) were co-cultivated with AC∆Evi in comparison to ACwtEvi controls. In vivo analysis of the AC∆Evi mice revealed that the AC-specific Evi deletion led to brain edema, indicating a partial breakdown of BBB structures that however, did not cause lethality of the mice. Additionally, AC∆Evi mice displayed alterations in vessel remodeling. Together these findings suggest that Wnt growth factors released by ACs play a role in brain vessel structure and regulation of the BBB phenotype. Moreover, we currently do not understand how vascular heterogeneity in the CNS is accomplished during development and how it is maintained in the adult. Besides the contribution of Wnt factors in BBB maintenance, their function in the differentiation of the leaky vascular phenotype in the circumventricular organs (CVOs), conferring neurosecretory and -sensory function, is not understood in detail. Neither during embryonic development, nor at early postnatal stages we detected activation of β-catenin signaling in CVO vessels of BAT-gal reporter mice. Dominant activation of the β-catenin pathway (gain-of-function, GOF) in endothelial cells (βCatGOF_EC) during early postnatal development led to expression of claudin-5 in vessels of the sub-fornical organ (SFO), whereas Meca-32 immunoreactivity was reduced. Moreover, βCatGOF_EC generated a thinner vascular phenotype within the SFO. Currently, we investigate in detail the circuitry of the Wnt pathway in the CVOs and its specific role in CVO differentiation. In summary, we can show that activation of the Wnt pathway in ECs is not only important during early brain vascularization, but also during later stages of life and particularly in the aged CNS. Herein AC-derived Wnts may contribute to brain EC differentiation and vascular remodeling. At the same time the lack of Wnt activation from certain vessels such as those of the CVO is crucial for the formation of their specific phenotype and function. Further investigations are required to better understand vascular heterogeneity in the brain in general and during aging in particular. Acknowledgments: Theodor Kocher Institute Bern, Switzerland, Engelhardt B. References: 1. Engelhardt, B.; Liebner, S.: Cell Tissue Res. 2014, 355: 687–699. DPhG Annual Meeting 2015 Conference Book • 27 SCIENTIFIC LECTURES SL.09 Toxicity of poly(n-butylcyano-acrylate) (PBCA)-nanoparticles designed to overcome the bloodbrain barrier Kolter, M.1; Fricker, G.1 1 Department of Pharmaceutical Technology and Biopharmacy, Institute of Pharmacy and Molecular Biotechnology, University of Heidelberg, Im Neuenheimer Feld 329, 69120 Heidelberg, Germany The therapy of diseases of the central nervous system is a major challenge, since drugs have to overcome the blood–brain barrier (BBB), which separates the brain from the peripheral blood stream. A powerful strategy to circumvent the BBB is the intravenous administration of drug-loaded poly(n-butylcyano-acrylate) (PBCA)nanoparticles coated with polysorbate 80 (PS80), which are able to cross the BBB via receptor-mediated transcytosis. The aim of this project was the preparation, physicochemical characterization and in particular the toxicological evaluation of PBCA-nanoparticles at the BBB (in vitro), representing the target organ, in human whole blood (ex vivo), as the site of administration and in a rat model (in vivo). A reproducible preparation of endotoxin-free PBCA-nanoparticles, composed of n-butylcyano-acrylate, poloxamer 188, sodium dodecyl sulfate (SDS) and soybean oil, was developed using a miniemulsion technique [1]. The nanoparticles were approximately 140 nm in size and exhibited a narrow particle size distribution, a negative surface charge, a spherical shape and a low agglomeration potential. The toxicological evaluation of PBCA-nanoparticles in an in vitro BBB model, which was composed of porcine brain capillary endothelial cells, included the effects on metabolic activity, cell viability, cellular oxidative stress and BBB integrity. Cells were affected in a strongly concentration-, time- and surface-dependent manner, but no cell death or loss of metabolic activity and only a moderate generation of oxidative stress was observed for nanoparticle concentrations ≤ 500 μg/ml up to 3 h of treatment. However, the BBB integrity was strongly affected by PBCAnanoparticles even at very low concentrations (≥ 5 µg/ml) resulting in a concentration-dependent, reversible and repeatable BBB opening. Further experiments indicated that this effect allows a passive and size-dependent paracellular transport of high molecular weight drugs into the brain. In human whole blood, PBCA-nanoparticles caused no inflammatory burst. Only IL-8 displayed a significant release after nanoparticle exposure, whereas other proinflammatory cytokines, which are typically released by nanoparticles (e.g. IL-1β, IL-6, TNF-α), remained unaffected. In addition, human leukocytes did not suffer major cell death after incubation with nanoparticles in concentrations up to 500 µg/ml over 3 h. The good blood compatibility was endorsed by the absence of any hemolytic effect at concentrations of ≤ 500 µg/ml over 24 h. Finally, the toxicological examination in rats (in vivo) identified neither inflammatory processes nor severe organ damages indicating an even lower in vivo than in vitro or ex vivo toxicity. In conclusion, PBCA-nanoparticles are an effective and promising drug delivery system to overcome the BBB with a low toxic potential demonstrated by hardly any or only moderate cytotoxic and inflammatory effects at therapeutic concentrations (up to 500 µg/ml) and incubation times (up to 3 h) at the BBB, in human whole blood and in vivo. Acknowledgments: Deutsche Forschungsgemeinschaft (DFG) (DFG FR1211-17/1) References: 1. Reimold, I. et al.: Eur. J. Pharm. Biopharm. 2008, 70(2): 627-32. 28 • DPhG Annual Meeting 2015 Conference Book REGULATION OF BETA-CELL FUNCTION – IMPLICATIONS FOR DIABETES 2.3 Regulation of Beta-Cell Function – Implications for Diabetes Chairs: G. Drews, E. Oetjen SL.10 Role of bile acids in beta-cell functions Drews, G.1 1 Institute of Pharmacy, Dept. Pharmacology, Toxicology, and Clinical Pharmacy, University of Tübingen Background: Meanwhile it is well established that bile acids (BAs) play an important role as signaling molecules besides their role in fat digestion. The most important BA receptors are the G-protein-coupled TGR5 and the nuclear farnesoid X receptor (FXR). They are involved in the maintenance of glucose homeostasis but also affect lipid metabolism and energy expenditure by acting on liver, skeletal muscle, and adipose tissue. It has been shown that the composition of the bile acid pool and the pool size are altered in type-2 diabetes mellitus (T2DM). We have studied the effects of BAs on pancreatic beta-cells and focused our research on the role of FXR in stimulus secretion coupling. Methods: Islet of Langerhans and dispersed beta-cells from wildtype (WT) mice and mice with genetic deletion of the FXR (FXR-KO) and of KATP channels (SUR1-KO) were used for in vitro experiments to study insulin secretion (radioimmunoassay), the cytosolic Ca2+ concentration (fluorescence technique), and KATP currents (patch-clamp technique). In vivo experiments were performed with mice fed a control or high fed diet (HFD) for 12 weeks. Glucose tolerance and insulin sensitivity were measured after i.p. application of glucose and insulin, respectively. Results: Chenodeoxycholic acid (CDC) and its conjugates enhance glucose-induced insulin secretion in islets of WT mice and increase the cytosolic Ca2+ concentration. These effects are lacking in beta-cells of FXR-KO mice, demonstrating that the effects of these BAs are mediated by FXR activation. Interestingly, activation of glucosedependent insulin secretion by BAs is also absent in islets from SUR1-KO mice, indicating that KATP channels are involved in the BA effect. Accordingly, the increase in the cytosolic Ca2+ concentration is a result of an indirect effect of BAs on KATP channels, i.e. closure of KATP channels by BAs only occurs in metabolically intact beta-cells but not in excised membrane patches. Changes in the cytosolic Ca2+ concentration and KATP channel activity occur within a few seconds or minutes after drug application, pointing to a non-genomic mode of action of BAs. Consistently with these in vitro experiments, glucose tolerance is impaired in lean FXR-KO mice whereas insulin sensitivity remains unchanged, demonstrating that the effect is due to altered beta-cell function. As expected, a HFD diet induces an impairment of glucose tolerance in WT mice. Remarkably, after a HFD the glucose tolerance of FXRKO mice does not impair but is in the same range as in WT mice under control diet. This clearly shows that the FXR has different functions in lean and obese mice. It is hypothesized that this shift in function is linked to a translocation of the FXR from the cytosol to the nucleus. Conclusions: FXR agonists exert rapid, non-genomic effects in beta-cells. FXR activation in beta-cell clearly affects glucose homeostasis. FXR stimulation has different effects in lean and obese mice. DPhG Annual Meeting 2015 Conference Book • 29 SCIENTIFIC LECTURES SL.11 Mobility of submembrane insulin granules Rustenbeck, I.1; Brüning, D.1; Schumacher, K.1; Matz, M.2; Baumann, K.2 1 Institute 2 Institute of Pharmacology and Toxicology, University of Braunschweig, D-38106 Braunschweig, Germany of Medicinal and Pharmaceutical Chemistry, University of Braunschweig, D-38106 Braunschweig, Germany Background and aims: The biphasic pattern of insulin secretion is essential for the maintenance of glucose homeostasis. The currently predominant hypothesis explains this pattern as the consequence of two different pools of insulin granules. One pool consists of secretion-ready granules attached to the plasma membrane and the other of granules located further inside the cell. A number of observations have put this hypothesis into question. To characterize glucose-induced and depolarization-induced insulin secretion, we compared insulin secretion and parameters of insulin granule mobility in insulin-secreting MIN6 cells and in normal mouse beta-cells. Methods: The number, mobility and exocytosis of submembrane insulin granules can be visualized by TIRF (total internal reflection fluorescence) microscopy. For a quantitative evaluation a computerized, observer-independent evaluation of the video files is required. Insulin granules were fluorescently labeled by transient transfection with insulin-EGFP in MIN6 cells and by adenoviral transduction in primary mouse beta-cells. The cells were continuously perifused with oxygenated medium containing the respective stimuli. The TIRF microscopy image files (1 sequence = 200 images = 25 seconds) were evaluated by an in-house written program. The insulin secretion of MIN6 pseudoislets and mouse islets was measured by perifusion with ELISA of the fractionated efflux. Results: The perifused islets and pseudo-islets were stimulated by 40 mM K+ and, after a wash-out period, by 30 mM glucose. In MIN6 pseudo-islets 30 mM glucose was much less effective than 40 mM K+. When the K+ depolarization preceded the glucose stimulus, glucose was entirely ineffective. In mouse islets glucose was about equi-effective with K+ depolarization. Under control condition, the number of granules in the first image of the sequences was 337 ± 34 per MIN6 cell but only 144 ± 14 per beta cell. In both cell types this parameter remained essentially stable during imaging. During basal and during stimulated activity of the cells, there was a continuous arrival of granules at the submembrane space which was closely mirrored by the departure of granules back into the cell interior. Under basal condition, the total number of granules identified per sequence was 6972 ± 873 in MIN6 cells and 1966 ± 172 in beta-cells. In MIN6 cells 82% of these granules remained at the plasma membrane for less than 1 s (short-term residents), and only 1.7% were visible for the entire sequence = 25 s (long-term residents). The corresponding values for beta-cells were similar, 80% and 3.0%, respectively. Most of the granules that fused with the plasma membrane were visible from the beginning of the sequence (potential long-term residents) and had a low lateral mobility, but there were also fusion events a few seconds after arrival at the plasma membrane.Unexpectedly, the rate of granule fusion was not increased during phases of stimulation, whereas the vertical mobility (rate of arrival and departures) was significantly increased. Conclusion: In insulin-secreting cells there is a continuous turnover of granules in the submembrane space. A small proportion of the granules becomes more firmly attached to the membrane. These are the granules that preferentially fuse with the plasma membrane. The link between secretion and fusion may be less direct than currently assumed. Even though a general similarity exists between the situation in MIN6-cells and primary beta cells, only beta cells are adequate to characterize the response to glucose. Acknowledgments: Supported by DFG Ru 368/5-2 30 • DPhG Annual Meeting 2015 Conference Book REGULATION OF BETA-CELL FUNCTION – IMPLICATIONS FOR DIABETES SL.12 Reactive oxygen species: Pharmacological target or physical prerequisite for beta-cell function? Düfer, M.1 1 Institute of Pharmaceutical and Medicinal Chemistry, Dept. of Pharmacology, Münster University, Germany Elevated generation of reactive oxygen species (ROS), e.g. in response to overnutrition, is known to play a pivotal role for manifestation and progression of type 2 diabetes mellitus. It is well established that key steps for regulation of insulin release are severely impaired by oxidative stress. As a consequence, therapeutic strategies to reduce ROS production or accumulation in pancreatic beta-cells are investigated to protect against the deterioration of beta-cell function in patients suffering from impaired glycemic control. However, there is increasing evidence that apart from their involvement in beta-cell failure ROS are important signalling molecules necessary for physiological regulation of insulin release. Acute stimulation of insulin secretion by glucose coincides with an elevation of hydrogen peroxide and with a decrease in superoxide radical anions. Challenging beta-cell metabolism by high glucose and lipid concentrations does not cause permanent oxidative stress but drastically reduces the ability of nutrients to alter intracellular ROS concentrations. This effect is presumably mediated by malfunction of antioxidant enzymes as activation of the transcription factor Nrf-2, which increases gene transcription of antioxidant pathways, restores the physiological ROS profile. Based on these observations, the objective of strategies targeting antioxidant capacity of beta-cells should not be complete prevention of ROS formation but preservation of the physiological balance between different species of ROS. DPhG Annual Meeting 2015 Conference Book • 31 SCIENTIFIC LECTURES SL.13 Regulation of beta-cell function and mass by the dual leucine zipper kinase Oetjen, E.1 1 Department of Clinical Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Martinistr. 52, 20246 Hamburg; Institute of Pharmacy, University of Hamburg, Bundesstr. 45, 20146 Hamburg, Germany The dual leucine zipper kinase DLK induces β-cell apoptosis by inhibiting the transcriptional activity conferred by the β-cell protective transcription factor cAMP response element binding protein CREB. This action might contribute to β-cell loss and ultimately diabetes. Within its kinase domain DLK shares high homology with the mixed lineage kinase (MLK) 3, which is activated by tumor necrosis factor (TNF) α and interleukin (IL)-1β, known prediabetic signals. In the present study, the regulation of DLK in β-cells by these cytokines was investigated. Both, TNFα and IL-1β induced the nuclear translocation of DLK. Mutations within a putative nuclear localization signal (NLS) prevented basal and cytokine-induced nuclear localization of DLK and binding to the importin receptor importin α, thereby demonstrating a functional NLS within DLK. DLK NLS mutants were catalytically active as they phosphorylated their down-stream kinase c-Jun N-terminal kinase to the same extent as DLK wild-type but did neither inhibit CREB-dependent gene transcription nor transcription conferred by the promoter of the anti-apoptotic protein BCL-xL. In addition, the β-cell apoptosis-inducing effect of DLK was severely diminished by mutation of its NLS. In two distinct murine models of prediabetes, enhanced nuclear DLK was found. These data demonstrate that DLK exerts distinct functions, depending on its subcellular localization and thus provide a novel level of regulating DLK action. Furthermore, the prevention of the nuclear localization of DLK as induced by prediabetic signals with consecutive suppression of β-cell apoptosis might constitute a novel target in the therapy of diabetes mellitus. 32 • DPhG Annual Meeting 2015 Conference Book GLISTEN – GPCR MEDICINAL CHEMISTRY 2.4 GLISTEN – GPCR Medicinal Chemistry Chairs: N. Tschammer, P. Kolb SL.14 Selective orthosteric agonists in class C GPCRs Acher F. C.1; Brabet, I.2; Rigault, D.1; Busserolles, J.3; Bertrand, H.-O.4; Eschalier, A.3; Goudet, C.2; Pin, J.-P.2 Université Paris Descartes, UMR8601-CNRS, 45 rue des Saints-Pères, Paris 06, France Institut de Génomique Fonctionnelle, CNRS UMR5203, INSERM U661, Université Montpellier, 141 rue de la Cardonille, 34094 Montpellier, France 3 Université d’Auvergne INSERM U 1107, Neuro-Dol, 63001 Clermont-Ferrand, France 4 Biovia Dassault Systems, Parc Club Orsay Université, 20 rue Jean Rostand, 91893 Orsay, France 1 2 G Protein Coupled Receptors (GPCRs) are characterized by a seven helix transmembrane domain where the binding site of agonists is found. However in class C GPCRs (e.g. metabotropic glutamate receptors, mGluRs) the orthosteric binding site is located in the large amino terminal domain that folds in two lobes connected by a flexible hinge building the Venus FlyTrap (VFT) domain. The closed conformation of this domain triggers activation of the receptors. It has been difficult to discover subtype selective agonists because of the high conservation of the orthosteric binding site. However by means of a virtual high throughput screening of mGluR subtype 4 VFT homology model, we have discovered a selective pocket adjacent to the glutamate binding site which is in fact a critical chloride binding site [1,2]. Binding to this site confers selectivity and higher potency to ligands. Indeed optimization of the hit, led to the discovery of LSP4-2022 that is mGlu4 receptor selective [3]. Further chemical modulation provided new agonists with higher potency. These agonists were successfully evaluated for their antihyperalgesic properties in inflammatory and neuropathic pain models [4]. References: 1. Selvam, C. et al.: J. Med. Chem. 2010, 53, 2797-2813. 2. Tora, A. S. et al.: FASEB J. 2015 [Epub ahead of print] 3. Goudet, C. et al.: FASEB J. 2012, 26, 1682-1693. 4. Vilar, B. et al.: J. Neurosci. 2013, 33, 18951-18965 DPhG Annual Meeting 2015 Conference Book • 33 SCIENTIFIC LECTURES SL.15 Identification and pharmacological characterization of endogenous and surrogate ligands for orphan G protein-coupled receptors Bräuner-Osborne, H.1; Gloriam, D. E.1 Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Fruebjergvej 3, 2100 Copenhagen, Denmark, 1 G protein-coupled receptors (GPCRs) are a large family of cell membrane embedded signal receivers that are involved in many biological processes and is one of the main target families for approved drugs. GPCRs are activated by a wide variety of ligands, including neurotransmitters, hormones and food constituents such as amino acids, sugars and fatty acids. GPCRs activate intracellular G proteins of which four classes exist (Gαq, Gαi, Gαs and Gα12/13) as well as other intracellular proteins such as ß-arrestins. 358 non-olfactory GPCRs have been identified in the human genome of which ~1/3 remain orphan receptors without confirmed endogenous agonists. Given the involvement of GPRCs in many important biological processes and their success as drug targets, there has been many efforts in academia and industry aiming at identifying the endogenous ligands or developing surrogate ligands for orphan receptors. Many different strategies have been adopted mainly focusing on employment of 'generic' pharmacological assays and screening of tissue extracts or libraries of potential endogenous ligands. This has led to important ligand-receptor matches, but the deorphanization rate has been slow. In contrast we have chosen to use a range of computational methods to select ligands for pharmacological testing to increase the likelihood of hit identification. This has so far led to the following ligand-receptor matches: 1. Identification of basic L-amino acids as potential endogenous agonists for the GPRC6A receptor using receptor modelling and ligand docking [1]. 2. Identification of the first selective GPRC6A antagonists using chemogenomics and 'privileged structures' - multitarget chemical scaffolds [2]. 3. Identification of aromatic L-amino acids and dipeptides as potential endogenous agonists for the GPR139 receptor using a pharmacophore based on previously identified surrogate agonists [3]. 4. Identification of the first surrogate agonist for the GPR132 receptor using GPCR-directed compound libraries [4]. The present lecture will present these case stories and outline our current strategy for future deorphanizations. Acknowledgments: Major financial support for this work has obtained from the Danish Council for Independent Research, the Lundbeck Foundation, the European Research Council and the Hørslev Foundation. References: 1. Wellendorph, P. et al.: Mol. Pharmacol. 2005, 67: 589-597. 2. Gloriam, D. E. et al.: Chem. Biol. 2011, 18: 1489-1498. 3. Isberg, V. et al.: J. Chem. Inf. Model. 2014, 54: 1553-1557. 4. Shehata, M. A. et al.: RSC Advances. 2015, 5: 48551-48557. 34 • DPhG Annual Meeting 2015 Conference Book GLISTEN – GPCR MEDICINAL CHEMISTRY SL.16 Structure free optimisation of fragments for the β2-adrenoreceptor Gawron, S.1; Aristotelous, T.1; Chevillard, F.2; Hopkins, A. L.1; Kolb, P.2; Hopkins-Navratilova, I.1; Gilbert, I. H.1 1 Division of Biological Chemistry and Drug Discovery, College of Life Sciences, Sir James Black Centre, University of Dundee, Dundee, DD1 5EH, UK 2 Philipps-Universität Marburg, Department of Pharmaceutical Chemistry, Marbacher Weg 6, 35032, Marburg, Germany G-protein coupled receptors (GPCRs) are the primary target class of currently marketed drugs, accounting for about a quarter of all drug targets of approved medicines. Here we describe a process of structure-free optimisation of fragments binding to β2-adrenoreceptor in the absence of structure. A decision tree is introduced for optimisation of both the core scaffold and the different vectors. This approach allowed a generation of ‘virtual’ model of the binding site. Subsequently it was possible to rationalise the data by docking the fragments into a crystal structure of the β2-adrenoreceptor. Acknowledgments: University of Dundee, GLISTEN, Marburg University References: 1. Aristotelous, T. et al.: ACS Med. Chem. Lett. 2013, 4: 1005-1010. DPhG Annual Meeting 2015 Conference Book • 35 SCIENTIFIC LECTURES SL.17 Comprehensive analysis of heterotrimeric G-protein complex diversity and their interactions with GPCRs in solution Hillenbrand, M. 1; Schori, C.1; Schöppe, J.1; Plückthun, A.1 1 Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland Agonist binding to G–protein–coupled receptors (GPCRs) triggers signal transduction cascades involving heterotrimeric G proteins as key players. A major obstacle for drug design is the limited knowledge of conformational changes upon agonist binding, the details of interaction with the different G proteins, and the transmission to movements within the G protein. Although a variety of different GPCR/G protein complex structures would be needed, the transient nature of this complex and the intrinsic instability against dissociation make this endeavor very challenging. We have previously evolved GPCR mutants that display higher stability and retain their interaction with G proteins. We aimed at finding all G-protein combinations that preferentially interact with neurotensin receptor 1 (NTR1) and our stabilized mutants. We first systematically analyzed by coimmunoprecipitation the capability of 120 different G-protein combinations consisting of αi1 or αsL and all possible βγ-dimers to form a heterotrimeric complex. This analysis revealed a surprisingly unrestricted ability of the G-protein subunits to form heterotrimeric complexes, including βγ-dimers previously thought to be nonexistent, except for combinations containing β5. A second screen on coupling preference of all G-protein heterotrimers to NTR1 wild type and a stabilized mutant indicated a preference for those Gαi1βγ combinations containing γ1 and γ11. Heterotrimeric G proteins, including combinations believed to be nonexistent, were purified, and complexes with the GPCR were prepared. Our results shed new light on the combinatorial diversity of G proteins and their coupling to GPCRs and open new approaches to improve the stability of GPCR/G-protein complexes. References: Hillenbrand, M. et al.: Proc. Natl. Acad. Sci. USA 2015, 112(11): E1181-1190. 36 • DPhG Annual Meeting 2015 Conference Book EVIDENCE BASED MEDICATION MANAGEMENT 2.5 Evidence based Medication Management Chairs: G. Hempel, S. Läer SL.18 Medication management in clinical care Bertsche, T.1,2 1 Institute 2 Drug of Pharmacy, Clinical Pharmacy, Leipzig University Safety Center, Leipzig University and University Hospital of Leipzig DRP (Drug-Related Problems) in hospitalized patients have a high risk to cause acute harm and even death to the patient. Additionally, after discharge from hospital DRP can continuously occur and cause problems in ambulatory care. Vulnerable patient groups of special interest for strategies to improve patient safety are children and adolescents as well as geriatric patients (including patients requiring intensive/intermediate or palliative care). Clinical pharmacists join the interdisciplinary health care team to perform a structured medication management in clinical care in order to prevent DRP before they occur. Within the hospital setting apart from medication data also laboratory data are regularly used. Additionally, physicians and nurses can be contacted directly when DRP have been identified. If pharmacists are on duty on the ward, a patient interview helps to gain a more sophisticated overview of the patients’ situation. Our group perform medication management projects based on monitoring procedures in the wards with direct patient contact, medication and patient chart reviews including electronic prescription, regular participation in ward rounds, questionnaire surveys, and (semi-) structured interviews. This way, we identified DRP and implemented interventional strategies such as information leaflets, algorithm-based decision support or teaching and training sessions as well as interdisciplinary guidelines. After admission to hospital the medication has regularly to be switched to the local drug formulary. For a reconciliation management at admission and also during the patients’ hospital stay the following aspects should be particularly considered in a medication management: indication, effectiveness, dosing, route of administration appropriate and feasible, drug-drug interactions and interactions with food and comorbidity, double prescriptions, duration of therapy, pharmaco-economic consideration such as cost/benefit relations. Patients’ medication data is then to be compared to data from drug information databases, systematic and narrative reviews, original data from RCT (Randomised Controlled Trials), SmPC (Summary of Product Characteristics) and (inter)national guidelines. However, as we found, information gained from different sources can highly differ from each other. Besides Rx-medication (requiring prescription by a physician) also OTC-drugs (Over-The-Counter) and CAM (Complementary and Alternative Medicine) should be taken into account, because patients frequently continue their drugs during their hospital stay without knowledge of the hospital physicians. At discharge, the medication recommended in the discharge letter by the physician should be finally assessed by a pharmacist. Information leaflets directly forwarded to the patient and a direct-to-patient counselling prevent DRP when the patient takes responsibility of drug administration by himself. Home visits with medicine reviews support a safer drug administration including the storage of drugs at home. Apart from patient- and medication-related knowledge, communication skills are essential to gain relevant information as well as to succeed in implementing the recommendations to physicians, nurses, patients, and relatives. Drug information should be offered in a personalized way and in written and verbal form to the responsible physician and (if appropriate) to the patient in agreement with the physician. Considering limited resources we are prioritizing strategies to the most frequent and severe problems according to decision-matrix models. We used particularly monitoring procedures to identify also knowledge-based DRP that are hardly detectable by CIRS (Critical Incident Reporting Systems). We found that the total medication process reaching from prescription to administration should be addressed. By using a feed-back strategy offering information on (in)appropriate drug administration to physicians avoids DRP most effectively. As we have recently shown, DRP derive not only from health care professionals and patients; also relatives and other groups such as teachers of children should be trained in save (emergency) drug administration. To reach sustainable effects we have proven that the pharmaceutical support should be continuously offered. As shown by our group, clinical pharmacists prevent on the one hand DRP and critical adverse drug events at the patient level. On the other hand, guideline adherence and effectiveness increase by strategies offered by pharmacists. To summarize, our results confirm that a structured and systematically performed medication management in routine clinical care is feasible and increases patient safety in drug therapy substantially. Acknowledgments: I would like to thank all patients, relatives, physicians, nurses and pharmacists participating in our studies and supporting our work. Special thanks to the members of my team: Susanne Schiek, Martina P. Neininger, Henriette K. Dumeier, Johanna Freyer, Claudia Greißing, Almuth Kaune, Dorothee Niemann, Pia M. Schumacher, Monika K. Sluzalek, Dominik Wilke, Janine Zimmer, and Sandra Paule. DPhG Annual Meeting 2015 Conference Book • 37 SCIENTIFIC LECTURES SL.19 Medication management in ambulatory care: Preliminary results of the WestGem-Study Rose, O.1,6; Schaffert, C.2; Czarnecki, K.2; Mennemann, H. S.3; John, C.6; Mertens-Keller, D.6; Richling, I.6; Waltering, I.4,6; Hamacher, S.5; Felsch, M.5, Herich, L.5; Köberlein-Neu, J.2 1 Department of Clinical Pharmacy, University of Bonn, An der Immenburg 4, 53121 Bonn Germany. of Health Care Management and Public Health, Schumpeter School of Business and Economics, University of Wuppertal, Rainer-Gruenter-Str. 21 Gebäude FN (1. OG), 42119 Wuppertal, Germany. 3 Muenster University of Applied Science, Robert-Koch-Str. 30, 48149 Muenster, Germany. 4 Department of Pharmacy, University of Muenster, Corrensstr. 48, 48149 Muenster. 5 Institute of Medical Statistics, Informatics and Epidemiology, University of Cologne, Kerpener Str. 62, 50937 Cologne, Germany. 6 Elefanten-Apotheke gegr 1575, Steinstr. 14, 48565 Steinfurt, Germany. 2 Department Background: Pharmaceutical practice worldwide experiences a shift towards patient oriented services. Medication Management (MM) is emphasized as the most prominent pharmaceutical care activity. A profound benefit is expected for the patients, the society and the health care providers. Data for Germany to show the outcomes of a Medication Management is still scarce. Purpose: This first RCT on MTM in Germany aims to provide data on the outcomes of a comprehensive MM by performing repeated PCNE-type-3 Medication Reviews (MR) and interprofessional collaboration. Methods: The study is designed as a pragmatic cluster-randomized controlled trial, involving 12 general practitioner (GP) clinics and 165 patients. It follows a stepped wedge design. Pharmacists receive anonymized data by GPs and home-care specialists. A PCNE type 3 Medication Review (MR) is performed and a SOAP-form is provided to the GPs. The MR is repeated after 6 months. The primary endpoint is a change in the quality of therapy, measured by the Medication Appropriateness Index (MAI). Secondary endpoints are changes in the number of drug related problems (DRPs). Clinical outcomes and interprofessional aspects of the collaboration are evaluated. Results: Preliminary data shows a significant reduction in the MAI-score, which correlates in time with a stronger effect in the patients who entered the intervention-phase earlier. Similar effects can be shown on the reduction of drug related problems (DRPs). A reduction in LDL-Cholesterol can be demonstrated in an eligible subgroup with the required laboratory data. Kidney function in another subgroup of the study with data on serum-creatinine declines less than expected from other studies. Pharmaceutical suggestions are accepted by more than 50% by the GPs. Conclusions: A collaborative MM in Germany can improve the quality of therapy and clinical endpoints. The effect of a Medication Management increases over time. 38 • DPhG Annual Meeting 2015 Conference Book EVIDENCE BASED MEDICATION MANAGEMENT SL.20 AMTS activities of the Pharmacists-Chamber of North-Rhine Krüger, M.1; Derix, S.1 1 Linner Apotheke, Appointee for Phamaceutical Care and AMTS, General Secretary North-Rhine Chamber of Pharmacists Qualification courses and projects, which ensure and enhance the patient oriented medication care in times of demographic and societal change ever since have belonged to the core strategic competencies of the NorthRhine chamber of pharmacists. AMTS and medication management should ideally become qualified and reimbursed pharmaceutical services. The multi-professional pharmacotherapeutic management is a central element of drug safety according to the remarkable and exemplary resolution of the federal state health conference of Northrhine-Westfalia in 2012. Thus, the qualification project ATHINA (Drug Safety in Pharmacies) is a priority amongst multiple measures of the North-Rhine chamber of pharmacists. With focus on geriatric patients the chamber initiated an evaluation study investigating implementation of pharmaceutical risk management systems in geriatric care units through specialized pharmacies. Co-operations and grants for several scientific projects (e.g. DIADEMA) have been established. Results show that AMTS and medication management enhance patient care. Competencies and job satisfaction of pharmacists are fostered as well. Better interdisciplinary collaboration and clearer responsibility definitions should be further optimized to ensure high quality patient care and medication management. The government is requested to ensure the reimbursement of these additional pharmaceutical services adequately. DPhG Annual Meeting 2015 Conference Book • 39 SCIENTIFIC LECTURES SL.21 Comparison of three lists of Potentially Iadequate Medications in old age (PIMs) in four German long-term care facilities with special regard to Adverse Drug Events (ADEs) Hildebrand, J.1,2; Hanke, F.1,2; Jaehde, U.3; Thürmann, P. A.1 1 Institute of Clinical Pharmacology, University of Witten/ Herdecke, Germany Society of Geriatric Pharmacy - Gero PharmCare Ltd, Cologne, Germany 3 Institute of Pharmacy, Clinical Pharmacy, University of Bonn, Germany 2 Background and aims: During the last 25 years, a variety of PIM lists has been developed in several countries. Some studies have shown that PIM use is associated with an increased risk for ADEs [1, 2], although this correlation is still being discussed controversially. We aimed at identifying the applicability and qualification of two international PIM lists for use in a German nursing home cohort in comparison to the German PRISCUS list [2]. Materials and methods: The medications of 339 nursing home residents living in four long-term care facilities in Northrhine-Westphalia between July and December 2010 were screened for PIMs. Observation period was 90 days retrospectively. We compared the German PRISCUS list, the French PIM list [3] and the 2012 Update of the American Beers list [4]. The prevalence of PIM prescription and the most frequently prescribed PIM substances were analysed. Additionally, prevalent ADEs were identified and analysed as described recently [5]. Possible correlations between PIM use and ADE occurrence were analysed using univariate regression analyses (statistical programme Minitab 17). Results: The prevalence of PIM prescriptions came to 45.3% (for regular medications and those documented “as needed”) according to the PRISCUS list and was remarkably higher when applying the French PIM list (50.6%) and even higher using the Beers list (60.9%). There was no statistically significant difference in PIM use between female and male nursing home residents (PRISCUS PIMs: p = 0.825, French PIMs: p = 0.802, Beers PIMs: p = 0.419). The most frequently prescribed PRISCUS PIMs were dimenhydrinate, zopiclone and acetyldigoxin, according to the French PIM list the most frequent substances were sodium-picosulfate, promethazine and dimenhydrinate, and the most frequent Beers PIMs were metoclopramide, risperidone and lorazepam. A correlation between PIM use and ADE occurrence was found for Beers PIMs (p = 0.006), but not for PRISCUS PIMs (p = 0.114) and French PIMs (p = 0.273). Conclusions: PIM prescriptions are very common for nursing home residents in Germany. Astonishingly, the percentage of PIMs was much higher with the Beers and the French PIM list. This comparison shows that there is variability between PIM lists which can be explained by the fact that all these lists base on expert consensus assessment rather than on evidence from randomised controlled trials in the elderly population. Apart from the PRISCUS list, which has been developed specifically for drugs authorised for the German market, international PIM lists may also offer important information for identifying patients at risk for ADEs. An association could be shown between the use of PIM drugs (according to the Beers list) and the occurrence of ADEs, which is in line with findings from other studies in the setting of nursing home residents [6]. References: 1. Dormann, H. et al.: Dtsch. Arztebl. Int. 2013, 110(13): 213–9. 2. Holt, S. et al.: Dtsch. Arztebl. Int. 2010, 107(31-32): 543-51. 3. Laroche, M. L. et al.: Eur. J. Clin. Pharmacol. 2007, 63: 725–31. 4. American Geriatrics Society 2012 Beers Criteria Update Expert Panel: American Geriatrics Society updated Beers Criteria for potentially inappropriate medication use in older adults, J. Am. Geriatr. Soc. 2012, 60 (4): 616–31. 5. Jaehde, U.; Thürmann, P. A.: Z. Evid. Fortbild. Qual. Gesundhwes. 2012, 106(10):712-6. 6. Lau, D. T. et al.: Arch. Intern. Med. 2005, 165(1):68-74. 40 • DPhG Annual Meeting 2015 Conference Book (BIO)ANALYTICS 2.6 (Bio)Analytics Chairs: J. Heilmann, M. Lämmerhofer SL.22 Protein mass spectrometry – Methods and applications in clinical proteomics Stühler, K.1,2 1 2 Institut für Molekulare Medizin, Universitätsklinikum Düsseldorf, Düsseldorf, Germany Molecular Proteomics Laboratory Biologisch-Medizinisches-Forschungszentrum, Heinrich-Heine-Universität, Düsseldorf, Germany In the last years, mass spectrometry (MS) has rapidly developed and beside other applications in proteomics it is meanwhile broadly applied for a wide range of protein analytical questions like for instance characterization of proteins’ primary structure, identification of posttranslational modifications, identification of protein interaction partners and even absolute protein quantification. Typically electrospray ionization MS instruments coupled with high performance nano liquid chromatography (nano LC-ESI-MS) systems are used to address especially quantitative question in biological research. The MS systems are available for high resolution measurements which are necessary for discovery driven analysis of complex protein mixtures using e.g. label based or label-free MS. For targeted quantification of proteins triple-quadrupol systems are indispensable tools and are applied in clinical proteomic. This triple-quadrupol instruments allow the relative and absolute quantification of known proteins by selected reaction monitoring (SRM)-analysis. Here, current research projects will be presented applying the above mentioned techniques for protein identification and quantification to exemplify actual applications of protein mass spectrometry in clinical proteomics. DPhG Annual Meeting 2015 Conference Book • 41 SCIENTIFIC LECTURES SL.23 Capillary electrophoresis-based stereospecific enzyme assay for methionine sulfoxide reductase Scriba, G.1; Zhu, Q.1; Schönherr, R.2; Heinemann, S.2 1 Department 2 Department of Pharmaceutical/Medicinal Chemistry, Friedrich Schiller University, Philosophenweg 14, 07743 Jena, Germany of Biochemistry, Friedrich Schiller University, Philosophenweg 14, 07743 Jena, Germany Under conditions of oxidative stress, L-methionine (Met) residues in peptides and proteins are easily oxidized to L-methionine sulfoxide [Met(O)] by reactive oxygen species [1]. Protein-bound as well as free Met(O) can be reduced by methionine sulfoxide reductase (Msr) enzymes, a group of thiol oxidoreductases, protecting cells against oxidative damage [2]. Because of the chirality of the sulfoxide moiety, Met(O) exists as the pair of diastereomers L-methionine-(S)-sulfoxide [Met-S-(O)] and L-methionine-(R)-sulfoxide [Met-R-(O)]. For the reduction of the diastereomers, stereospecific Msr enzymes exist, which share little sequence homology but possess mirror-like relationships of their active sites. MsrA catalyzes the reduction of either free or protein bound Met-S-(O). In contrast, MsrB reduces Met-R-(O) in proteins but displays only low activity for free Met-R-(O). In addition, an enzyme named free methionine-(R)-sulfoxide reductase (fRMsr), which specifically reduces free MetR-(O), has been found in bacteria [2,3]. In order to determine the stereospecific activity of Msr enzymes, capillary electrophoresis-based assays employing N-acetylated pentapeptides containing a dinitophenyl (Dnp) label at the C-terminus were developed. For example, the diastereomers of ac-Lys-Ile-Phe-Met(O)-Lys-DNP were separated in a background electrolyte using a dual elector system consisting of sulfated β-cyclodextrin and 15-crown-5. The optimized conditions were obtained by experimental design resulting in a 50 mM Tris buffer, pH 7.85, which contained 5 mM 15-crown-5 and 14.3 mg/mL sulfated β-cyclodextrin. The assay was validated and applied to the determination of the stereospecificity of recombinant human and fungal Msr enzymes as well as the determination of the Michaelis-Menten kinetic data of the enzymes. Furthermore, mutations of Aspergillus nidulans MsrA were analysed by using various peptide substrates. Acknowledgments: The financial Support of Q. Zhu by the China Scholarship Council is gratefully acknowledged. References: 1. Voigt, W.: Free Racid. Biol. Med. 1995, 18: 93-105. 2. Broschi-Muller, S.; Gnad, A.; Branlant, B.: Ann. Biochem. Biophys. 2008, 474: 266-273. 3. Lee, B. C.; Gladyshev, V. Y.: Free Racid. Biol. Med. 2011, 50: 211-227. 42 • DPhG Annual Meeting 2015 Conference Book (BIO)ANALYTICS SL.24 New approach for drug discovery and development: Prediction of human drug metabolism using chimeric mice transplanted with human hepatocytes Sanoh, S.1; Tateno, C.2; Ohta, S.1 1 Graduate 2 R&D School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8553, Japan. Dept, PhoenixBio, Co., Ltd., 3-4-1 Kagamiyama, Higashi-Hiroshima, 739-0046, Japan In drug discovery and development, it is essential to predict human drug metabolism and pharmacokinetics (PK) of new chemical entity to estimate efficacy and safety in humans. However, it is often difficult to predict these profiles from animal data because species differences of metabolic activities in drug-metabolizing enzymes are observed between animals and humans. Screening of metabolic stability of candidates for cytochrome P450 (CYP) has generally been conducted using human liver microsomes in pharmaceuticals. Human hepatocytes also have been widely used to predict PK parameters such as clearance (CL) and identify drug metabolites. Hepatocytes are useful to examine both CYP and non-CYP metabolic activities such as UDP-glucuronosyltransferase (UGT) and aldehyde oxidase (AO). Chimeric mice with humanized liver which were generated from urokinase-type plasminogen activator/severe combined immunodeficiency mice transfected with human hepatocytes, are currently used in drug discovery and development. The ratio of replacement of human hepatocytes is more than 70% in the liver of chimeric mice with humanized liver. The expression of human specific metabolizing enzymes, transporters has been confirmed in chimeric mice. Therefore, chimeric mice with humanized liver are expected as in vivo animal model to predict human specific drug metabolism and PK [1,2]. In fact, several reports show that metabolic profiles of drugs mediated by CYP and non-CYP in chimeric mice reflect those in humans. Six metabolites of ibuprofen by CYP and UGT were detected in urine of chimeric mice. The urinary excretion profile was similar to that of humans [3]. Furthermore, metabolic profiles of AO substrates also reflected those of humans although there are significant species differences of AO metabolism between animals and humans [4,5]. In contrast, chimeric mice transplanted with rat hepatocytes have also been developed as the control mice for chimeric mice with humanized liver in consideration of species differences between rats and humans [6]. The comparison of metabolic profiles between rats and chimeric mice with rat hepatocytes could help to improve predictability of human profiles from data of chimeric mice with humanized liver. The utility of chimeric mice transplanted with rat hepatocytes was demonstrated [3-5]. Predictability of not only drug metabolism but also PK profile has been evaluated. Good correlations of hepatic intrinsic CL values between chimeric mice and humans in several drugs metabolized through multiple pathways by CYP and non-CYP were observed although there are some limitations [7]. In addition, each relationship of total CL and the volume of distribution at steady state between predicted from single-species allometric scaling of chimeric mice and observed data of human indicated further good correlations, respectively. The plasma concentration-time curves in chimeric mice were also generally similar to the profiles in humans. However, the predicted plasma concentration-time curve of diazepam from chimeric mice was not superimposable with observed data of humans [8]. In conclusion, chimeric mice with high replacement of human hepatocytes have potential to predict drug metabolism and PK. Therefore, the approach using chimeric mice with humanized liver may contribute to PK/PD study as part of translational research. Furthermore, the development of chimeric mice transplanted with some individual human hepatocytes with various metabolic activities may be helpful to predict inter-individual differences of PK. However, it is necessary to consider the contribution of extra-hepatic organs not humanized such as intestine and metabolism in mouse residual hepatocytes in the liver of chimeric mice with humanized liver. Predictability of diazepam could be improved using chimeric mice with higher replacement exceeding 80% of hepatocytes. Recently, murine cyp3a knockout chimeric mice with humanized liver, which do not express murine cyp3a in intestine and liver, were developed. The mice are expected to reduce the effects of residual mouse metabolic contribution by Cyp3a [9]. In future, humanized mice with gene modification could become the useful animal model. References: 1. Tateno et al.: Am J Pathol 2004, 165(3): 901-912. 2. Sanoh; Ohta: Biopharm Drug Dispos 2014, 35: 71-86. 3. Sanoh et al.: Drug Metab Dispos 2012, 40(12): 2267-2272. 4. Sanoh et al.: Drug Metab Dispos 2012, 40(1): 76-82. 5. Tanoue et al: Xenobiotica 2013, 43(11): 956-962. 6. Utoh et al.: Am J Pathol 2010, 177(2): 654-665. 7. Sanoh et al.: Drug Metab Dispos 2012, 40(2): 322-328. 8. Sanoh et al.: Xenobiotica 2015, 45(7):605-614. 9. Kato et al.: Drug Metab Dispos 2015, 43(8): 1208-1217. DPhG Annual Meeting 2015 Conference Book • 43 SCIENTIFIC LECTURES SL.25 Importance of extrahepatic UDP-glucuronosyltransferase 1A1 in bilirubin metabolism Fujiwara, R.1; Itoh, T.1; Tukey, R. H.2 1 Department 2 Department of Pharmaceutics, School of Pharmacy, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo, 108-8641, JAPAN of Pharmacology, University of California San Diego, La Jolla, California, 92093, USA Background: Accumulating evidence indicates that breast-fed human infants have higher serum levels of bilirubin than formula-fed infants and are at a higher risk for bilirubin-induced neurological dysfunction. However, the mechanism underlying the breast milk-induced jaundice has not been clarified completely. While human infants develop physiological jaundice, experimental mice or rats do not show the induced serum bilirubin level during periods of developmental change, suggesting that the function of UDP-glucuronosyltransferase (UGT) 1A1, which is the sole enzyme responsible for bilirubin metabolism, is different between human and mouse. Methods: We developed humanized UGT1 (hUGT1) mice, in which the original Ugt1 locus was disrupted and replaced with the human UGT1 locus. We fed human breast milk or infant formula to neonatal hUGT1 mice and examined the UGT1A1 expression in the liver and small intestine as well as their serum bilirubin levels. Results: Breast milk-fed hUGT1 mice developed severe hyperbilirubinemia. Overly extreme hyperbilirubinemia occurs in 5-10% of newborn hUGT1 mice, with the TSB levels often reaching > 20 mg/dL, leading to the development of seizures and eventual death due to bilirubin accumulation in brain tissue (Figure). In the hUGT1 mice, serum bilirubin levels gradually increased and the peak was at 14 days after birth (Figure). While UGT1A1 was slightly expressed in the liver, a higher expression of UGT1A1 was observed in the small intestine. Interestingly, the pattern of serum bilirubin during the development correlated well to the expression pattern of intestinal UGT1A1. Formula-fed hUGT1 mice had lower serum levels of bilirubin. We found that the hepatic UGT1A1 level was similar between the breast- and formula-fed hUGT1 mice, but intestinal UGT1A1 was significantry higher in the formulafed hUGT1 mice than in breast-fed ones. A higher expression of intestinal UGT1A1 was observed in the preterm hUGT1 mice, indicating that breast milk is suppressing the expression of UGT1A1 in the small intestine after birth. Conclusions: The reduction of serum bilirubin correlates to the induction of the intestinal UGT1A1 expression in the hUGT1 mice, indicating that UGT1A1 expressed in the gastrointestinal tract plays an important role in bilirubin metabolism [1,2]. Figure: Serum bilirubin levels in neonatal hUGT1 and wild-type mice. References: 1. Fujiwara, R. et al.: Gastroenterology. 2012, 142(1): 109-118. 2. Fujiwara, R. et al.: Proc. Natl. Acad. Sci. USA. 2010, 107(11): 5024-5029. 44 • DPhG Annual Meeting 2015 Conference Book (BIO)ANALYTICS SL.26 Renaissance of supercritical fluid chromatography: Fast and sensitive analysis of polar drugs and their metabolites by hyphenated mass spectrometry Parr, M. K.1; Wüst, B.2; Nägele, E.2; Stanic, M.3; Schmidt, A.1,3 1 Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Str. 2+4, 14195 Berlin, Germany Technologies GmbH, Hewlett-Packard-Str.8, 76337 Waldbronn, Germany 3 Chromicent GmbH, Rudower Chaussee 29, 12489 Berlin, Germany 2 Agilent HPLC is considered as method of choice for the separation of various classes of drugs and their impurities. Pharmacopoeias show increasing numbers of methods for identification, purity and assay of APIs utilizing HPLC based methods. However, some analytes are still challenging as HPLC shows limited resolution capabilities and highly polar analytes interact only insufficiently on the conventional analytical reversed phase columns. Especially in combination with mass spectrometric detection only limited possibilities for alteration in the selectivity of the stationary phase are available. Some highly polar sympathetic drugs and their metabolites showed almost no retention on different reversed phase (RP) columns generally used for HPLC-MS. Even on the more polar RP phases like phenylhexyl their analysis remained challenging or even impossible. Supercritical fluid chromatography (SFC) as orthogonal separation technique to HPLC may help to overcome these issues. To check for the general potential selected polar drugs and drug metabolites were analysed by SFC separation as alternative. All compounds showed sharp peaks, good retention and resulted in retention times clearly separated from the dead time especially for the very polar analytes. Retention times and elution orders using the SFC method are different to both reversed phase and HILIC separations due to the orthogonality of the SFC technique. Short cycle times could be realized. As temperature and pressure strongly influence the polarity of supercritical fluids, a precise temperature and backpressure regulation is required for the stability of the retention times. As CO2 is the main constituent of the mobile phase in SFC solvent consumption and solvent waste are considerably reduced. Acknowledgments: The World Anti-Doping Agency is acknowledged for the financial support of this study within their research grant (14A03KP). DPhG Annual Meeting 2015 Conference Book • 45 SCIENTIFIC LECTURES 2.7 Allosteric Regulation Chairs: K. Mohr, M. Bünemann SL.27 Molecular Insights into the mechanosensitivity of histamine H1 receptors Storch, U.1; Erdogmus, S.1; Hoffmann, C.2; Gudermann, T.1; Mederos y Schnitzler, M.1 1 Walther 2 Institute Straub Institute of Pharmacology and Toxicology, Ludwig Maximilians University, Goethestr. 33, 80336 Munich, Germany of Pharmacology and Toxicology, University of Würzburg, Versbacher Straße 9, 97078, Würzburg, Germany Mechanosensation plays a role in several physiological processes for example in the vascular system where it is important for maintaining blood pressure and blood flow. Our previous findings indicate that in vascular smooth muscle cells Gq/11-protein coupled receptors and especially angiotensin II AT1 receptors are direct mechanosensors mediating myogenic vasoconstriction without the involvement of endogenous agonists. From all receptors we have tested, histamine H1 receptors showed the highest mechanosensitivity. Therefore, we aimed to analyse whether H1 receptors adopt an active receptor conformation which might be different from the agonist induced receptor conformation. For this, the technique of dynamic intramolecular fluorescence resonance energy transfer (FRET) was employed which allows monitoring of conformational changes of the receptor. Cerulean, a stable cyan fluorescent protein was attached to the C-terminus of the H1 receptor and a small tetracysteine-binding motif was inserted at different positions in the third intracellular loop which allows binding of the small yellow fluorescent molecule FlAsH. Agonist stimulations with histamine (100 µM) and mechanical stimulations with hypotonic bath solutions with decreasing osmolarities (275, 250, 225, 200, 150 mOsmol) caused FRET signal decreases. When compared with agonist challenge, mechanical stimulations resulted in significantly larger decreases of the FRET signal. Furthermore, the amplitude of mechanically induced FRET signals was concentration dependent since application of different hypoosmotic solutions with decreasing osmolarities evoked stepwise decreases in FRET amplitudes. Moreover, mechanically induced FRET signals were significantly reduced in the presence of selective H1 receptor blockers. Altogether, our findings point to the fact that histamine H1 receptors are mechanosensitive and adopt distinct active receptor conformations upon mechanical stimulation. 46 • DPhG Annual Meeting 2015 Conference Book ALLOSTERIC REGULATION SL.28 Biased signaling and probe dependence at the chemokine receptor CXCR3 Tschammer, N.1 1 Department of Chemistry and Pharmacy, Medicinal Chemistry, Emil Fischer Center, Friedrich Alexander University, Schuhstraße 19, 91052 Erlangen, Germany Dysfunctions of the chemokine receptor CXCR3 signaling network are linked to a myriad of pathologies including autoimmune diseases, cancer and vascular diseases. The efforts so far failed to produce therapeutics, which specifically modulate the activity of CXCR3 and thus contribute to the symptom relieve or cure. In the contrast to soluble proteins chemokines small synthetic ligands bind to the chemokine receptors at the allosteric site(s) inside the hydrophobic pocket formed by transmembrane helices. In general it can be seen that all important unique features of allosteric receptor control (permissivity, saturation of effect, probe dependence) are embodied in the chemokine receptor system. While this adds a level of complexity to analyses and approaches to drug discovery, it also introduces a remarkable capacity for pharmacologic control of this physiological system for therapeutic advantage. With the use of our allosteric radioligand RAMX3 [1] and novel boronic acid based negative allosteric modulators [2] we were able to detect complex mechanisms of allosteric modulation in this receptor. We discovered the first biased negative allosteric modulator of the receptor that preferentially inhibits the CXCL11-mediated recruitment of β-arrestin 2 over G protein activation [2]. When the second CXCR3 receptor chemokine ligand CXCL10 was used, the signalling bias was lost, indicating very clear probe dependence on the allosteric modulator effect [3]. Such probe-dependent allostery may serve to allow fine tuning of chemokine response in the seemingly redundant arena of multiple chemokine agonists for receptors. Our efforts to develop positive allosteric modulators (PAMs) resulted in a discovery of a potent biased PAM that promotes solely the CXCR3-mediated recruitment of β-arrestin2. Furthermore, our most prominent PAM behaves in the presence of endogenous agonists as functional negative allosteric modulator (NAM), while it causes the receptor desensitation and thus decreases the amount of receptors available for the interaction with endogenous chemokines. This type of compounds could mean a breakthrough in the treatment of inflammatory diseases, because they reduce the responsiveness of CXCR3-expressing cells to endogenous chemokines and thus attenuate chemotaxis, which would ultimately result in reduce recruitment of the CXCR3+ cells at the site of inflammation. Our discoveries expand the ways in which the function of CXCR3 can be manipulated. Moreover they provide new insights into interactions between allosteric ligands, chemokines and their receptor, which could be exploited for drug discovery. Acknowledgments: I thank the German Research Foundation for financial support (DFG; grant TS287/2-1,Graduate Training Schools GRK1910 and GRK1962), and the European COST Action CM1207 (GLISTEN: GPCR–Ligand Interactions, Structures, and Transmembrane Signalling: a European Research Network). References: 1. Bernat et al.: ChemMedChem 2012, 7(8): 1481-1489. 2. Bernat et al.: ACS Chem. Biol. 2014, 9(11):2664-2677. 3. Bernat et al.: ChemMedChem 2015, 10(3): 566-574. DPhG Annual Meeting 2015 Conference Book • 47 SCIENTIFIC LECTURES SL.29 Muscarinic M2 receptor allosterism: Context-sensitive signalling Seemann, W. K.1; Mohr, K.2 1 Department of Pharmacology, University of Cologne, Cologne, Germany and Toxicology Section, Institute of Pharmacy, University of Bonn, Bonn, Germany 2 Pharmacology “Conventional pharmacology” seeks to achieve selective G protein-coupled receptor (GPCR) modulation on the level of binding by discrimination between receptors and receptor subtypes. Here, we shift focus beyond the binding event towards signaling. For this, we employed the M2 subtype of muscarinic acetylcholine receptors, which signals into the Gi/o-pathway of guanyl-nucleotide-binding proteins. M2 receptors are widely distributed in the mammalian body and control a multitude of physiological functions. For example, as previously shown in M2 knock-out mice, M2 signaling includes reduction of pain, but also depression of cardiac function [1]. As cholinergic Gi/o-signaling and adrenergic Gs-signaling converge on the level of intracellular cAMP [2], we hypothesized less effective cholinergic signaling by dualsteric agonists to occur in tissues under conditions of elevated cAMP. In order to probe feasibility of this concept, we applied a novel type of agonist engineered to transduce receptor activation with down-tuned efficacy. These “dualsteric” (bitopic orthosteric/allosteric) ligands activate the receptor protein from the orthosteric transmitter binding site and simultaneously bind to the receptor’s allosteric vestibule [3], thereby limiting the receptors conformational transition to the fully active state [4]. With the main focus on cardiac function, in-vitro and in-vivo measurements showed that dualsteric intrinsic efficacy declines in different M2 receptor expression systems with elevated intracellular cAMP. In conclusion, the dualsteric design concept provides a novel approach for pharmacological selectivity and might provide a new avenue to generate antinociceptive therapeutics that lack major muscarinic side effects. Acknowledgments: W.K.S. was supported by the North-Rhine Westphalia International Graduate Research School BIOTECH-PHARMA at the University of Bonn. References: 1. Gomeza, J. et al.: Proc. Natl. Acad. Sci. USA 1999, 96: 1692-7. 2. Brodde, O. E.; Michel, M. C.: Pharmacol. Rev. 1999, 51: 651-90. 3. Antony, J. et al.: FASEB J. 2009,23(2): 442-50. 4. Bock, A. et al.: Nat. Commun. 2012, 3: 1044. 48 • DPhG Annual Meeting 2015 Conference Book ALLOSTERIC REGULATION SL.30 Detection of unexplored allosteric pockets using a “dummy” ligand approach Hermans, S. M. A.1; Pfleger, C.1; Gohlke, H.1 Department of Mathematics and Natural Sciences, Institute for Pharmaceutical and Medicinal Chemistry, Heinrich-Heine-University, Universitätsstr. 1, 40225, Düsseldorf, Germany 1 Allosteric regulation is the coupling between separated sites in biomacromolecules such that an action at one site changes the function at a distant site. The identification of novel allosteric pockets is complicated by the large variation in allosteric regulation, ranging from rigid body motions to disorder/order transitions, with dynamically dominated allostery in between [1]. Here, we present a new and efficient approach to probe information transfer through proteins in the context of dynamically dominated allostery that exploits ”dummy” ligands as surrogates for allosteric modulators. In a preliminary study to test the general feasibility, the approach was applied to conformations extracted from a MD trajectory of the holo and apo structures of LFA1. The grid-based PocketAnalyzer program [2] is used to detect putative binding sites. “Dummy” ligands were generated for each detected pocket along the ensemble. Finally, the Constraint Network Analysis (CNA) software, which links biomacromolecular structure, (thermo-)stability, and function, is used to probe the allosteric response by monitoring altered stability characteristics of the protein due to the presence of the “dummy” ligand [3,4,5]. The results were compared to those of the holo structure with the bound allosteric ligand to validate the “dummy” ligand approach. Remarkably, the usage of “dummy” ligands almost perfectly reproduced the results obtained from the known allosteric effector. Although it turned out that the intrinsic rigidity of the “dummy” ligands over-stabilizes the LFA1 structure, these results are already encouraging. Even for the LFA1 apo structures, where the allosteric pocket is partially closed, the results are in agreement with known allosteric effectors. Overall, the results obtained from the validation of the “dummy” ligand approach are encouraging. This suggests that our “dummy” ligand approach for the characterization of unexplored allosteric pockets is a promising step towards identifying novel drug targets. References: 1. Motlagh, H. N. et al.: Nature 2014, 508 (7496): 331–339. 2. Craig, I. R. et al.: J. Chem. Inf. Model. 2011, 51: 2666–2679. 3. Pfleger, C. et al.: J. Chem. Inf. Model. 2013, 53: 1007–1015. 4. Krüger, D. M. et al.: Nucleic Acids Res. 2013, 41 (April): 340–348. 5. Pfleger, C.: Ensemble-Based Framework for Analyzing Dynamically Dominated Allostery (Doctoral Thesis, Heinrich Heine University, Düsseldorf, Germany) 2014 DPhG Annual Meeting 2015 Conference Book • 49 SCIENTIFIC LECTURES 2.8 Anti-infective Compounds Chairs: U. Holzgrabe, P. Proksch SL.31 Vinylsulfone-based inhibitors of rhodesain as new antitrypanosomal compounds Schirmeister, T.1 1 Institut für Pharmazie und Biochemie, Johannes Gutenberg-Universität Mainz, Germany Vinylsulfones are a well-known and promising class of irreversible inhibitors of cysteine proteases (see Fig. 1). An example is K11777, a compound under clinical development for parasitic diseases like Chagas disease [1] or African trypanosomiasis. The major cysteine protease of the parasite Trypanosoma brucei rhodesiense causing the African sleeping sickness is rhodesain which is e.g. involved in parasite infiltration into the central nervous system [2]. Starting from the structure (pdb: 2P7U) of the enzyme-inhibitor complex of rhodesain with K11777[3] we are developing new vinylsulfones[4] with altered inhibitory properties in terms of chemical reactivity and affinity, using an iterative design cycle consisting of quantum mechanical/molecular mechanical (QM/MM) computations, docking, syntheses, and testing. Figure 1: Mechanism of inhibition of cysteine proteases by K11777. Acknowledgments: Financial support by the DFG (SFB 630) is gratefully acknowledged. References: 1. Rhee, S.-W. et al.: J. Label Compd. Radiopharm. 2013, 56: 461-463. 2. Ettari, R. et al.: Med. Res. Rev. 2010, 30: 136-167. 3. Kerr, I. D. et al.: J. Biol. Chem. 2009, 284: 25697-25703. 4. Schneider, T. et al.: New J. Chem. 2015, 39: 5841-5853. 50 • DPhG Annual Meeting 2015 Conference Book ANTI-INFECTIVE COMPOUNDS SL.32 Discovery of dengue protease inhibitors with nanomolar affinity Klein, C. D.1 1 Institut für Pharmazie und Molekulare Biotechnologie, Universität Heidelberg, Im Neuenheimer Feld 364, D-69120 Heidelberg, Germany This presentation will summarize research that resulted in the discovery of inhibitors of dengue virus protease with affinity in the nanomolar range. Infections with dengue virus and related flaviviruses are a major health concern in the tropical and subtropical climates, and an expansion of the vectors and viruses into temperate zones is to be expected. In analogy to the closely related hepatitis C virus, the protease of dengue virus represents an attractive drug target for the discovery of antiviral drugs [1]. Our work started with the development of a robust assay procedure [2,3], followed by the screening of potential covalent-reversible electrophilic fragments and other small molecules [4,5], and continued with fragment-merging and optimization based on the preferred substrate recognition sequence [6,7]. This allowed us to increase the affinity of dengue protease inhibitors by more than three orders of magnitude, while maintaining ligand efficiency and selectivity towards other serine proteases. A major factor in this success was the evaluation of non-natural amino acids with modulated or abolished basicity as replacements for a key arginine residue. Building blocks with potential promiscuity liabilites could be eliminated in the optimization process. Future tasks are the optimization of PK properties and the antiviral activity in cell culture, which remain unsatisfying. In this respect, the introduction of electrophilic fragments with covalent-reversible binding behavior to the catalytic serine appears to be attractive, in that pharmacokinetic factors such as fast metabolic clearance become less critical. Acknowledgments: The work summarized here was supported by grants from the Deutsche Forschungsgemeinschaft, Studienstiftung des Deutschen Volkes and Konrad-Adenauer-Stiftung. References: 1. Nitsche, C. et al.: Chem. Rev. 2014, 114: 11348–11381. 2. Steuer, C. et al.: J. Biomol. Screening 2009, 14: 1102-1108. 3. Nitsche, C.; Klein, C. D.: Methods Mol. Biol.2013, 1030: 221-236. 4. Nitsche, C.; Steuer, C.; Klein, C. D.: Bioorgan. Med. Chem. 2011, 19: 7318-7337. 5. Steuer, C. et al.: Bioorgan. Med. Chem. 2011, 19: 4067-4074. 6. Behnam, M. A. et al.: ACS Med. Chem. Lett. 2014, 5: 1037. 7. Bastos Lima, A. et al.: Bioorg. Med. Chem. 2015, in press. DPhG Annual Meeting 2015 Conference Book • 51 SCIENTIFIC LECTURES SL.33 The lectin LecB as target for anti-infectives against chronic Pseudomonas aeruginosa infections Titz, A.1 1 Chemical Biology of Carbohydrates, Helmholtz-Institute for Pharmaceutical Research Saarland, 66123 Saarbrücken, Germany The opportunistic pathogen Pseudomonas aeruginosa is currently considered as a major threat to public health care. The Gram-negative bacterium forms biofilms which protects itself from host defense and antibiotic therapy resulting in chronic infections. The bacterial lectin LecB is a virulence factor and plays a prominent role in biofilm formation. Inhibition of lectin function with carbohydrate-based ligands was shown to disrupt bacterial biofilm formation. Based on the crystal structure of the lectin with its glycan ligands, we dissected the contributions of individual functional groups to protein binding in a biophysicsguided approach. This knowledge was then used for the development of small and drug-like glycan-based molecules as LecB inhibitors as future anti-biofilm compounds in chronic Pseudomonas aeruginosa infections. References: 1. Sommer, R. et al. ChemistryOpen 2015, doi: 10.1002/open.201500162. 2. Hofmann, A. et al.: Carbohydr. Res. 2015, 412, 34-42. 3. Sommer, R.; Exner, T. E.; Titz, A. PLoS ONE 2014, 9(11): e112822. 4. Hauck, D. et al.: ACS Chem. Biol. 2013, 8(8), 1775–1784. 52 • DPhG Annual Meeting 2015 Conference Book ANTI-INFECTIVE COMPOUNDS SL.34 Chlorflavonin inhibits growth of Mycobacterium tuberculosis by targeting branched-chain amino acid biosynthesis Rehberg, N.1; Akone, H. S.2; Proksch, P.2; Kalscheuer, R.1 1 Institute for Medical Microbiology and Hospital Hygiene, Heinrich-Heine-University Düsseldorf, Universitätsstr. 1, 40225 Düsseldorf, Germany 2 Institute of Pharmaceutical Biology and Biotechnology, Heinrich-Heine-University Düsseldorf, Universitätsstr. 1, 40225 Düsseldorf, Germany Mycobacterium tuberculosis, the etiologic agent of Tuberculosis (TB), is one of the leading causes of human mortality and morbidity due to pathogenic microorganisms. In recent years treatment of TB is increasingly facing serious problems due to the alarming rise of multidrug-resistant (MDR) and extensively drug-resistant (XDR) Mycobacterium tuberculosis strains, the latter of which are virtually untreatable with currently available anti-TB drugs. Thus, there is an urgent need for new antibiotics against TB. Employing bioactivity guided fractionation, chlorflavonin was isolated from an ethylacetate extract obtained from the endophytic fungus Mucor irregularis, which has been isolated from the Cameroonian medicinal plant Moringa stenopetala. Chlorflavonin exhibited strong growth inhibitory activity in vitro against Mycobacterium tuberculosis (MIC90 1.5 μM), which was strictly dependent on presence of the chlorine atom. In contrast to its potent antitubercular activity, chlorflavonin exhibited no cytotoxicity towards the human fibroblast cell line MRC-5 and toward the macrophage like human acute monocytic leukemia cell line THP-1 up to concentrations of 100 µM. The isolation of spontaneous resistant mutants against chlorflavonin, which occurred at a low frequency of 10-7, allowed us to understand its mechanism of action. Mapping of resistance-mediating mutations employing whole-genome sequencing as well as chemical supplementation assays revealed that chlorflavonin specifically inhibits the acetohydroxyacid synthase IlvB1, which mediates the first common step in branched chain amino acids (BCAA) and pantothenic acid biosynthesis. Thus, chlorflavonin inhibits growth of Mycobacterium tuberculosis by causing combined auxotrophy to BCAA and pantothenic acid. While exhibiting a bacteriostatic effect in monotreatment, chlorflavonin displayed synergistic effects with the first-line antibiotic isoniazid leading to a complete sterilization and no resistance in liquid culture in combination treatment. Using a fluorescent reporter strain, intracellular activity of chlorflavonin against Mycobacterium tuberculosis inside infected macrophages was demonstrated that was superior to streptomycin treatment. Acknowledgments: This work was supported by the Jürgen Manchot Foundation and the Research Commission of the Medical Faculty of the Heinrich-Heine-University Düsseldorf. DPhG Annual Meeting 2015 Conference Book • 53 SCIENTIFIC LECTURES SL.35 Marine natural products as potential sources for new antitubercular agents Daletos, G.1 1 Institute of Pharmaceutical Biology and Biotechnology, Heinrich-Heine University, Universitaetsstrasse 1, 40225 Düsseldorf, Germany Tuberculosis (TB) remains one of the world’s lethal infectious diseases. In 2013, the World Health Organization (WHO) reported 9.0 million new cases and 1.5 million deaths caused by TB worldwide [1]. Marine organisms represent a rich source of structurally unique compounds that can be utilized for the search for new antitubercular drugs from nature [2]. Invertebrates, such as sponges, have hitherto provided the largest number of secondary metabolites with unique structural features and pronounced biological activities [3]. Our ongoing search for new anti-TB marine natural products already yielded several promising compounds including manzamine A [4], aerothionin [5], sceptrin [6], and papuamine [7]. Among these active metabolites a group of unusual cyclic peptides called callyaerins, from the Indonesian sponge Callyspongia aerizusa appeared particularly interesting for further investigation. The planar structures of the isolated compounds were unambiguously elucidated on the basis of 1D and 2D NMR spectroscopic data and MS interpretation. The absolute configurations of their constituent amino acid residues were determined using the Marfey’s method [8]. The basic structural unit of the callyaerins comprises a cyclic peptide with a linear peptide side chain, both of variable size, linked through a non-proteinogenic (Z)-2,3diaminoacrylic acid (DAA) functional group [9,10]. All compounds were investigated in vitro against Mycobacterium tuberculosis, as well as against THP-1 (human acute monocytic leukemia), and MRC-5 (human fetal lung fibroblast) cell lines in order to assess their general cytotoxicity. Callyaerins were found to inhibit M. tuberculosis at low micromolar concentrations, which highlights the potential of these compounds as promising anti-TB agents. Acknowledgments: My sincere gratitude to Prof. Dr. P. Proksch (Heinrich-Heine University, Duesseldorf, Germany) for his admirable supervision and expertise guidance during my work. I wish to thank Dr. R. Kalscheuer (Heinrich-Heine University, Duesseldorf, Germany) for carrying out the experiments with M. tuberculosis. References: 1. Global tuberculosis report 2014; World Health Organization: Geneva, Switzerland, 2014. 2. García, A. et al.: Eur. J. Med. Chem. 2012, 49, 1-23. 3. Proksch, P. et al.: Phytochem. Rev. 2010, 9, 475-489. 4. Sakai, R. et al.: J. Am. Chem. Soc. 1986, 108, 6404-6405. 5. Fattorusso, E. et al.: J. Chem. Soc. D 1970, 12, 752-753. 6. Walker, R. P.; Faulkner, D. J.: J. Am. Chem. Soc. 1981, 103, 6772-6773. 7. Baker, B. J.; Scheuer P. J.; Shoolery J. N.: J. Am. Chem. Soc. 1988, 110, 965-966. 8. Marfey, P.: Carlsberg Res. Commun. 1984, 49, 591-596. 9. Ibrahim, S. R. M. et al.: ARKIVOC 2008, xii, 164-171. 10. Ibrahim, S. R. M. et al.: Biorg. Med. Chem. 2010, 18, 4947-4956. 54 • DPhG Annual Meeting 2015 Conference Book POORLY SOLUBLES 2.9 Poorly Solubles Chairs: P. Kleinebudde, W. Weitschies SL.36 The use of screening tools in the development of amorphous solid dispersions Page, S.1 1 Pharmaceutical Research and Development, F. Hoffmann-La Roche Ltd, CH-4070 Basel, Switzerland With the implementation of high-throughput screening in drug development the number of poorly soluble compounds increased over time. This is not only reflected in the development pipelines in the pharmaceutical industry, but also in the number of publications in the area of poorly soluble compounds and in particular in the field of amorphous solid dispersions. Solid dispersions can be classified according to the physical state of their components as well as the type of stabilizer used. The first generation of solid dispersions contained crystalline carriers, whereas the second generation of solid dispersions consists of the drug substance and an polymeric carrier [1]. These polymeric carriers are added to the system in order to stabilize the amorphous form of the drug and/or modify their dissolution behaviour. In particular the later property led to the third generation of solid dispersions, which contain a mixture of a polymer and surfactant, a mixture of polymers or surfactants [1]. An initial stage in the development of amorphous solid dispersions is therefore the selection of an appropriate polymeric carrier and/or surfactant with the aim to obtain a physical stable amorphous system showing a high degree of supersaturation. A screening assay should therefore focus on two aspects: physical stability and drug release. Over the years, different screening tools where developed to enable a good and quick selection of promising prototype formulations which are later manufactured and tested in vivo. An overview on different screening tools is presented in this talk [2,3,4], which are used for the selection of an appropriate formulation composition. In addition, examples underlining the suitability of certain analytical tools for the later development of the process [5,6] are given. Acknowledgments: The presenter would like to thank Nicole Wyttenbach, Matthias Eckhard Lauer, Monira Siam, Olaf Grassmann, Reto Maurer, Laurence Jacob, Joseph Tardio, Emmanuel Scheubel, Christine Janas, Christoph Marschall, Jochem Alsenz, Johannes H Kindt and Andreas Engel for the collaboration in the last years which enabled the development of suitable screening tools, which foster formulation development of amorphous solid dispersions and solid solutions. References: 1. Vasconcelos, T.: Drug Discov. Today 2007, doi: 10.1016/j.drudis.2007.09.005. 2. Wyttenbach, N. et al.: Eur. J. Pharm. Biopharm. 2013 84(3):583-98. 3. Lauer, M.-E. et al.: Pharm. Res. 2011 28(3):572-84. 4. Lauer, M.-E. et al.: Pharm. Res. 2013 30(8):2010-22. 5. Lamm, M. S. et al.: AAPS PharmSciTech 2015, doi: 10.1208/s12249-015-0387-9. 6. Marschall, C.´; Maurer, R.; Page, S.: 1st European Conference on Pharmaceutics – Drug Delivery 2015, Reims. DPhG Annual Meeting 2015 Conference Book • 55 SCIENTIFIC LECTURES SL.37 Colloidal carrier systems for the formulation of poorly water-soluble drugs Bunjes, H.1 1 Institut für Pharmazeutische Technologie, TU Braunschweig, Mendelssohnstr. 1, 38106 Braunschweig, Germany Poorly soluble drugs pose a considerable formulation challenge, in particular with regard to their peroral and parenteral administration. The need for finding strategies enabling their pharmaceutical administration becomes increasingly urgent as more and more drug candidates emerging from the development pipeline display poor aqueous solubility. An interesting option to formulate these substances is the use of colloidal carrier systems which even allow intravenous administration due to the small size of the particles acting as drug carriers. Among the different types of colloidal carrier systems available, lipid dispersions are particularly promising since they mainly consist of components that can be considered to be physiologically well compatible. Accordingly, colloidal lipid dispersions like fat emulsions or liposomes have proven to be a valuable option for the delivery of poorly soluble drugs over many years of therapeutic use. A broad variety of different colloidal lipid carriers systems (emulsions, liposomes, mixed micelles with phospholipids, solid and liquid crystalline lipid nanoparticles) is available to the pharmaceutical formulation scientist. Thus, the question arises which of them would be the most promising for a given drug. In order to address this question in an efficient way, our group has started to develop a screening method that is based on the loading of preformed carrier dispersions with the drug in the bulk state. After incubation of the drug with the dispersion, excess drug is filtered off and the amount of drug loaded into the dispersion can be determined analytically [1]. This method has already been successfully employed to select promising carrier dispersions for an antileishmanial drug candidate [2]. It has also proven to be very helpful in solving more fundamentally oriented questions as which parameters affect the loading capacity of carrier particles prepared from a given lipid [3]. As the latter study required the availability of lipid dispersions with different, very well defined particle size, the newly established method of membrane emulsification was included in the processes of dispersion preparation [4]. Particle size and physical state of the particle matrix were found to be of influence depending, however, on the type of drug substance under investigation. While, for example, curcumin and amphotericin B could better be loaded in dispersions containing solid triglyceride particles fenofibrate showed a high preference for the corresponding emulsion particles. The use of dispersions containing smaller particles was always favorable for drug loading albeit to a different extent [3]. Current work on this method is directed towards understanding the factors that determine the extent of drug loading and the loading kinetics. Very slow kinetics are probably the reason why this passive loading approach fails for some drugs that are known to be stably incorporated into the respective dispersions when they are processed in the conventional way by directly loading them during dispersion preparation. Acknowledgments: The scientific contributions of Eva Kupetz, Katrin Göke, Karin Rosenblatt, Sonja Joseph, and Sandra Gehrmann are gratefully acknowledged as is financial support from the Deutsche Forschungsgemeinschaft and the Niedersächsisches Ministerium für Wissenschaft und Kultur. References: 1. Rosenblatt, K.; Bunjes, H.: Poster, 6th World Meeting Pharm., Biopharm., Pharm. Technol., Barcelona, April 2008. 2. Kupetz, E. et al.: Eur. J. Pharm. Biopharm. 2013, 85(3A): 511-520. 3. Kupetz, E.; Bunjes, H.: J. Control. Release 2014, 189: 54-64. 4. Joseph, S.; Bunjes, H.: J. Pharm. Sci. 2012, 101(7): 2479-2489. 56 • DPhG Annual Meeting 2015 Conference Book POORLY SOLUBLES SL.38 Oligomeric cross-linkers for hydrogel formulation from ECM-derived macromolecules Hacker, M.1 1 Pharmaceutical Technology, Institute of Pharmacy, Universität Leipzig, Leipzig, 04317, Germany Hydrogels are well-established delivery devices for APIs of different molecular sizes and properties, including small molecules, proteins, peptides and nucleic acids. Hydrogel materials have also gained attention as widely adjustable artificial extracellular matrices (ECM) for applications in regenerative medicine and tissue engineering. In these applications the control of hydrogel mechanical characteristics, micro- and nano-structural properties and biological interactions with cells and tissues are of key importance. ECM components or degradation products thereof such as collagen, hyaluronan, elastin or gelatin, have become interesting hydrogel building blocks due to their availability, biocompatibility, degradability and biological function. In order to control stability of such ECM component-derived gels, cross-linking is typically necessary. Despite the availability of numerous cross-linking strategies, one that achieves high degrees of cross-linking with low toxicity concerns and options for additional functional modifications is still to be established. We have recently described the synthesis and multi-functionality of maleic anhydride(MA)containing oligoacrylamides that have a high reactivity against primary amines and can be used for the synthesis of cytocompatible hydrogels from macromolecules with amine functionalities [1]. With these oligomeric MAcontaining cross-linkers (abbreviated oPNMA1), we developed and characterized a platform of gelatin-based hydrogels and microparticles that can be easily derivatized by pre-cross-linking functionalization of oPNMA with monovalent amines [2]. This contribution illustrates our current strategies to expand this platform of chemically cross-linked hydrogels towards different applications and physico-chemical functionalities important for regenerative applications. First, we made use of the variety of readily available acrylate and acrylamide monomers in order to find suitable comonomers that impart further functional characteristics to the oligomeric MA-containing cross-linkers and ultimately to the resulting hydrogels. We have integrated diacetone acrylamide which introduced ketone groups to the oligomer chains and characterized the ketone reactivity with the objective to immobilize small molecules but also biological macromolecules to cross-linked hydrogels via biocomjugation using this new functional group. Through the selection of predominantly hydrophilic acrylates or acrylamides we were able to synthesize MA-containing oligomers that allowed for the formulation of injectable gelatin-based hydrogels. Second, tubular hydrogel conduits were fabricated by means of a static mixer and a tube molding procedure. The tubes were extensively characterized and are envisioned for applications in peripheral nerve regeneration. In these attempts we also focus on hydrogel derivatization with small molecules to control cell adhesion and differentiation. Acknowledgments: The authors thank the German Research Council (DFG SFB/Transregio 67 A1) for financial support. MCH gratefully acknowledges a travel grant by the Galenus Privatstiftung. References: 1. Loth, T. et al.: React. Funct. Polym. 2013, 73: 1480-1492. 2. Loth, T. et al.: Biomacromolecules. 2014, 15: 2104-2118. DPhG Annual Meeting 2015 Conference Book • 57 SCIENTIFIC LECTURES SL.39 Contribution of Confocal Raman Microscopy (CRM) to the validation of an ex vivo skin penetration method Lunter, D.1 Department of Pharmaceutical Technology, Institute of Pharmacy and Biochemistry, Eberhard Karls University Tuebingen, Auf der Morgenstelle 8, 72076 Tuebingen, Germany 1 Skin diseases are conventionally treated with topical formulations. Depending on the disease, the active may be intended to act on the skin surface (e. g. disinfectants) or within the skin (e. g. antipruritics). It is therefore of interest to evaluate the concentration of the active within the skin, particularly if an intradermal action is aimed for. To this end, in vivo, ex vivo or in vitro penetration experiments can be conducted. Conventionally, ex vivo and in vitro experiments rely on the segmentation of the skin and subsequent extraction and quantification of the active. The stratum corneum usually withholds the highest amount of the active. In order to prevent bias of the drug amounts detected in the deeper skin layers, it is pivotal to ensure complete separation of the stratum corneum from the deeper skin layers. This can be done by tape stripping, cyanoacrylates biopsy, trypsinization or cryo-segmentation. In this work the effectiveness of tape stripping, cyanoacrylates biopsy and cryo-segmentation to remove the stratum corneum is evaluated by confocal Raman microscopy (CRM) and an ex vivo skin penetration method is validated on the basis of this evaluation. As CRM can give spatially resolved information of the chemical composition of a sample, it can be employed to distinguish between the stratum corneum and deeper skin layers as they exhibit different chemical compositions and therefore give different Raman signals. In the course of the experiment skin samples were either treated with 30 tape strips, one or five cyanoacrylates biopsies or cryo-segmentation. Then cross sections of the samples were prepared and average spectra of the skin surfaces were acquired. For the subsequent analysis, the spectra were normalized. The normalized average spectra were compared to the normalized average spectrum of the viable epidermis which was extracted from untreated skin samples. This was done by calculating the root mean square of the differences between the normalized average spectra of the differently treated samples and the normalized average spectrum of the viable epidermis. The separation method that yielded the lowest values removed the stratum corneum most effectively and was selected to be used in a penetration experiment. Here, a hydrophilic procaine HCl containing gel with or without the penetration enhancer POE-23-lauryl ether served as model formulation [1,2]. The results were validated by CRM. To this end, skin samples were incubated with the model formulations and cross sections were made with a cryo-microtome. The cross sections were subsequently analysed by CRM. Colour coded images of the procaine distribution within the skin were calculated. Furthermore, the procaine content in the skin samples was evaluated by calculating the procaine-peak intensities in the Raman maps. The analysis of the Raman spectra showed that the deviations from the spectrum of the viable epidermis decreased in the order: cryo-segmented skin>tape stripped skin>skin biopsied once>skin biopsied five times. Further analysis of the spectra revealed, that tape stripping did not remove the SC completely and that the use of a cryo-microtome lead to removal of not only the stratum corneum but also of parts of the viable epidermis. In the case of the cyanoacrylates biopsies, one biopsy did not lead to complete removal of the SC but five biopsies were found to remove the SC completely. Furthermore, the deviation of the spectrum of the five times biopsied skin sample was found to be almost as low as the deviations between spectra from the viable epidermis of different skin samples. Thus, a penetration experiment was conducted with this method. It could be shown that the method was capable of elucidating the effect of the penetration enhancer on the skin penetration of procaine HCl. CRM penetration experiments were performed with the same formulations. They yielded similar results and thereby confirmed the feasibility of the method. It was thus proven that the method of cyanoacrylates biopsy is most effective in separating the stratum corneum from the deeper skin layers. The developed method was capable of detecting differences in penetrated amounts of the model active which were induced by the addition of a penetration enhancer. CRM was found to be a valuable tool in the validation of the penetration method. Acknowledgments: Institute of Experimental Medicine, University of Tuebingen, Schenk, M. References: 1. Shin, C-S. et al.: Int. J. Pharm. 2004, 287: 73-78. 2. Lunter, D.; Daniels, R.: I. Biomed. Opt. 2014, 19: 126015-126027 58 • DPhG Annual Meeting 2015 Conference Book PPP IN DRUG DEVELOPMENT 2.10 PPP in Drug Development Chairs: S. Knapp, F. Bracher SL.40 A PPP for the development of chemical tool compounds Knapp, S.1,2,3 1 Johann Wolfgang Goethe-University, Institute for Pharmaceutical Chemistry, Building N240 Room 3.03, Max-von-Laue-Str. 9, D-60438 Frankfurt am Main. 2 Buchmann Institute for Molecular Life Sciences, Riedberg Campus, Max-von-Laue-Str. 15, D-60438 Frankfurt am Main 3 University of Oxford, Nuffield Department of Clinical Medicine, SGC and Target Discovery Institute, Oxford, OX3 7DQ, United Kingdom Selective small molecule inhibitors (chemical probes) have a major impact on our understanding of human biology and for the validation of novel disease associated targets for the development of new treatment therapies. However, the development and characterization of chemical probes is a cost intensive multidisciplinary process requiring significant efforts in medicinal chemistry, structural biochemistry, screening and cell biology that can rarely be accomplished by an isolated laboratory [1,2]. These complications led to many probe molecules that have initially been characterized inadequately and have since been proven to be nonselective or associated with poor characteristics such as the presence of reactive functionality that can interfere with common assay features. Unfortunately such reagents are still widely using in academic research leading to wrong target-disease association. To tackle this challenging problem and to combine expertise and resources in different areas of chemical biology we formed a large multinational group involving academic research laboratories and also currently 8 large pharmaceutical companies. This consortium was established based on the Structural Genomics Consortium (SGC) open access model, which distributes and publishes reagents promptly and without constrains imposed by intellectual property. The consortium has focussed initially on chemical tools developed in the epigenetic target area but it has now been extended to include protein kinases, protein interaction and potential targets identified by genetic analysis such as genome wide association studies. In this presentation I will present the general setup of the project as well as some selected examples (e.g. BET bromodomain inhibitors [3,4] and protein kinases [5]) that led to comprehensive validation of new disease targets and to a fast translation into clinical studies. Acknowledgments: This work was supported by the SGC, a registered charity (number 1097737) that receives funds from AbbVie, Bayer Pharma AG, Boehringer Ingelheim, Canada Foundation for Innovation, Eshelman Institute for Innovation, Genome Canada, Innovative Medicines Initiative (EU/EFPIA), Janssen, Merck & Co., Novartis Pharma AG, Ontario Ministry of Economic Development and Innovation, Pfizer, São Paulo Research Foundation-FAPESP, Takeda, and Wellcome Trust. References: 1. Arrowsmith, C. H. et al.: Nat. Chem. Biol. 2015, 11(8), 536–541. 2. Knapp, S. et al.: Nat. Chem. Biol. 2013, 9(1), 3-6. 3. Filippakopoulos, P. et al.: Cell, 2012, 49 (1), 214-231. 4. Filippakopoulos, P.; Knapp, S.: Nat. Rev. Drug. Discov. 2014, 13 (5), 337-356. 5. Gammons, MV. et al.: Invest Ophthalmol, 2013, 54 (9), 6052-6062. DPhG Annual Meeting 2015 Conference Book • 59 SCIENTIFIC LECTURES SL.41 Public Private Partnerships in lead discovery: Overview and case study on binding kinetics Müller-Fahrnow, A.1 1 Bayer Healthcare AG, 13353 Berlin Pharmaceutical industry is faced by significant challenges in its effort to discover new drugs that address unmet medical needs. Safety concerns and lack of efficacy are the two main technical reasons for attrition. Improved early research tools including predictive in vitro and in vivo models as well as a deeper understanding of the disease biology therefore have the potential to improve success rates. In order to foster innovation and to meet the challenges, we need to combine internal activities with external collaborations in line with the interests and needs of all partners. Different types of partnerships ranging from fee-for-service activities to strategic partnerships with significant and long-term partnerships will be discussed [1]. Kinetics for Drug Discovery (K4DD) is one example for a public private partnership in which Bayer Healthcare is actively involved. The overall aim of the K4DD project, supported by the Innovative Medicines Initiative (IMI), is to enable the adoption of drug-target binding kinetics analysis in the drug discovery decision-making process [2]. Together with 19 other partners, we have defined three focus areas, the so-called work packages: 1. Understanding of binding kinetics at a molecular level Ability to design appropriate kinetic behaviour rationally 2. Assay technologies Information feedback in line with cycle times in drug discovery 3. Understanding translation to complex systems Confidence that differences in binding kinetics lead to differentiation in cell-based assays and in vivo The status of the project including our work on a new database for binding kinetics will be presented. Acknowledgments: K4DD has received support from the Innovative Medicines Initiative Joint Undertaking under grant agreement no. 115366, 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. More info: www.imi.europa.eu References: 1. Lessl, M. et al.: Angew. Chem. Int. Ed. Engl. 2013, 2684-268 2. Copeland, R. et al.: Nature Reviews Drug Discovery 2006, 5 (9), 730-739 60 • DPhG Annual Meeting 2015 Conference Book PPP IN DRUG DEVELPOMENT SL.42 Development of a selective DYRK1A inhibitor in a PPP framework Kunick, C.; Falke, H.1; Chaikuad, A.2; Becker, A.1; Loaëc, N.3,4; Lozach, O.3,4; Abu Jhaisha, S.5; Becker, W.5; Jones, P. G.6; Preu, L.1; Baumann, K.1; Knapp, S.2; Meijer, L.3,4 1 Technische Universität Braunschweig, Institut für Medizinische und Pharmazeutische Chemie, Beethovenstraße 55, 38106 Braunschweig, Germany. 2 University of Oxford, Nuffield Department of Clinical Medicine, Structural Genomics Consortium, Old Road Campus Research Building, Roosevelt Drive, Headington, Oxford OX3 7DQ, U.K. 3 ManRos Therapeutics, Perharidy Research Center, 29680 Roscoff, Bretagne, France. 4 CNRS, Station Biologique de Roscoff, ‘Protein Phosphorylation & Human Disease’ group, 29680 Roscoff, France. 5 RWTH Aachen University, Institute of Pharmacology and Toxicology, Wendlingweg 2, 52074 Aachen, Germany. 6 Technische Universität Braunschweig, Institut für Anorganische und Analytische Chemie, Hagenring 30, 38106 Braunschweig, Germany. Dual-specificity tyrosine phosphorylation-regulated kinase 1A (DYRK1A) is a protein kinase which is constitutively activated by autophosphorylation at Tyr321. After activation, DYRK1A phosphorylates serine or threonine residues preferentially on substrates with small hydrophobic residues at the P+1 position [1]. Physiological mechanisms regulated by DYRK1A comprise cell cycling, Notch signaling, apoptosis, and pre-mRNA splicing, etc. The gene encoding for DYRK1A is located on chromosome 21 which is present in three copies in Down Syndrome (DS, or trisomy 21), the most frequent congenital reason of mental retardation. DS individuals indeed show increased DYRK1A expression in the brain [2,3]. Several observations also link DYRK1A to Alzheimer’s disease (AD). DYRK1A acts as a priming kinase for phosphorylation of Tau protein by glycogen synthase kinase-3 (GSK-3) and thus may contribute to the formation of hyperphosphorylated Tau aggregates which appear as intracellular neurofibrillary tangles in the brains of AD patients. By phosphorylating the β-Amyloid Precursor Protein (APP), DYRK1A increases APP cleavage by γ- and β-secretases. This cleavage produces amyloid peptides which form extracellular plaques and lead to DYRK1A upregulation. Based on this evidence, DYRK1A has been suggested as a target for the development of drugs against neurodegenerative diseases [2,3]. For a detailed investigation of DYRK1A’s physiological role in various test conditions and as starting points for a rational drug design, small organic DYRK1A inhibitors would be rather helpful. Although up to now more than a dozen of such DYRK1Ainhibitory chemotypes have been published, none has yet exhibited clear selectivity versus protein kinases of the structurally related cdc-2 like kinase (CLK) family [2,3]. We here report a structure-guided DYRK1A inhibitor development campaign during which the screening hit 11H-indolo[3,2-c]quinoline-6-carboxylic acid was modified. X-ray structure determination of three congeners co-crystallized with DYRK1A confirmed the predicted binding mode within the ATP binding site in the target kinase and led to the design of to KuFal194, a DYRK1A inhibitor (IC50 = 6 nM) with cellular activity and a two orders of magnitude selectivity over CLK1 [4]. Ideas, services, results and conclusions were contributed to this collaborative project by a private biotechnology company (ManRos), a registered charity (SGC), a public research organization (CNRS) and three universities (University of Oxford, RWTH Aachen and TU Braunschweig). Acknowledgments: This research was supported by grants from the ‘Fonds Unique Interministériel” (FUI) TRIAD project (LM), the “Association France-Alzheimer (Finistère)” (LM), the “Fondation Jérôme Lejeune” (LM), the German research foundation (DFG grant Be 1967/3-1, to WB), and an FP7-KBBE-2012 grant (BlueGenics) (LM). SK receives funding from the SGC, a registered charity (number 1097737) that receives funds from AbbVie, Bayer Pharma AG, Boehringer Ingelheim, the Canada Foundation for Innovation, Genome Canada, GlaxoSmithKline, Janssen, Lilly Canada, the Novartis Research Foundation, the Ontario Ministry of Economic Development and Innovation, Pfizer, Takeda, and the Wellcome Trust [092809/Z/10/Z]. AC is supported by the European Union FP7 Grant No. 278568 “PRIMES” (Protein interaction machines in oncogenic EGF receptor signalling). We thank Sophie Kaspar for performing cell-based assays and Dr. Matthias Engel (Saarland University, Saarbrücken, Germany) for providing the HEK293-tau-DYRK1A cell line. References: 1. Soundararajan, M. et al.: Structure 2013, 21, 986–996. 2. Becker, W.; Soppa, U.; Tejedor, F. J.: CNS Neurol. Disord. – Drug Targets 2014, 13, 26-33. 3. Abbassi, R. et al.: Pharmacol. Ther. 2015, 151, 87-98. 4. Falke, H. et al.: J. Med. Chem. 2015, 58, 3131-3143. DPhG Annual Meeting 2015 Conference Book • 61 SCIENTIFIC LECTURES SL.43 The role of adenosine in colonic inflammation – A study in rat colon preparations in vitro Voß, U.1; Müller, C. E.2; Abdel-Aziz, H.3; Kelber, O.3; Nieber, K.1 1 Institute of Pharmacy, University of Leipzig, Brüderstr. 34, 04103 Leipzig, Germany of Pharmacy, Rheinische Friedrich-Wilhems Universität Bonn, An der Immenburg 4, 53121 Bonn, Germany 3 Scientific Department, Steigerwald Arzneimittelwerk GmbH, Havelstr. 5, 64295 Darmstadt, Germany 2 Institute Adenosine is a purine nucleoside involved in several physiological functions, regulating a wide variety of immune and inflammatory responses and acting as modulator of gut functions. While it is usually present at low concentrations in the extracellular space, stressful conditions, such as inflammation, can markedly increase its extracellular level up to micromolar range. Recent evidence suggests a prominent role of A2A receptors (A2AR) in the anti-inflammatory effect of herbal preparations as e.g. an Iberis amara extract (STW 6). Also A2B receptors (A2BR) have been shown to be involved in the pathophysiology of inflammation. In the current study we investigated the role of A2AR and A2BR to regulate contractility in untreated and inflamed rat colon preparations using specific receptor agonists and antagonists. Inflammation was induced by intraluminal instillation of 2,4,6-trinitrobenzenesulfonic acid (TNBS, 0.01/0.1M, 30min). mRNA-expression was determined using RT-PCR. Contractions were measured isometrically in an organ bath setup. All four adenosine receptor subtypes were expressed in untreated colon preparations. Activation of A1, A2B, and A3 receptor with specific agonists reduced the acetylcholine (ACh, 10µM)-induced contractions, while activation of A2BR enhanced it. After incubation with TNBS morphological damages in colonic mucosa and muscle walls were detectable followed by reduced ACh-contractions. The TNBS-mediated decrease of ACh-contractions as well as the morphological damages were partially normalized by co-incubation of TNBS with the A2AR agonist 2-p-[carboxyethyl]phenethylamino-5’-N-ethylcarboxamido-adenosine (CGS 21680, 10µM) or the A2BR antagonist 4-(2,3,6,7-tetrahydro-2,6-dioxo-1-propyl-1H-purin-8-yl-benzenesulfonic acid (PSB 1115, 100µM). In this study using an in-vitro inflammatory model, we demonstrate that the A2AR agonist CGS 21680 or the A2BR antagonist PSB 1115 effectively counteracted the development of TNBS-induced disturbances in colon preparations. This model therefore opens new options for uncovering anti-inflammatory mechanisms of action of herbal medicinal products. 62 • DPhG Annual Meeting 2015 Conference Book SCREENING TECHNIQUES IN PHARMACOLOGY & DRUG DEVELOPMENT 2.11 Screening Techniques in Pharmacology & Drug Development Chairs: S. Knapp, F. Bracher SL.44 Data quality in drug discovery – The role of analytical performance in ligand binding assays Wätzig, H.1; Oltmann-Norden, I.1; Steinicke, F.1; Alhazmi, H. A.1; Nachbar, M.1; Baumann, K.1; Exner, T.2; Böckler, F. M.2; El Deeb, S.1 1 Institute of Medicinal and Pharmaceutical Chemistry, TU Braunschweig, 38106 Braunschweig, Germany. Institute, University of Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, Germany 2 Pharmaceutical Despite its importance and all the considerable efforts made, the progress in drug discovery is limited. One main reason for this is the partly questionable data quality. Models relating biological activity and structures and in-silico predictions rely on precisely and accurately measured binding data. However, these data vary so strongly, such that only variations by orders of magnitude are considered as unreliable. This can certainly be improved considering the high analytical performance in pharmaceutical quality control. Thus the principles, properties and performances of biochemical and cell-based assays are revisited and evaluated. In the part of biochemical assays immunoassays, fluorescence assays, Surface Plasmon Resonance (SPR), Isothermal Calorimetry (ITC), Nuclear Magnetic Resonance (NMR) and Affinity Capillary Electrophoresis (ACE) are discussed in details, in addition radiation-based Ligand Binding Assays (LBA), Affinity Chromatography, Mass Spectrometry (MS), Atomic Force Microscopy (AFM) and Microscale Thermophoresis (MST) are briefly evaluated. In addition, general sources of error, such as solvent, dilution, sample pretreatment and the quality of reagents and reference materials are discussed. Biochemical assays can be optimized to provide good accuracy and precision (e.g. RSD% < 10%). Cell-based assays are often considered superior related to the biological significance, however, typically they cannot still be considered as really quantitative, in particular when results are compared over longer periods of time or between laboratories. A very careful choice of assays is therefore recommended. Strategies to further optimize assays are outlined, considering the evaluation and the decrease of the relevant error sources. After avoiding the common error sources and characterizing assay performances by control charts, a simple as possible representative method for the respective technique needs to be identified and optimized by an experimental design. After reducing the overall variability, other hidden error sources can be identified more easily. Furthermore, the degree of method complexity can be subsequently increased, to study the influence of additional parameters. Analytical performance and data quality are still advancing and will further advance the progress in drug development. References: 1. Wätzig, H. et al.: J. Comput. Aided Mol. Des. 2015, DOI: 10.1007/s10822-015-9851-6. DPhG Annual Meeting 2015 Conference Book • 63 SCIENTIFIC LECTURES SL.45 Dynamic mass redistribution to probe the signaling repertoire of GPCRs Schrage, R.1; Mohr, K.1 1 Pharmacology and Toxicology Section, Institute of Pharmacy, University of Bonn, Germany G protein-coupled receptors are cell surface receptors which are essential for transmitting extracellular signals across cell membranes and represent one of the largest and most diverse protein families in the human genome [1,2]. In humans, GPCRs respond to a plethora of different stimuli including hormones, neurotransmitters, and other sensory inputs like vision, olfaction, and taste [e.g. 3]. More than one third of all marketed drugs target GPCRs [4] and a lot of effort is still being put into GPCR drug discovery as an emerging number of “undrugged” receptors display association with various diseases [e.g. 5]. The receptor conformation of a GPCR is sensitive to the shape and physicochemical properties of a bound ligand as well as to the type of intracellular adaptor protein [6]. As GPCRs exist in ensembles of possible conformations, different ligands may stabilize distinct subsets of these conformations [7]. Therefore, the cellular response may be of unique character in case a specific ligand favors certain signaling pathways over others, a phenomenon termed “biased signaling” or “functional selectivity”. Unlike classic pharmacological “snapshot assays” that quantify a particular intracellular pathway such as the generation of second messengers, optical biosensors have the advantage to capture whole cell responses in intact cells [8]. In case of GPCR drug discovery, the application of specific toxins like pertussis toxin, which specifically interferes with Gi protein-mediated pathways, allows unraveling which particular pathways contribute to a whole cell response induced by an agonist. Here, we present the muscarinic M2 receptor as a paradigm for a GPCR that promiscuously activates Gi- and Gs-dependent signaling pathways, both of which can be quantified in dynamic mass redistribution (DMR) experiments applying the EPICTM system [9]. Relative to the endogenous messenger acetylcholine, the oxotremorine M-related compound iperoxo was identified as a non-biased agonist with outstanding potency and efficacy in DMR experiments [9]. The physicochemical properties and the high affinity of iperoxo made it the compound of choice to generate a novel muscarinic radioagonist, yielding [3H]iperoxo. [3H]iperoxo was the first radioagonist to probe all five muscarinic receptors and allowed detection of activationrelated conformational changes in the receptor protein [10]. Taken together, we present DMR experiments as extremely useful in GPCR drug discovery, especially for GPCRs that promiscuously interact with a variety of intracellular adaptor proteins. Acknowledgments: We are grateful for the excellent collaboration with Prof. Dr. Evi Kostenis (University of Bonn, Germany), Prof. Dr. Marco De Amici (Università degli Studi di Milano, Italy), and Prof. Dr. Ulrike Holzgrabe (University of Würzburg, Germany). References: 1. Millar, R. P.; Newton, C. L.: Mol. Endocrinol. 2010, 24(1): 261-74. 2. Oldham, W. M.; Hamm, H. E.: Nat. Rev. Mol. Cell. Biol. 2008, 9(1): 60-71. 3. Rosenbaum, D. M.; Rasmussen, S. G.; Kobilka, B. K.: Nature. 2009, 459(7245): 356-63. 4. Overington, J. P.; Al-Lazikani, B.; Hopkins, A. L.: Nat. Rev. Drug Discov. 2006, 5(12): 993-6. 5. Garland, S. L.: J. Biomol. Screen. 2013, 18(9): 947-66. 6. Kobilka, B. K.; Deupi, X.: Trends Pharmacol. Sci. 2007, 28(8): 397-406. 7. Nygaard, R. et al.: Cells 2013, 152(3): 532-42. 8. Schröder, R. et al.: Nat. Biotechnol. 2010, 28(9): 943-9. 9. Schrage, R. et al.: Br. J. Pharmacol. 2013, 169(2): 357-70. 10. Schrage, R. et al.: Biochem. Pharmacol. 2014, 90(3): 307-19. 64 • DPhG Annual Meeting 2015 Conference Book SCREENING TECHNIQUES IN PHARMACOLOGY & DRUG DEVELOPMENT SL.46 Label-free biosensors help to reveal a new mechanism of GPCR activation Grundmann, M.1; Hudson, B. D.2; Tikhonova, I. G.3; Smith, N. J.4; Mohr, K.5; Ulven, T.6; Milligan, G.2; Kenakin, T.7; Kostenis, E.1 1 Molecular-, Cellular- and Pharmacobiology Section, Institute of Pharmaceutical Biology, University of Bonn, Nussallee 6, 53115 Bonn, Germany 2 Molecular Pharmacology Group, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, Scotland, UK 3 Molecular Therapeutics, School of Pharmacy, Medical Biology Centre, Queen's University, Belfast, Northern Ireland, UK 4 Molecular Cardiology Division, Victor Chang Cardiac Research Institute, Faculty of Medicine, University of New South Wales, Sydney, Australia 5 Pharmacology and Toxicology, University of Bonn, Bonn, Germany 6 Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Odense M, Denmark 7 Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, NC, USA G protein-coupled receptors (GPCRs) are long runners among drug targets and have an outstanding history in pharmacology. In recent years, high attrition rates in the drug discovery process tarnish the picture of success. Insufficient preclinical compound evaluation and the development of drug molecules lacking an innovative modeof-action were identified as key issues during this process. The implementation of new holistic cellular readouts into early phases of drug research raises hopes to overcome the issues and to reinitiate the development of effective and successful drugs. Label-free assays can provide an important means to achieve a comprehensive compound profile already in early drug discovery stages including the identification of the mode-of-action. GPCRs are highly versatile and regulate complex cell signaling. Since a plethora of signal transduction concepts are mirrored herein, GPCR biology is a treasure trove for ligands with new mechanism of action. To date, ligands that target GPCRs can be classified either as orthosteric, allosteric or bitopic. In this talk, I will present data that underpin the capability of cell-based biosensor assays to disclose a novel molecular mechanism of drug-like compound action. Embedded into an array of classical GPCR signaling readouts and by using pharmacological and mutational approaches, we made use of the possibility to track cell activation in real time to discover a new form of bitopic agonism to spatio-temporally modulate a class A GPCR. Acknowledgments: We are grateful to Ulrike Rick for expert technical assistance. We thank Sunil Pandey for the synthesis of CATPB. This work was supported by the Danish Council for Strategic Research (grant 11-11619). The authors are grateful to Corning Inc., Perkin Elmer, and Molecular Devices for their support on the dynamic mass redistribution as well as cellular dielectric spectroscopy biosensors. DPhG Annual Meeting 2015 Conference Book • 65 SCIENTIFIC LECTURES SL.47 Assay technologies addressing GPCRs in drug discovery Koch, M.1 1 Bayer Pharma AG, Lead Discovery Wuppertal, Aprather Weg 18a, 42096 Wuppertal Approximately 40% of all approved drugs target G protein-coupled receptors (GPCRs). This large family of transmembrane receptors transduces extracellular signals into intracellular effector pathways through the activation of heterotrimeric G proteins. Therefore, they constitute a prominent class of validated pharmacological targets with an obvious high druggability. Consequently, extensive efforts are under way to explore and validate further members of the GPCR family as promising drug targets for various therapeutic areas and to identify novel lead compounds [1,2]. High throughput screening (HTS) is a well-established technology to identify novel starting points for lead generation out of a large collection of several millions of small molecules. At Bayer Pharma AG we have established a unique HTS technology platform capable of testing up to 300,000 compounds per day. Furthermore, we select a suitable set of HTS-compatible functional assays from an assay toolbox fitting to the specific objectives of the particular target and disease hypothesis. The identification of compounds with a reasonable profile for further development is a rare event. Most probably, primary HTS hits are potential artefacts. Therefore, compound characterization with respect to specificity and selectivity is an essential component in the HTS workflow. Orthogonal readout systems are applied to exclude unspecific target interactions or assay readout artefacts. Additionally, selectivity tests against closely related receptors, isoforms and orthologs provide further crucial information to aid the selection of the most promising lead candidates. In the post HTS phase, competitive ligand binding assays provide supplementary evidence on the molecular mode of action, e.g. for differentiation between orthosteric and allosteric modulation of receptor activity [3]. Furthermore, radiolabeling of selected candidates allows a detailed investigation of ligand binding kinetics, especially of residence time which is a key parameter for receptor occupancy. The presentation will exemplify how different assay technologies addressing GPCRs are applied to support lead discovery and lead optimization towards new molecular entities. References: 1. Pierce, K. L. et al.: Nature Rev. Mol. Cell Biol. 2002, 3(9): 639-50. 2. Eglen, R. M.; Raisine T.: Methods Mol. Biol. 2009, 552: 1-13. 3. Tschammer N.: Top. Med. Chem. 2015, 14: 87-118. 66 • DPhG Annual Meeting 2015 Conference Book SCREENING TECHNIQUES IN PHARMACOLOGY & DRUG DEVELOPMENT SL.48 Semi-automatic fluorescence anisotropy titrations (saFLAT) enhance screening throughput and data quality Boeckler, F. M.1; Lange, A.1; Heidrich, J.1; Ansideri, F.1.; Koch, P.1 Laboratory for Molecular Design and Pharmaceutical Biophysics, Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmaceutical Sciences, Eberhard Karls Universität Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, Germany. 1 Reliable detection of binding constants and improved screening throughput are typically opposing aims in drug discovery. Still, both are of utmost importance [1] for the hit and lead identification and optimization stages in academic and industrial labs. Of course, automatization and robotics have strongly contributed to speeding up data generation and to enhancing data quality. However, such solutions typically require considerable investments, constituting an obstacle particularly for academic research. Smart solutions for redesigning the experimental setup and semi-automatic liquid handling systems can help to accelerate research and enhance data quality at a bargain price. Based on similar approaches for the measurement of protein-DNA binding [2], we have designed a fluorescence anisotropy assay which is performed as a titration in microtitre plates [3]. Fluorescein-labelled probe molecules (as reporters) are displaced from the target binding site by increasing concentrations of competitor ligands. Changes in fluorescence anisotropy measured using a CLARIOstar plate reader [4] reveal changes in the proportion of bound to free fluorescein-labelled probe. Using a VIAFLO 96-channel pipette [5] in a semi-automatic mode, one titration step can be performed in seconds. The sample volume during the titration is kept constant by aspirating the same volume of the sample prior to addition of an aliquot of the competitor compound from a source plate. In order to keep the concentration of the fluoresceinlabelled probe and the protein constant, stock solutions of the competitor compounds are mixed with the same concentration of reporter and protein as in the initial sample. By this process, only the concentration of the competitor compound changes during the titration. To minimize the errors associated with handling small volumes and still perform a titration covering several pKD units, different stock solutions of the competitor compounds (in multiple source plates) can be used subsequently for the titration. Based on this experimental setup, the KD value of 96 compounds can be determined simultaneously in one single screening experiment. The number of titration steps and the possible pKD range is predominantly limited by the solubility of the competitor compounds. Alternatively, the possibility to determine many KD values in parallel can be used as well to increase the number of replications of the experiment and, therefore, the data reliability. References: 1. Wätzig, H. et al.: J. Comput. Aided Mol. Des. 2015, in press, DOI: 10.1007/s10822-015-9851-6 2. Veprintsev, D. B.; Fersht, A. R.: C. Nucleic Acids Res. 2008, 36(5): 1589-1598. 3. Vogel, S. et al.: Proc. Natl. Acad. Sci. U. S. A. 2012, 109(42): 16906-16910. 4. BMG Labtech CLARIOstar, http://www.bmglabtech.com/en/products/clariostar/ 5. INTEGRA Biosciences VIAFLO 96, http://www.integra-biosciences.com/sites/96_384_channel_pipette.html DPhG Annual Meeting 2015 Conference Book • 67 SCIENTIFIC LECTURES 2.12 Signaling in Cell Death Chairs: S. Knapp, F. Bracher SL.49 Sensitization of the anti-cancer efficacy of the Bcl-2 family inhibitor ABT-263 by natural compounds Polier, G.1; Giaisi, M.1; Köhler, R.1; Müller, W. W.1; Krammer, H. P.1; Li-Weber, M.1 1 Tumorimmunology Program (D030), German Cancer Research Center (DKFZ), D-69120 Heidelberg, Germany Tumor initiation, progression and resistance to therapies are tightly associated with over-expression of antiapoptotic proteins Bcl-2, Bcl-xL, Bcl-w and Mcl-1. ABT-263 (Navitoclax), an orally bio-available small-molecule mimetic of the Bcl-2 homology domain 3, inhibits Bcl-2, Bcl-xL, and Bcl-w and has shown anti-cancer effects mainly on lymphomas and lymphocytic leukemia. Despite promising results obtained from the clinical trials, the use of ABT-263 in patients is dose-limited due to induction of death of thrombocytes via inhibition of Bcl-xL and subsequent thrombocytopenia. Besides, many tumors resist treatment due to high levels of Mcl-1 expression or develop resistance via up-regulation of Mcl-1 during long-term exposure. These obstacles highlight the demand to improve the ABT-263-based therapy. Recently, we show that agents that inhibit the translation initiation or transcription elongation, such as the natural compounds Rocaglamides and flavones, e.g. wogonin, baicalein, apigenin, chrysin and luteolin, can enhance ABT-263-induced apoptosis and thereby decrease its effective dose in different cancer cell lines and in primary AML and ALL cells by down-regulation of Mcl-1 expression. Importantly, these natural compounds do not enhance the toxicity of ABT-263 to proliferating normal T cells and thrombocytes. These compounds also potentiate the lethality of ABT-263 in cancer cells which have acquired resistance to ABT-263. Finely, we show that combination of wogonin with ABT-263 promotes in vivo tumor regression in a human T-cell leukemia xenograft mouse model. Our study demonstrates that the combination therapy can lower the effective dose of ABT-263 and thereby possibly decreasing the risk of adverse side effects. Acknowledgments: This work was supported by the Helmholtz Alliance on Immunotherapy of Cancer in the Helmholtz association and Deutsches Konsortium für Translationale Krebsforschung (DKTK). References: 1. Ebada, S. S. et al.: Prog. Chem. Org. Nat. Prod. 2011, 94: 1-58. 2. Polier, G. et al.: Cell Death Dis. 2011, 2:e182. 3. Li-Weber, M.: Int. J. Cancer. 2015, 137(8):1791-9. 4. Polier, G. et al.: Int. J. Cancer. 2015, 136(3):688-98. 68 • DPhG Annual Meeting 2015 Conference Book SINGALING IN CELL DEATH SL.50 miR181b is induced by the chemopreventive polyphenol curcumin and inhibits breast cancer metastasis via downregulation of the inflammatory cytokine CXCL1 and 2 Bachmeier, B. E.1; Kronski, E.1; Fiori, M. E.2; Barbieri, O.3,4; Astigiano, S.3; Mirisola, V.5; Killian, P. H.1; Bruno, A.6; Pagani, A.6; Rovera, F.7; Pfeffer, U.8; Sommerhoff, C. P.1; Noonan, D. M.6,7; Nerlich, A. G.9 1 Institute of Laboratory Medicine, Ludwig-Maximilians-University, Munich, Germany of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy 3 Department of Experimental Medicine, University of Genoa, Genoa, Italy 4 Embryogenesis and Tumorigenesis in Animal Models, IRCCS AOU San Martino-IST National Cancer Research Institute, Genoa, Italy 5 Integrated Molecular Pathology, IRCCS AOU San Martino-IST National Cancer Research Institute, Genoa, Italy 6 Scientific and Technologic Pole, Fondazione Onlus MultiMedica, Milan, Italy 7 Department of Biotechnologies and Life Sciences, University of Insubria, Varese, Italy 8 Department of Surgical and Morphological Sciences, University of Insubria, Varese, Italy 9 Institute of Pathology, Academic Hospital Munich-Bogenhausen, Munich, Germany 2 Department Chronic inflammation is a major risk factor for the development and metastatic progression of cancer. We have previously reported that the chemopreventive polyphenol Curcumin inhibits the expression of the proinflammatory cytokines CXCL1 and -2 leading to diminished formation of breast and prostate cancer metastases. In the present study, we have analyzed the effects of Curcumin on miRNA expression and its correlation to the anti-tumorigenic properties of this natural occurring polyphenol. Using microarray miRNA expression analyses, we show here that Curcumin modulates the expression of a series of miRNAs, including miR181b, in metastatic breast cancer cells. Interestingly, we found that miR181b down-modulates CXCL1 and -2 through a direct binding to their 3’-UTR. Overexpression or inhibition of miR181b in metastatic breast cancer cells has a significant impact on CXCL1 and 2 and is required for the effect of Curcumin on these two cytokines. miR181b also mediates the effects of Curcumin on inhibition of proliferation and invasion as well as induction of apoptosis. Importantly, overexpression of miR181b in metastatic breast cancer cells inhibits metastasis formation in vivo in immunodeficient mice. Finally, we demonstrated that Curcumin up-regulates miR181b and down-regulates CXCL1 and -2 in cells isolated from several primary human breast cancers. DPhG Annual Meeting 2015 Conference Book • 69 SCIENTIFIC LECTURES SL.51 Shikonin and its derivatives inhibit phosphorylation of the epidermal growth factor receptor signaling and synergistically kill glioblastoma cells in combination with erlotinib Zhao, Q.1; Efferth, T.1 1 Department of Pharmaceutical Biology, Institute of Pharmacy and Biochemistry, Johannes Gutenberg University, Staudinger Weg 5, Mainz, 55128, Germany Overexpression and mutation of the epidermal growth factor receptor (EGFR) gene play a causal role in tumorigenesis and resistance to treatment of glioblastoma (GBM) [1]. EGFR inhibitors such as erlotinib are currently used for the treatment of GBM; however, their efficacy has been limited due to drug resistance [2]. New treatment strategies are therefore urgently needed. Shikonin, a natural naphthoquinone, induces both apoptosis and necroptosis in human glioma cells [3,4], but the effectiveness of erlotinib-shikonin combination treatment as well as the underlying molecular mechanisms is unknown yet. In this study, we investigated erlotinib in combination with shikonin and 14 shikonin derivatives in parental U87MG and transfected U87MG.∆EGFR GBM cells. Most of the shikonin derivatives revealed strong cytotoxicity. Shikonin together with five other derivatives, namely deoxyshikonin, isobutyrylshikonin, acetylshikonin, β,β-dimethylacrylshikonin and acetylalkannin showed synergistic cytotoxicity toward U87MG.∆EGFR in combination with erlotinib. Moreover, the combined cytotoxic effect of shikonin and erlotinib was further confirmed with another three EGFR-expressing cell lines, BS153, A431 and DKMG. Shikonin not only dose-dependently inhibited EGFR phosphorylation and decreased phosphorylation of EGFR downstream molecules, including AKT, P44/42MAPK and PLCγ1, but also together with erlotinib synergistically inhibited ∆EGFR phosphorylation in U87MG.∆EGFR cells as determined by Loewe additivity and Bliss independence drug interaction models. These results suggest that the combination of erlotinib with shikonin or its derivatives might be a potential strategy to overcome drug resistance to erlotinib. Acknowledgments: Department of Pharmacognosy, Institute of Pharmaceutical Sciences, University of Graz, Graz; Austria Faculty of Pharmacy, University of Athens, Athens, Greece; Departament de Farmacologia, Facultat de Farmacia, Universitat de Vale`ncia,Valencia, Spain; Prof. Rudolf Bauer, Prof. Ioanna Chinou, Prof. José-Luis Rios. References: 1. Furnari, F.B. et al.: Genes & development 2007, 21: 2683-710. 2. Zhu, J. J.; Wong, E. T.: Current molecular medicine 2013, 13: 358-67. 3. Chen, C. H. et al.: Ann. Surg. Oncol. 2012, 19: 3097–106. 4. Huang, C. et al.: Plos One 2013, 8(6): e66326. 70 • DPhG Annual Meeting 2015 Conference Book SINGALING IN CELL DEATH SL.52 A novel autodisplay based screening assay for the identification of small molecules that inhibit the dimerization of human chaperone Hsp90 Bopp, B.1: Ciglia, E.2; Ouald-Chaib, A.2; Groth, G.3; Gohlke, H.2; Jose, J.1 1 Institute of Pharmaceutical and Medicinal Chemistry, Westfälische Wilhelms-Universität. PharmaCampus. Corrensstr. 48, 48149 Münster, Germany 2 Institute of Pharmaceutical and Medicinal Chemistry, Heinrich-Heine-University Düsseldorf, Universitätsstr. 1, 40225 Düsseldorf, Germany 3 Institute for Biochemical Plant Physiology, Heinrich-Heine-University Düsseldorf, Universitätsstr. 1, 40225 Düsseldorf, Germany Human Hsp90 (Hsp90) is a homodimeric chaperone, essential for the maturation of numerous proteins. Some of these proteins are involved in tumor formation and growth, which makes Hsp90 an interesting drug target for cancer treatment [1]. Here, we describe a novel autodisplay based method to screen for small molecules that inhibit PPI. The Autodisplay technology was used to express the chaperone Hsp90 on the surface of Escherichia coli. Functional folding and dimerization was confirmed by binding of FITC-labeled p53, a natural client protein of Hsp90 and subsequent analysis by flow cytometry. By computational analysis hot spots in the C-terminal domain of Hsp90 important for dimerization were identified [2]. These hot spot predictions were used to design peptides that were supposed to inhibit Hsp90 dimerization. Because binding of FITC-labeled p53 is dependent on Hsp90 dimer formation, a loss of fluorescence in the flow cytometer analysis indicates inhibition. This reduction in fluorescence turned out to be dose-dependent with respect to the inhibitor concentration, and an IC50 of 8.96 µM could be determined for the best inhibitor H3. By microscale thermophoresis measurement with the purified C-terminal domain of Hsp90 it was verified that H3 indeed binds the C-terminal domain of Hsp90 with a KD value of 2.19 µM. Up to now, H3 is the first inhibitor shown to target the C-terminal dimerization domain of Hsp90 with a KD value in the low micromolar range [3]. References: 1. Wegele, H.; Müller, L.; Buchner, J.: Rev. Physiol. Biochem. Pharmacol. 2004, 151: 1-44. 2. Ciglia, E. et al.: PLoS One 2014, 9: e96031. 3. Bopp, B. et al.: 2015, submitted DPhG Annual Meeting 2015 Conference Book • 71 SCIENTIFIC LECTURES 2.13 Anticancer and Epigenetic Drugs Chairs: M. Kassack, M. Jung SL.53 Targeted cancer therapies Rauh, D.1 1 Technische Universität Dortmund, Fakultät für Chemie und Chemische Biologie, Otto-Hahn-Strasse 6, 44227 Dortmund (Germany) The goal of my lab is to develop, synthesize and facilitate desperately needed tools to study target protein function in cells and organisms to push research to exciting new frontiers. In our research, we employ organic synthesis, biochemical and cellular compound screening, protein X-ray crystallography, structure-based design as well as target identification for the development of inhibitors and functional probes to perturb proteins of interest. A strong focus in the lab is on personalized medicine for the treatment of cancer. Here, we closely collaborate with clinical oncologists to better understand the mechanisms of drug resistance and to develop compounds to overcome acquired drug resistance in non-small-cell lung cancer and gastrointestinal stromal tumors. We are motivated that our techniques and investigations will lead to a better understanding of the molecular and cellular causes of fatal diseases and stimulate the development of new drugs. Some of the labs’ research initiatives on targeted cancer therapies will be outlined [1-5]. References: 1. Engel, J. et al.: J. Med. Chem. 2015, in press. 2. Richters, A. et al.: ACS Chem. Biol. 2015,10, 289-298. 4. Fang, Z. et al.: ACS Chem. Biol. 2015, 10, 279-288. 4. Mayer-Wrangowski, S. C. et al.: Angew. Chem. Int. Ed. Engl. 2015, 54, 4379-4382. 5. Weisner, J. et al.: Angew. Chem. Int. Ed. Engl. 2015, 54, 10313-10316. 72 • DPhG Annual Meeting 2015 Conference Book ANTICANCER AND EPIGENETIC DRUGS SL.54 Role of apoptosis signaling in tumorigenesis and therapy resistance Wesselborg, S.1 1 Institute for Molecular Medicine I, University of Düsseldorf, Universitätsstr. 1, 40225 Duesseldorf, Germany Activation of the endogenous suicide program (apoptosis) is one of the major mechanisms by that cancer cells are eliminated by antitumor agents. Though the primary site of action might differ, the basic outcome of most anticancer drugs is usually the same – induction of the apoptotic program in the cancer cell. Conversely, defects within the apoptotic signaling machinery convey resistance of tumors to radio- and chemotherapy. Understanding of the molecular events that regulate the apoptotic machinery induced by anticancer drugs and the measures that the tumor cell takes to avoid apoptosis might help to develop new therapeutic strategies and drug development. Theoretically, a cancer cell is nothing but a cell propagating its anarchistic death defying principle of unlimited self dissemination that – if cellular safeguard mechanisms or the immune system fail to keep under control – is finally terminated by the demise of the whole organism itself. The basic objective of chemotherapy is therefore to exploit the different activities that distinguish malignant tumor cells from non-malignant cells in order to specifically eliminate the 'anarchistic dropouts'. An ideal anticancer drug would accompolish to specifically target the malignant cancer cell and leave the rest of the non-malignant cells in our body unaffected. Due to detrimental side effects on non-malignant tissues and the virtue of some tumors not to respond to the respective chemotherapy or to develop resistance during chemotherapy this ideal concept is unfortunately rarely achieved. However, research of the last three decades improved our knowledge of tumor biology and enabled the identification of new targets for therapy. Thus, in addition to uncontrolled cell proliferation, it is well established now that cancer cells also need to induce their supply with oxygen and nutrients by the induction of angiogenesis of surrounding blood vessels, extravasation and metastasis by releasing matrix metalloproteases for extracellular matrix degradation and so on. Whereas, the 1980's were dominated by research on the mechanisms of uncontrolled proliferation the 1990's also brought another feature of cancer cells into focus – that is the counterpart of mitosis i.e. apoptosis. Thus, it turned out that tumor cells not only propagate their anarchistic desire for unlimited proliferation but they also inactivate external and internal triggers of the cellular suicide program. Anticancer drugs are used as therapeutic agents since 1940 with more or less success. Per definition a chemotherapeutic agent is any drug that contributes to tumor destruction. For decades the major feature of chemotherapeutic agents had been the infliction of DNA-damage. This kind of therapy basically targets cells with a high proliferation rate that cannot evade the consequences of DNA-damage by cell cycle arrest or DNA-repair. Consequently, other benign but highly proliferating tissues such as bone marrow, mucosal membranes, hair follicles and cells of the gastrointestinal are also affected. Recently, due to the rapidly expanding knowledge of the signal transduction pathways in tumor biology, a new generation of anticancer drugs evolved in form of ‘targeted therapy’ that complements conventional DNA-damaging radio- and chemotherapy. This new generation of antitumor agents utilizes genuine features of cancer cells as targets for anti-tumor therapy. Hence, these new approaches target receptor tyrosine kinases for growth factors (such as EGF, PDGF, TGF-α), inhibit intracellular protein kinases (such as PKC, PKA, RAF), the proteasome or histone deacetylases (HDACs), and so forth. DPhG Annual Meeting 2015 Conference Book • 73 SCIENTIFIC LECTURES SL.55 A novel CREBBP/p300 inhibitor and its molecular effects in cancer cells Lucas, X.1; Hügle, M.2; Ostrovsky, D.3; Decker, S.4, Gerhardt, S.2; Einsle, O.2; Dierks, C.4; Breit, B.3; Wohlwend, D.2; Günter, S.1 Institute of Pharmaceutical Sciences, Albert-Ludwigs-University, Hermann-Herder-Str. 9, 79104 Freiburg, Germany of Biochemistry, Albert-Ludwigs-University, Albertstr. 21, 79104 Freiburg, Germany 3 Institute of Organic Chemistry, Albert-Ludwigs-University, Albertstr. 21, 79104 Freiburg, Germany 4 Department of Hematology/Oncology, University Medical Centre Freiburg, Hugstetter Str. 55, 79106 Freiburg, Germany 1 2 Institute Proteins that are involved in the regulation of chromatin and transcriptional control represent an important targetclass and provide new opportunities for the development of small molecules as anti-cancer agents. Histone modification proteins are classified as 'writers', 'erasers' and 'readers'. The last comprise, among others, the wellstudied bromodomain (BRD) family, which specifically recognises acetylated lysine residues. Within the 61 different BRDs especially the eight members of BET-subfamily have been subjected to several drug discovery campaigns, and some of the resulting drugs are now in clinical trials for atherosclerosis, acute myeloid leukemia (AML) and other hematologic malignancies. Here, we present the results of a model-based drug discovery campaign aiming for the identification of new inhibitors against CREBBP and p300. Their bromodomains are outside the BET subfamily, and their role in cancer development is not yet completely understood. An identified high-affinity fragment [1] was further optimized by a fragment-growing approach. We could show that the resulting inhibitor can successfully inhibit the proliferation of cells from different cancer types. In vitro screenings with primary blood cells from chronic lymphocytic leukemia (CLL) patients could show that the observed cytotoxic effects are specific for CLL-cells. Additionally, RNA-seq analysis could show that the activity of the inhibitor can be related to the down-regulation of important driver genes involved in CLL progression. References: 1. Lucas X, et al.: Angew Chem Int Ed Engl. 2013, 52: 14055-14059. 74 • DPhG Annual Meeting 2015 Conference Book ANTICANCER AND EPIGENETIC DRUGS SL.56 Macrocyclic inhibitors of human histone deacetylase enzymes Olsen, C. A.1 1 Center for Biopharmaceuticals and Department of Drug Design & Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100, Copenhagen, Denmark. A brief introduction to the histone deacetylase (HDAC) enzymes, or perhaps more appropriately the lysine deacylase (KDAC) enzymes [1], will be given, and the lecture will then be devoted to work on macrocyclic peptidebased inhibitors of this class of enzymes. Several cyclic tetrapeptide and depsipeptide natural products have proven useful as biological probes and drug candidates due to their potent activities as HDAC inhibitors. These chemotypes exert their activity through a dual mechanism of action involving binding to the catalytic pocket as well as the surface of the enzyme where substrate proteins dock. Here, the synthesis of such a class of cyclic tetrapeptide HDAC inhibitors, the azumamides, will be presented along with structurally modified analogues. We thus achieved total syntheses of azumamides B–D for the first time, which corroborated the originally assigned structures and enabled full profiling of the HDAC inhibitory properties of the entire selection of azumamides A–E [2]. Furthermore, an extended series of analogues containing various structural modifications was evaluated by HDAC profiling, NMR structure determination, and molecular docking to HDAC crystal structures to reveal insight into the requirements for potent HDAC inhibition by macrocyclic peptides that disrupt the interaction between HDAC enzyme and substrate protein [3,4]. References: 1. Olsen C. A. Angew. Chem. Int. Ed. 2012, 51: 3755-3756. 2. Villadsen J. S. et al.: J. Med. Chem. 2013, 56: 6512–6520. 3. Maolanon, A. R. et al.: J. Med. Chem. 2014, 57: 9644–9657. 4. Villadsen, J. S. et al.: Med. Chem. Commun. 2014, 5: 1849–1855. DPhG Annual Meeting 2015 Conference Book • 75 SCIENTIFIC LECTURES SL.57 Correlation of conformation with cytotoxic activity of α-aminoxy oligopeptides: A circular dichroism study Rüther, A.1; Diedrich, D.2; Rodrigues Moita, A. J. 2; Kurz, T.2; Kassack, M. U.2; Hansen, F. K.2; Lüdeke, S.1 1 Institute of Pharmaceutical Sciences, Albert-Ludwigs-Universität Freiburg, 79104 Freiburg, Germany of Pharmaceutical and Medicinal Chemistry, Heinrich-Heine-Universität Düsseldorf, 40225 Düsseldorf, Germany 2 Institute Therapeutic peptides may have active and inactive conformations. The equilibrium between these conformations depends both on the amino acid sequence and on external parameters, such as pH, or interaction with membranes. Therefore, in order to understand their influence a therapeutic context, it is critical to identify the conformational response of peptides or peptidomimetics on environmental stimuli [1]. As circularly polarized light interacts with helically disposed transition dipoles in the peptide backbone, circular dichroism (CD) spectroscopy is exceptionally sensitive toward conformational changes and provides spectra with characteristic patterns that are typical for secondary structure [2]. While CD is a widely used tool to study the conformation of canonical peptides, there is little CD data available for peptidomimetics, such as peptoids [3], or αaminoxy peptides [4]. In particular studies on α-aminoxy peptides are of interest. Due to their proteolytic and conformational stability they are promising drug candidates [5]. We studied spectra recorded for different cytotoxic α-aminoxy peptides obtained through combined solutionphase/solid-phase synthesis. The α-aminoxy peptides show a remarkable pH-dependent change of their secondary structure. The presence of liposomes as a model for cell membranes also induces changes in the CD spectrum. This suggests that membranolytic effects observed in preliminary mode of action studies with cancer cells are concomitant with conformational changes that occur upon binding of the α-aminoxy peptides to the cell membranes. References: 1. Sinthuvanich C. et al.: J. Am. Chem. Soc. 2012, 134: 6210–6217. 2. Toniolo, C.; Formaggio, F.; Woody, R. W.: Comprehensive Chiroptical Spectroscopy. (Wiley) 2012. 3. Laursen, J. S. et al.: Nat. Commun. 2015, 6: DOI: 10.1038/ncomms8013. 4. a) Li X.; Yang, D.: Chem. Commun. 2006: 3367–3379. b) Yang D. et al.: Chem. Asian. J. 2010, 5: 1356–1363. 5. a) Draghici, B. et al.: RSC Adv. 2011, 1: 602–606. b) Li X, Wu Y. D.; Yang, D.: Acc. Chem. Res. 2008, 41: 1428–1438. 76 • DPhG Annual Meeting 2015 Conference Book PERSONALIZED MEDICINE – BIOMARKER AND DIAGNOSTICS 2.14 Personalized Medicine – Biomarker and Diagnostics Chairs: O. Queckenberg, T. Dingermann SL.58 The challenge to leverage genetic biomarkers from research level to routine clinical IVD testing Ortmann, R.1 1 QIAGEN GmbH, Hilden, Germany During recent years companion diagnostics reached high importance for pharmaceutical development programs and guidance on clinical treatment decisions. European countries achieved a leading position through early uptake of companion diagnostics in clinical routine, with the highest patient testing coverage worldwide. Commercial CEin-vitro-diagnostic products and laboratory developed tests, mainly developed by European universities, are applied side by side. Quality assessment systems, like EQA, are in place. Still, there are some key challenges in personalized medicine, which will impact future companion diagnostic development and commercialization projects: 1. the increasing complexity of testing, e.g. biomarker panel testing and NGS technologies, 2. the higher throughput of samples, esp. in liquid biopsy based treatment monitoring, 3. the consolidation of test results to clinically actionable result reports, through bioinformatics, 4. the EU directive on higher regulatory requirements on companion diagnostics, and 5. the proof of clinical utility to payers. Previously, clinical drug development projects guided the evolution of companion diagnostics in general. Now, commercialization success of new drug-companion diagnostic co-launches increasingly impacts decisions on biomarker strategy in pharmaceutical companies. Further growth of a strong European position in personalized medicine can be achieved only through a close collaboration of pharmaceutical and diagnostic companies with clinicians and molecular diagnostic laboratories, as the maxim of decisions needs to move from early development towards routine clinical application. The consequences of this change will be addressed during the talk. DPhG Annual Meeting 2015 Conference Book • 77 SCIENTIFIC LECTURES SL.59 The potential of biomarkers to support decisions making in clinical studies in cardiovascular indications Kramer, F.1 1 Clinical Sciences – Experimental Medicine, BAYER HealthCare AG, Aprather Weg 18a, 42113 Wuppertal, Germany Cardiovascular (CV) diseases such as heart failure (HF) are complex syndromes, which are driven by multiple pathomechanisms. Therefore, a comprehensive assessment of the status of different pathomechanisms in every individual patient is the key to a therapy serving patient`s needs. Due to the complexity of CV disease, a large group of pharmacotherapies have been established as standard of care (SoC) in CV indications. This comprises, amongst others, beta-blockers (BB), angiotensin converting enzyme inhibitors (ACEIs), angiotensin receptor blockers (ARBs), diuretics, calcium channel blockers and mineral corticoid receptor antagonists (MRAs). Use of these agents has led to a significant improvement of patient care and a decline in deaths from CV diseases [1]. As many of those therapies target the periphery rather than the heart, there is still a high medical need for therapeutic concepts which target for example inflammation processes, tissue remodelling or energy metabolism in the myocardium. Furthermore, the fact that a significant number of patients do not respond to a given therapy stresses the need for new therapies [2]. To enable successful clinical development of drug candidates in CV indications a comprehensive diagnosis, including a definition of underlying pathomechanisms in the target population, is essential. Furthermore, the ability to reliably monitor safety and to detect early signs of efficacy during a clinical study are key elements of modern drug development. Biomarkers serve as a tool for both: first to stratify patients for a clinical study investigating a certain mode of action (MoA) and second, to detect early signs of efficacy or undesired side effects. Today the term “biomarker” is used in a much broader sense and comprises more than just molecular markers to be quantified in biospecimens, such as blood, urine, saliva or tissue. A comprehensive characterization of patient`s disease status and drug candidate properties can only be achieved when information provided by molecular biomarkers is complemented by information derived from a diversity of approaches such as imaging, emerging functional measurements and medical device-derived data. Especially, the use of data derived from devices such as implanted cardiac defibrillators (ICDs), cardiac re-synchronization therapy (CRT) devices and pacemakers bear the potential to allow better surveillance of patient`s disease status upon therapy. Last but not least, data which in the past could only be generated by implanted devices can nowadays be derived from wearable patches with telemonitoring features. This is the basis for the generation of precious real life data and an increased compliance of patients in clinical studies because tele-monitoring allows a paradigm shift from an in-hospital to an at-home diagnosis and thereby reduces frequency of scheduled hospital visits in clinical studies. Taken together, the availability to have a diversity of biomarker approaches builds the basis for the design of smarter and potentially smaller clinical studies in well characterized patient populations. A comprehensive assessment of different pathomechanisms in every individual patient by using different biomarker types allows selection of the right drug for the right patient and thereby paves the way to a more personalized cardiovascular medicine. References: 1. Nabel E. G.; Braunwald E.: A Tale of Coronary Artery Disease and Myocardial Infarction. N. Engl. J. Med. 2012; 366:54-63. 2. FDA Report: Paving the Way for Personalized Medicine: FDA’s Role in a New Era of Medical Product Development. October 2013. 78 • DPhG Annual Meeting 2015 Conference Book PERSONALIZED MEDICINE – BIOMARKERS AND DIAGNOSTICS SL.60 From “omics”-technologies to stratified medicine in autoimmune diseases Lautscham, G.1; Budde, P.1; Zucht, H.1; Chamrad, D.1; Telaar, A.1; Vordenbäumen, S.2; Schulz-Knappe, P.1; Schneider, M.2 1 Protagen 2 Centre AG, Otto-Hahn-Straße 15, 44227 Dortmund, Germany of Rheumatology, Heinrich-Heine- University Düsseldorf, Düsseldorf, Germany. The inherent complexity of clinical manifestations and variety of therapeutic has made treating autoimmune diseases like lupus especially challenging. Systemic lupus erythematosus (SLE) is an autoimmune disease affecting approximately 5 million people worldwide, but early diagnosis, differentiation to other autoimmune diseases and prognostic stratification are still great challenges. Hence, SLE represents an enormous challenge for the clinical development of effective therapies and therapeutic options for patients remain limited, with the last half a century seeing only Benlysta® approved by the FDA. The degree of heterogeneity amongst SLE patients is a major obstacle for pharmaceutical development and needs to be overcome before effective and curative therapies can be identified. In order for this to happen, there is a clear need for both diagnostic biomarkers (Dx) and assays that enable precise disease characterization, patient stratification and response prediction (CDx). In SLE, several immune defects have been described, leading to the overproduction of autoantibodies against cellular and nuclear antigens and immune-mediated damage of several organs and tissues. Although, the process by which self-molecules become immunogenic is not yet fully understood, specific autoantibodies serve as important diagnostic reporter for SLE. While most of the current diagnostic autoantibody assays are based on antigens that have been identified in tissue extracts, the application of “omics” technologies allows to the address the complexity of the disease. The SeroTag® autoantibody profiling technology enables systematic screening for disease-specific changes in autoantibody reactivities using recombinant proteins produced from tissue specific expression libraries. Thus, the detection of a broad set of SLE-associated autoantibodies (AABs) might help to investigate the number, co-prevalence and similarities of AAB reactivities in SLE patients supporting a personalized disease management approach. We have recently conducted autoantibody profiling studies of SLE, systemic autoimmune diseases, and healthy controls and found known diagnostic autoantibodies and novel SLE-associated autoantibodies [1]. Confirmed antigens were employed to identify clustered autoantibodies and their clinical association in SLE and a multiplexed AAB array was developed. The discovery phase constituted the broad characterization of serum samples from SLE patients, AID patients (SSc, RA, AS) and healthy controls. against 6,912 recombinant human proteins using the bead-based Luminex technology. This work resulted in the development of NavigAID SLE, enabling stratification of SLE into distinct subgroups. The Protagen NavigAID SLE is a stratification array based on a solid database of more than 700 SLE patients, combining 87 selected known and proprietary biomarkers for the diagnosis and differential diagnosis of SLE, analysis of disease activity, detection of potential organ involvement, and activity of interferon type I response genes, as well as characterisation of four homogenous SLE patient subgroups. This array distinguishes between SLE patients ranging from a highly reactive patient group, who have a high disease activity score and possess broad and homogenous positive autoantibody reactivity, through to a smaller group of patients who have comparatively low levels of autoantibody reactivity. The co-prevalence of multiple autoantibodies in SLE patients has rarely been analysed [2,3]. However, this has now been examined in substantially greater detail and has proven to be invaluable for defining more homogeneous patient groups and solving the problem of heterogeneity in SLE. This array forms the basis for the alignment of CDx development with clinical programmes for SLE treatment, thereby increasing the probability of successful drug development. A greater emphasis on this co-development model, bringing together CDx and drug development groups, will allow for a much greater understanding of the disease in the earliest possible stages. This can result in the earlier identification of possible side effects, a shortening of overall trial lengths and numerous improvements to drug efficacy and safety. References: 1. Lueking A. et al.: Ann. Rheum. Dis. 2013;72:A535. 2. Voss A. et. al.: J. Rheumatol. 2008, 35(4), 625–630. 3. Meilof J. F. et. al.: J. Autoimmun. 1997, 10(1), 67–75. DPhG Annual Meeting 2015 Conference Book • 79 SCIENTIFIC LECTURES 2.15 Focused Pharmaceutical Research Chairs: S. Laufer, D. Steinhilber SL.61 Project group translational medicine and pharmacology, Fraunhofer IME Steinhilber, D.1,2; Parnham, M.1; Geisslinger, G.1,3 1 Fraunhofer IME-TMP, Theodor-Stern-Kai 7, 60596 Frankfurt am Main für Pharm. Chemie, Max-von-Laue-Str. 9, 60438 Frankfurt am Main 3 Institut für Klinische Pharmakologie, Klinikum der Goethe-Universität, Theodor-Stern-Kai 7, 60596 Frankfurt am Main 2 Institut Pharmaceutical research promotes the development of new drugs and enhances our understanding of how they work. However, R&D costs have increased exponentially whereas the number of new drug registrations has declined steadily over the last 10 years. One critical factor is that the identification of drug targets for inadequately understood diseases requires more extensive investment in discovery research but has a high attrition rate. This reflects the lack of validated clinical models for efficacy and safety, and intensive efforts are currently underway to develop new disease models and preclinical/clinical biomarkers, allowing R&D projects to be translated into benefits for patients. The research focus of the project group is drug research, development of predictive preclinical and clinical models of disease and clinical research. The synergy generated by housing predictive preclinical and clinical models under one roof will make it easier to take early go/no-go project decisions. We have developed validated disease models covering the fields of cardiovascular, neurodegenerative and chronic inflammatory gastrointestinal diseases, acute inflammation and pain (inflammatory, neuropathic, oncological and post-operative), arthritic and skin disorders. Based on the internal expertise in the field of pathophysiological signalling pathways, we perform research on novel innovative therapeutic approaches (systems medicine). Drawing on cutting-edge research activities and intellectual property within Goethe University Frankfurt, we apply the latest technology and research concepts to our collaborative projects, with pre-competitive research focusing on the treatment of chronic inflammatory joint disease, pain, neurodegenerative disorders and cardiovascular disease. The project group covers a portfolio of technologies for drug research and development across the value chain. 80 • DPhG Annual Meeting 2015 Conference Book FOCUSED PHARMACEUTICAL RESEARCH Neuroallianz consortium – Example of an academic-industrial collaboration SL.62 Müller, C. E.1; Pfeifer, A.2 1 PharmaCenter Bonn, 1Pharmaceutical Institute, Pharmaceutical Chemistry I, University of Bonn, An der Immenburg 4, D-53121 Bonn; of Pharmacology and Toxicology, Biomedical Center, University of Bonn, Sigmund-Freud-Str. 25, D-53105 Bonn 2 Institute The lengthy process of drug development up to market approval and the associated high risk, rising costs, increasing complexity, as well as high regulatory demands, require novel strategies. In the framework of the BioPharma competition by the BMBF (German Federal Ministery of Education and Research) strategic alliances between pharma-/biotech-companies and academic research institutions are financially supported, which cover the complete value chain from target identification and validation up to marketing of the developed drugs. Neuroallianz focuses thematically on the development of drugs for neurodegenerative diseases such as Alzheimer’s and Parkinson’s. For these indications there is a high, rapidly increasing medical demand due to the ageing population in our country. The consortium “Neuroallianz” implements a novel strategic partnership model between universities (Bonn University is the leading university partner), non-university, publicly funded research institutions (e.g. Research Center Jülich, Fraunhofer Institute St. Augustin), biotech- and pharma-companies. We have currently 20 partners, 10 from academia and 10 from industry; the leading industry partner in the consortium is UCB Pharma, Monheim. Neuroallianz is currently working on 14 projects, 6 therapeutic, and 5 diagnostic ones; moreover 2 infrastructure projects belong to the Neuroallianz portfolio: “compound library”, and “information technology”; in addition we set up a management project. Importantly, we have a strategy in place to foster public relations activities including information for the general public, as well as scientific publications and presentations. In addition, we organize advanced training programs on a regular basis for coworkers and students. Our optimized model of academic-industrial collaboration is a success story, which results in a win-win situation for all partners – for the benefit of the patients. DPhG Annual Meeting 2015 Conference Book • 81 SCIENTIFIC LECTURES The Interfaculty Centre for Pharmacogenomics and Pharma Research (ICEPHA) SL.63 Laufer, S.1; Schwab M.2 1 Institute of Pharmacy, Eberhard-Karls-University Tübingen Fischer-Bosch-Institute of Clinical Pharmacology (IKP), Stuttgart 2 Margarete Both Tübingen University and the IKP have a long history in academic drug discovery with several recent success stories in bringing ideas from bench to bedside. The ICEPHA was found in 2007 to concentrate competences and capabilities of the faculty of science, the faculty of medicine and the University Hospital Tübingen, the Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology and the Robert-Bosch-Hospital, Stuttgart. The ICEPHA established an interconnection between chemical and biological sciences and human medicine, thus forming a dynamic network in focussed research areas across academic expertise and pharmaceutical industry users. ICEPHA is a research network as well as a service and development center for innovative drugs and therapies. Such a construction allows to carry out research projects that can not be handled efficiently through one single institution. ICEPHA’s main interest is concentrated on genes which (A) affect the susceptibility of patients to drugs or (B) are associated with the manifestation of disease. Program (A) provides knowledge for defining and investigating targets for custom-tailored drugs. Program (B) is the basis for a predictable individualized therapy with patient-directed drugs and dosage. This is of prime importance for the maximum benefit for the patient’s health after therapeutic intervention, but of equal importance for the economy of the public health system. Seed and basic financing is provided from the founding organisation. Substantial funding for basic research could be raised as well from industry and Helmholtz (2 professorships each). BMBF and EU programs are sources for project funding as well. Beside the research mission, the ICEPHA developed a graduate program as well. Major aim of the ICEPHA graduate program are “membrane-associated drug targets in personalized cancer medicine” to improve future outcomes for people with cancer. In order to achieve this goal, the members of the program take advantage of the synergy achieved through their combined expertise in the basic and clinical sciences. The following key activities provide a general framework of the research in our consortium: • Validation of novel drug targets for personalized anti-tumor therapy • Identification of molecular markers as reliable predictors of treatment response • Establishment of new drugs for innovative treatment regimes • Optimization of the transfer of resources, technologies and expertise from academia to healthcare research and development of clinical trials in pharmaceutical industry 82 • DPhG Annual Meeting 2015 Conference Book FUTURE MOLECULAR DESIGN 2.16 Future Molecular Design Chairs: G. Schneider, H. Gohlke SL.64 Automated design of bispecific small molecule drugs Hopkins, A. L.1,2 1 2 Division of Biochemical Chemistry and Drug Discovvery, School of Life Science, University of Dundee, Dundee, DD1 3DF ex scientia Ltd. Dundee Incubator, James Lindsay Place, Dundee, DD1 5JJ Many effective drugs act via modulation of multiple proteins rather than single targets. Advances in systems biology are revealing a phenotypic robustness and a network structure that strongly suggests bispecific compounds targeting two nodes in a network may exhibit superior clinical efficacy. In this presentation I will discuss the development of our automated drug design platform and its successful application to the discovery and optimization of novel, bispecific small molecules that deliberately target proteins from distinct gene families. The application of the design algorithms is demonstrated by the automated invention of bispecific ligands that hits a novel combination of two enzymes from distinct gene families. Screening and X-ray crystallography experiments confirm the initial designs exhibit nanomolar potency against both primary targets, as well as high selectivity. DPhG Annual Meeting 2015 Conference Book • 83 SCIENTIFIC LECTURES SL.65 KRIPO-Protein binding site similarities for drug design Ritschel, T.1,2 1 Centre for Molecular and Biomolecular Informatics (CMBI), Radboud university medical center, 6500 HB Nijmegen, The Netherlands of Medicinal Chemistry, Faculty of Sciences, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), VU University Amsterdam, Amsterdam, The Netherlands 2 Division Key Representations of Interaction in POckets (KRIPO) is a computational method to detect similar protein binding sites independent of sequence or structural alignment [1]. The characteristics of binding pockets containing a ligand are encoded in 3D-pharmacophore fingerprints. The binding site fingerprints were optimized to improve their performance. A variety of attributes of the fingerprints were considered for the optimization, including the placement of pharmacophore features, whether or not the fingerprints are fuzzified, and the resolution and complexity of the pharmacophore fingerprints (2-, 3- and 4-point fingerprints). Finally, fuzzy 3-point pharmacophore fingerprints were chosen as an optimum representation of localized binding sites in a searchable fragment database. The KRIPO fingerprint representation is key for fast and efficient processing of PDB scale crystal structure databases. Therefore, an on-the-fly searching of PDB scale crystal structure database for application in drug design is feasible. As application examples bioisosteric replacement, off-target prediction and GPCR binding site analysis will be presented. References: 1. Wood, J. D. et al.: J. Chem. Inf. Model. 2012, 52(8):2031-43. 84 • DPhG Annual Meeting 2015 Conference Book FUTURE MOLECULAR DESIGN SL.66 Predicting the sites and products of drug metabolism Kirchmair, J.1; Glen, R. C.2; Tyzack, J. D.3 1 Center for Bioinformatics, University of Hamburg, Bundesstr. 43, 22763 Hamburg, Germany for Molecular Informatics, University of Cambridge, Lensfield Road, CB2 1EW, Cambridge, United Kingdom 3 Optibrium Ltd., 7221 Cambridge Research Park, Beach Drive, CB25 9TL, Cambridge, United Kingdom 2 Center The metabolic system has evolved as the main line of defence of living organisms against foreign, potentially hazardous substances, by transforming them into readily excretable metabolites [1]. Metabolites can have physicochemical and pharmacological properties that differ substantially from those of the parent compound [2]. Hence understanding the metabolic fate of xenobiotics is of utmost importance to the development and use of drugs, cosmetics, agrochemicals, and in fact, any chemicals exposed to biological systems [3]. Today a plethora of computational methods for predicting the sites (atom positions in a molecule where biotransformations are initiated) and products of drug metabolism are available [3,4]. In this contribution we provide an overview of the capabilities and limitations of current predictors and present two of our own methods for site of metabolism prediction: FAME (FAst MEtabolizer) [5] and an approach based on a new probabilistic machine learning method (RASCAL - Random Attribute Subsampling Classification ALgorithm) [6]. FAME is based on a set of random forest models that are derived from a set of >100k automatically annotated biotransformations. It covers phase 1 and phase 2 metabolism of xenobiotics (including drugs and natural products) in various different species and obtains top-1, top-2 and top-3 rates (i.e. percentage of molecules for which at least one known site of metabolism is predicted among the top-k highest ranked atom positions) of 71%, 81% and 87% on an external test set. The second set of models, derived using the RASCAL algorithm, is focused on cytochrome P450-mediated metabolism. While trained on a much smaller dataset, the method obtains top-1, top-2 and top-3 rates of up to 78%, 91% and 94% (depending on the isozyme). References: 1. Testa, B.: In Drug Metabolism Prediction (ed. Kirchmair, J.) (Wiley-VCH) 2014. 2. Kirchmair, J. et al.: J. Chem. Inf. Model. 2013, 53(2): 354-367. 3. Kirchmair, J. et al.: Nature Rev. Drug Discov. 2015, 14: 387-404. 4. Kirchmair, J. et al.: J. Chem. Inf. Model. 2012, 52(3): 617-648. 5. Kirchmair, J. et al.: J. Chem. Inf. Model. 2013, 53(11): 2896-2907. 6. Tyzack, J. D. et al.: J. Cheminf. 2014, 6(1): 29. DPhG Annual Meeting 2015 Conference Book • 85 SCIENTIFIC LECTURES SL.67 Enforcing drug discovery by computational molecular design Schneider, G.1 1 Department of Chemistry and Applied Biosciences, ETH Zürich Innovative bioactive agents fuel sustained drug discovery and the development of new medicines. Future success in chemical biology and pharmaceutical research will fundamentally rely on the combination of advanced synthetic and analytical technologies that are embedded in a theoretical framework that provides a rationale for the interplay between chemical structure and biological effect. A driving role in this setting falls on leading edge concepts in computer-assisted molecular design and engineering, by providing real-time access to a virtually infinite source of novel tool compounds and lead structures, and guiding experimental screening campaigns. We will present concepts and ideas for the representation of molecular structure, predictive models of structure-activity relationships, the de-orphaning of bioactive compounds, automated molecular design, and discuss de novo design approaches that have proven their usefulness and will contribute to future drug discovery by generating innovative bioactive agents. Emphasis will be put on the reaction-based construction of potent and selective enzyme inhibitors and modulators of G-protein coupled receptors. As we are currently witnessing strong renewed interest in bioactive natural products we will showcase new methods for natural-product inspired molecular design and macromolecular target prediction. References: 1. Reker, D. et al.: Nature Chem. 2014, 6: 1072–1078. 2. Reker, D. et al.: Proc. Natl. Acad. Sci. USA 2014, 111: 4067–4072. 3. Reutlinger, M. et al.: Angew. Chem. Int. Ed. 2014, 53: 4244–4248. 4. Rodrigues, T.; Schneider, P.; Schneider, G.: Angew. Chem. Int. Ed. 2014, 53: 5750–5758. 5. Spänkuch, B. et al.: Angew. Chem. Int. Ed. 2013, 52: 4676–4681. 6. Schneider, G.: Nat. Rev. Drug Discov. 2010, 9: 273–276. 86 • DPhG Annual Meeting 2015 Conference Book MEDICATION SAFETY IN SPECIAL PATIENT GROUPS 2.17 Medication Safety in Special Patient Groups Chairs: K. Friedland, U. Jaehde SL.68 Medication safety of pregnant and lactating women Scherneck, S.1; Schaefer C.2 1 Institute of Pharmacology, Toxicology and Clinical Pharmacy, Technical University of Braunschweig, Mendelssohnstr. 1, 38106 Braunschweig, Germany 2 Institute for Clinical Teratology and Drug Risk Assessment in Pregnancy, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany The use of drugs during pregnancy and lactation is a critical situation for the health care professional. A possible risk of embryonic or fetal damage caused by a drug has to be weighed against an inadequate treatment of the mother. For the most pharmaceuticals data are insufficient to precisely assess their risk and safety. Randomized controlled trials exclude pregnant and breast-feeding women for ethical reasons. In addition, most therapies in these special patient groups are off-label. For a qualified risk assessment, common reference books like the “Rote Liste” lack adequate information. Drug labels are often imprecise and tend to overestimate potential risks. Readymade drug risk information/labelling requires detailed information on the magnitude of risks including specification of the severity of expected toxic effects such as birth defects, if applicable. In case no risk has been found, the extent of human data should be specified on which safety conclusions were based. Furthermore, detailed recommendations are required how to counsel and manage in inadvertent drug exposure during an unplanned pregnancy. In contrast to ready-made risk labelling, Teratology Information Services (TIS) provide individual counselling and allow studies on drug safety. The largest German TIS, the Institute for Clinical Teratology and Drug Risk Assessment in Pregnancy, counsels 15,000 information requests per year. Data of drug-exposed pregnant patients are prospectively ascertained using structured questionnaires. Birth defect rates, effects on prematurity, miscarriage rates and other pregnancy complications can be investigated in association with prenatal drug exposure. During recent years several studies based on TIS-data have substantially contributed to risk estimation of a wide range of pharmaceuticals such as fluoroquinolones, antirheumatic MTX, rivaroxaban, TNF-alpha blockers, and atypical neuroleptics. These studies improve both the knowledge of drug effects on the unborn and the care of pregnant women. Acknowledgments: This work was supported by the German Federal Institute for Drugs and Medical Devices (BfArM). References: 1. Schaefer C. et al.: Arzneimittel in Schwangerschaft und Stillzeit (Urban & Fischer/Elsevier), 8th ed. 2012. 2. Schaefer C.; Peters P.; Miller R. K.: Drugs during Pregnancy and Lactation (Elsevier), 4th ed. 2015. DPhG Annual Meeting 2015 Conference Book • 87 SCIENTIFIC LECTURES SL.69 Medication for neonates, infants and children Läer, S.1* Institute of Clinical Pharmacy and Pharmacotherapy. Heinrich-Heine Universitaet Duesseldorf * on behalf of the LENA Consortium http://www.lena-med.eu/ 1 Drug use in children is characterized by a high rate of off-label use of about 30-50%. A systematic drug development in Europe has not started before 2007 when the European Paediatric Regulation has come into force. Drug development for children provides systematic data for drugs on dosing, dosing regimens according to safety measures, efficacy, short term safety and longterm safety. The LENA project acknowledges the current shortcomings of paediatric drug development and aims to develop an orally administered age-appropriate formulation of enalapril for use in neonates, infants and children. The development process will include investigatordriven trials that will collectively generate all necessary data for devising a paediatric-use marketing authorization (PUMA). The project will therefore conclude with a completely tested product ready for broad dissemination to the pediatric population in the European Union Member States. Acknowledgments: The research leading to these results has received funding from the EU’s Seventh Framework Programme (FP7/20072013) under grant agreement n°602295 (LENA). 88 • DPhG Annual Meeting 2015 Conference Book MEDICATION SAFETY IN SPECIAL PATIENT GROUPS SL.70 Medication safety of elderly patients in nursing homes Jaehde, U.1; Kulick, M.1; Bitter, K.1 1 Institute of Pharmacy, Department of Clinical Pharmacy, University of Bonn The multimorbidity of elderly patients often leads to polymedication. Therefore, these patients are at particular risk to suffer from drug-related problems and adverse drug reactions. Several studies have shown that the incidence of adverse drug reactions is particularly high in long-term care facilities leading to severe consequences such as hospitalization and death. In a prospectively designed cross-section analysis in North Rhine-Westfalia the incidence of adverse drug reactions was found to be 8 per 100 resident-months with a preventability rate of 60%. The majority of adverse drug events were caused by CNS and cardiovascular drugs [1]. Based on these data, a structured multiprofessional intervention was designed consisting of five measures: (1) intensive seminars for nurses and pharmacists, (2) advanced training for the prescribing general practitioners, (3) the implementation of a reminder card summarizing high-risk drugs and monitoring issues, (4) the formation of medication safety teams in each nursing home consisting of a nurse and a pharmacist, and (5) structured documentation and communication regarding individual drug therapy. The feasibility of this intervention was studied in 4 nursing homes in North Rhine-Westphalia [1]. Its efficacy is currently being evaluated in a large interventional trial (AMTS-AMPEL) in 18 nursing homes in North Rhine-Westphalia and Mecklenburg-Western Pomerania funded by the German Ministry of Health (BMG). Another approach to enhance medication safety of nursing home residents is based on a simple medication analysis by community pharmacists which is currently being evaluated in collaboration with a health insurance (AOK Rheinland/Hamburg) and the Pharmacists’ Association North Rhine. Based on prescription data of the AOK, the current medication and further information from the nursing home, the pharmacists detect drug-related problems (DRP) and propose potential solutions to the prescribing general practitioner. In a feasibility study including five community pharmacies, the pharmacists detected two DRP per patient in average. 33% of the DRP were drugdrug interactions of which 40% were considered as relevant for the residents’ medication safety. 39% of the patients took drugs considered as potentially inadequate in the elderly [2]. This new approach has currently been extended to 17 further community pharmacies and 20 nursing homes in North Rhine-Westphalia. In conclusion, incidence and severity of adverse drug events indicate serious deficiencies in the health care of elderly patients living in nursing homes. Medication safety-enhancing interventions are currently being developed in Germany that have the potential to improve the health status of the residents and to reduce costs, e.g. by avoiding unnecessary falls and hospitalization. References: 1. Jaehde U.; Thürmann P.: Z Evid Fortbild. Qual. Gesundh. wesen. 2012, 106: 712-6. 2. Bitter K. et al: DPhG Annual Meeting, September 23rd-25th, Düsseldorf, 2015. DPhG Annual Meeting 2015 Conference Book • 89 SCIENTIFIC LECTURES 2.18 Hot Topics in Pharmaceutical Biology – Young Investigators in the Spotlight Chairs: S. Alban, A. Vollmar SL.71 Cold atmospheric plasma – A future therapeutic approach? Bioanalytics as route to fundamental understanding Wende, K.1; Schmidt, A.1; Bekeschus, S.1; Hasse, S.1; Lalk, M.2; Masur, K.1; von Woedtke, T.1,3 Leibniz Institute for Plasma Science and Technology, INP Greifswald e.V., Felix-Hausdorff-Str. 2, 17489 Greifswald, Germany EMA University of Greifswald, Institute of Biochemistry, Felix-Hausdorff-Str. 4, 17489 Greifswald, Germany 3 Greifswald School of Medicine Institute for hygiene and environmental health, Walter-Rathenau-Str. 49A, 17475 Greifswald, Germany 1 2 Plasma medicine is an emerging biomedical field in which cold physical plasmas are generated to deliver a delicate mixture of reactive components to cells and tissues. Cold plasmas are complex gaseous systems, delivering energy to the tissue mainly as light and chemical entities. Of these, short lived oxygen or nitrogen centred reactive species gained the most attraction due to their potential ability to interact with biomolecules in a direct or indirect way. While knowledge on their generation and distribution in gas phase is rich, less is known about the composition of species in liquids [1]. Recent studies suggested a beneficial role of cold plasmas in the healing of chronic wounds wherein cells of the immune system are assumed to be responsible for the misled inflammatory response in the area of neoplastic diseases [2]. The mechanisms beyond these observations remain elusive and require exploration. Using bioanalytic techniques like molecular biology, metabolomics, transcriptomics and proteomics, the mechanism of plasma – tissue interaction was investigated. Data show, that cellular processes of human cells could both be stimulated or inhibited. Accordingly, gene and protein expression was affected by cold plasma treatment. The nuclear erythroid-related factor 2 (Nrf2) and phase II enzyme-pathway components were found to act as key controllers orchestrating the response [3]. Paracrine signals for cell-cell communication were detected, e.g. interleukin release. Wound relevant immune cells showed a markedly increased differentiation and changes in the metabolism [4]. For some conditions, an impact on the p53 – mitogen activated kinase pathway (MAPK) was found which led to pro- and anti-apoptotic events. No genotoxicity was detected in HRPT1 gene mutation assay [5]. Cold plasma Redox balance & oxidative signaling Eukaryotic cells & tissues p53 – MAPK signaling Nrf2 pathway activation Protein expression/degradation Interleukin secretion Pro- and antiapoptotic signals Metabolism Migration These data show that plasma impacts the cellular redox balance and intra- and extracellular signalling events. By this, hormesis-like effects bracing the cells against stress from within or without are activated, e.g. by the expression of defence enzymes, stimulation of cell differentiation, or metabolic changes. The findings spotlight the complex interaction of cold plasma with eukaryotic tissues and underline the demand for further elucidation. Results obtained so far foster the understanding of the current clinical observations and contribute to the recognition of the molecular processes leading to cold plasma generated effects in tissues. Acknowledgments: This work was founded in part by German Federal Ministry of Education and Research (grant number 03Z2DN11). References: 1. Wende, K. et al.: Biointerphases 2015, 10(2): 029518 2. Klebes, M. et al: J Biophotonics 2015, 8(5): 382 3. Schmidt, A. et al.: J. Biol. Chem 2015, 290(11): 6731 4. Bekeschus, S. et al.: Ox. Med. Cell. Longevity 2015, article 607969 5. Wende, K. et al.: Mutat. Res-Gen. Tox. En. 2015, submitted 90 • DPhG Annual Meeting 2015 Conference Book HOT TOPICS IN PHARMACEUTICAL BIOLOGY – YOUNG INVESTIGATORS IN THE SPOTLIGHT SL.72 About the potential of plant senescence as a new source for drug discovery Sendker, J.1 1 Institute of Pharmaceutical Biology and Phytochemistry, University of Münster, Correnstrasse 48, 48149 Münster, Germany In the course of plant senescence, a genetically controlled program is started that accounts for nutrient reallocation and establishes a strong oxidative environment in the senescing plant tissue. While the impact of senescence conditions on the proteome and primary metabolites has been intensely investigated, only little effort has been made towards elucidating senescence-associated changes of the secondary metabolome. Studies on the plant species of Prununs laurocerasus L., Hedera helix L., Juglans regia L. and Solanum dulcamara L. indicate that secondary metabolites undergo reproducible alterations in senescing leaves, ranging from subtle modifications to total decomposition of major compounds giving rise to catabolites hardly detectable in green leaves. Notably, senescence-associated changes can be biotechnologically provoked using the well-established and cheap treatment of ethylene-fumigation, which makes senescent plant material a promising source for drug discovery. Moreover, senescent plant material has proven to effectively aid the identification of bioactive natural products in metabolomic drug discovery studies: biotesting and LC-MS-profiling of 36 different Juglans extracts has led to the prediction of hydrojuglonglucoside as antitrypanosomal agent by means of Partial Least Squares (PLS) regression. The sample set included 9 senescent samples which by their distinct metabolic profiles and bioactivities significantly added on to sample variability, which is an essential prerequisite for the successful application of PLS or comparable methods of multivariate statistics. In order to confirm the predicted bioactivity of hydrojuglonglucoside, activity against Trypanosoma brucei was confirmed by testing the isolated compound [1]. Acknowledgments: Parts of the work were performed as part of the activities of the Research Network Natural Products against Neglected Diseases (ResNetNPND; http://www.resnetnpnd.org). References: 1. Ellendorf et al.: Molecules 2015, 20: 10082-10094. DPhG Annual Meeting 2015 Conference Book • 91 SCIENTIFIC LECTURES SL.73 Regulation of the heterologously expressed novobiocin gene cluster by the host strain Streptomyces coelicolor M512 Bekiesch, P.1; Maček, B.2; Forchhammer, K.3; Apel, A. K.1 Pharmaceutical Biology, Pharmaceutical Institute, Eberhard-Karls-Universität Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, Germany. 2 Proteome Center Tübingen, Eberhard-Karls-Universität Tübingen, Auf der Morgenstelle 15, 72076 Tübingen, Germany 3 Microbiology/ Department of Organismic Interactions, Interfaculty Institute of Microbiology and Infection, Eberhard Karls Universität Tübingen, Auf der Morgenstelle 28, 72076 Tübingen 1 The fast growing genome databases provide us with a large number of so far unknown secondary metabolite biosynthetic gene clusters. For actinomycetes these clusters are often heterologously expressed in an engineered host strain, as is Streptomyces coelicolor M512, to identify and investigate the corresponding compounds. However, production rates of secondary metabolites often are lower in the heterologous host strain. To better understand why this is the case, one has to know how the heterologously expressed gene cluster is regulated by the host strain itself. As a model system we chose the novobiocin biosynthetic gene cluster expressed in S. coelicolor M512 with a wellstudied operon structure and pathway specific regulation. Performing DNA affinity capturing assays (DACA) [1] combined with label-free mass spectrometry at three different time points on the promoters of the pathway specific regulatory genes novE (PnovE) and novG (PnovG), of the first biosynthetic gene novH (PnovH) and of the vegetative sigma factor gene hrdB (PhrdB) as a negative control we could identify and quantify a total of 2475 captured proteins. The proteins were binding to one or more of the tested promoter regions with intensities reaching from 103 to 1011. Among them the pathway specific regulator NovG was only captured on its binding site on PnovH, whereas most of the identified proteins bound to all tested promoter regions, including PhrdB. As positive charges on the surfaces of the proteins might lead to a low unspecific binding to negatively charged DNA in this very sensitive assay, we used the lowest NovG binding intensity, in the stationary phase, as threshold to exclude proteins with lower binding intensities. Interestingly we identified some well-known global regulators as e.g. NdgR, AdpA, SlbR or WhiA among those proteins binding with high intensity, but to all tested promoter regions. We then selected four proteins binding specifically to PnovH or PnovG to study them more in detail. Deletion and overexpression studies could confirm their regulatory function in novobiocin production. Furthermore for two proteins binding was confirmed by surface plasmon resonance and binding sites were determined likewise. These results indicate that DACA combined with label-free mass spectrometry is a very sensitive method and useful to get a broader view of the regulation of biosynthetic gene clusters. References: 1. Park, S. S. et al.: Ind. Microbiol. Biotechnol. 2009, 36: 1073-1083. 2. Dangel, V. et al.: Arch. Microbiol. 2008, 190: 509-519. 92 • DPhG Annual Meeting 2015 Conference Book HOT TOPICS IN PHARMACEUTICAL BIOLOGY – YOUNG INVESTIGATORS IN THE SPOTLIGHT SL.74 mRNA binding proteins in metaflammation and hepatocarcinogenesis Keßler, S.1 1 Department of Pharmacy, Pharmaceutical Biology, Saarland University, Saarbücken, Germany Hepatocellular carcinoma (HCC) is the second most cancer-related death and the sixth most common tumor type world-wide. Incidence of HCC is rising especially in Western countries due to alcohol abuse and metabolic diseases leading to fatty liver disease, which displays an important risk factor for HCC development. HCC typically develops within an inflammatory environment based on chronic inflammatory liver diseases. Since mRNA binding proteins can bind different transcripts and regulate their translation they are able to interfere with many different pathways. Therefore, mRNA binding proteins might display key factors in chronic liver disease and hepatocarcinogenesis, in which metabolism, inflammation, and carcinogensis engage with each other. The insulin-like growth factor 2 (IGF2) mRNA binding protein 2 (p62/IMP2) was originally isolated as an autoantigen from an HCC patient and shown to induce a fatty liver in transgenic mice [1]. We could decipher that p62-induced hepatic lipid accumulation is due to an IGF2-dependent activation of the lipogenic key regulator sterol regulatory element binding protein 1 (SREBP1) resulting in an induction of the elongase ELOVL6 [2]. During progression of fatty liver disease p62 is able to promote the manifestation of hepatic inflammation and fibrogenesis [3]. Analysis of human HCC samples revealed a strong overexpression of p62, which was correlated with poor prognosis and a more aggressive HCC phenotype [4]. Functional studies in p62 transgenic mice confirmed a stem cell-like and therefore more aggressive phenotype characterized by an increased number of chromosomal aberrations. In this context p62 induced the imprinted gene DLK1, which is able to activate the small Rho GTPase Rac1 and thereby leads to the generation of reactive oxygen species (ROS) [5]. Increased ROS levels can cause genomic instability, which is related to a more stem-like tumor phenotype. Taken together, the mRNA binding protein p62 displays an important regulator involved in all states of chronic liver disease. Therefore, p62 might display an interesting target for new treatment options for fatty liver and HCC therapy. References: 1. Tybl, E. et al.: J. Hepatol. 2011, 54:994-1001. 2. Laggai, S. et al.: J. Lipid. Res. 2014, 55:1087-1097. 3. Simon, Y. et al.: Gut 2014, 63:861-863. 4. Kessler, S. M. et al.: Am. J. Physiol. Gastrointest. Liver Physiol. 2013, 304:G328-G336. 5. Kessler, S. M. et al.: Cell death and disease 2015, in press. DPhG Annual Meeting 2015 Conference Book • 93 SCIENTIFIC LECTURES SL.75 Cyanobateria in natural product research – An interesting source for the discovery of new leads and motivation for developing tools to aid structure elucidation Niedermeyer, T.1; Kramer, D.2; Strohalm, M.3 1 Institute for Microbiology and Infection Research, Tübingen University, Auf der Morgenstelle 28, 72076 Tübingen, and German Center for Infection Research (DZIF), partner site Tübingen, Germany 2 Cyano Biotech GmbH, Magnusstr. 11, 12489 Berlin, Germany 3 Institute of Microbiology, Academy of Sciences of the Czech Republic, v.v.i., Videnska 1083, 142 20 Prague, Czech Republic Cyanobacteria have in the past been neglected by natural product scientists and received much less attention than other sources of secondary metabolites. However, nowadays they are acknowledged as a promising yet underexplored source of natural products with both novel structures and potent bioactivities, and the first drug product based on a cyanobacterial secondary metabolite has reached the clinic in 2011. Many cyanobacterial compounds exhibit toxic effects, such as e.g. the well-known microcystins, anatoxins, and saxitoxins. Other compounds, e.g. the microviridins, anabaenopeptins and microginins, show remarkably potent inhibition of proteases [1]. The majority of natural products from cyanobacteria are biosynthesised by non-ribosomal peptide synthetases and polyketide synthases (NRPS/PKS) [2]. Often, these compounds are synthesised as cyclic peptides. Being of non-ribosomal origin, they often feature uncommon and highly modified amino acids, making them attractive both from the point of view of their structure elucidation as well as their biosynthesis. In the first part of this talk I will introduce cyanobacteria as a prolific source of novel natural products and will shortly present some lead discovery projects I have been involved in. In the second part of the talk I will present a software tool we developed to aid in the annotation and interpretation of cyclic peptide tandem mass spectra. The mass spectrometric characterization of non-ribosomal cyclic peptides is challenging due to the predominant occurrence of non-proteinogenic amino acid monomers and the complex fragmentation patterns observed. Even though several software tools for the annotation of cyclic peptide tandem mass spectra have been published, these tools are still unable to annotate a majority of the signals observed in experimentally obtained spectra. They are thus not suitable for extensive mass spectrometric characterization of these compounds. This lack of an advanced and user-friendly software tool has motivated us to extend a freely available open-source software, mMass (http://www.mmass.org), to allow for cyclic peptide tandem mass spectra annotation and interpretation. The resulting software has been tested on several cyanobacterial and other naturally occurring peptides and has been found to be superior to all other tools currently available with regard to both usability and annotation extensiveness. Thus it is highly useful for accelerating the structure confirmation and elucidation of cyclic as well as linear peptides and depsipeptides [3]. Using this tool, we identified a large number of novel microginin derivatives in the Microsystis aeruginosa strain HUB 5-3. Microginins are short linear lipopeptides, and potent inhibitors of the angiotensin converting enzyme (ACE) [4]. Thus we decided to study their biosynthesis in this strain [5]. Although the PKS/NRPS gene cluster responsible for microginin biosynthesis on a first glance does not possess unique features, we were surprized to observe microginins that seem to be synthesized by only a part of the biosynthesis machinery. References: 1. Niedermeyer, T.; Brönstrup, M.: Microalgal Biotechnology: Integration and Economy (de Gruyter) 2012, 169-200. 2. Welker, M.; von Döhren, H.: FEMS Microbiol. Rev. 2006, 30: 530-563. 3. Niedermeyer, T. H. J.; Strohalm, M.: PLoS ONE 2012, 7, e44913. 4. Ishida, K. et al.: Tetrahedron 2000, 56, 8643-8656. 5. Kramer, D.: 2010, US 7,846,686 94 • DPhG Annual Meeting 2015 Conference Book HOT TOPICS IN PHARMACEUTICAL BIOLOGY – YOUNG INVESTIGATORS IN THE SPOTLIGHT SL.76 Analysis of natural product – Biosynthesis in the post-genomic era Schäberle, T. F.1 1 Pharmaceutical Biology, University of Bonn, Bonn/Germany Natural products have always been and will continue to the best source of lead structures for drug development. Over the last decades however, industry started doubting the use of natural products for the generation of new leads, since many known compounds have been rediscovered in bioactivity-based screening approaches. But in recent years, more and more insights into the molecular mechanisms of natural product synthesis were obtained. Most of the bioactive molecules are built up by polyketide synthase (PKS) and nonribosomal peptide synthetase (NRPS) biosynthetic assembly lines. Connecting the products to the genes encoding the biosynthetic machinery of these modular systems has stimulated new interest in natural product research, since this knowledge gave rise to genome mining approaches (Figure 1). Using the growing number of available genomic data, it became clear that the potential for the discovery of ever new structures has been by far underestimated, thus illustrating the lasting importance of natural products for drug discovery as an endless frontier instead of an ending era. The elucidation of biosynthetic gene clusters linked to natural products will be exemplified by recent examples from our laboratory. The gene loci of the antibiotically active compounds teixobactin and corallopyronin A will be presented [1,2]. From the latter, in detail analysis revealed even the basis for new enzymatic properties of double bond isomerization and of head to head polyketide fusion [3,4]. Figure 1. Duplex mode of the interconnection between a gene cluster and its corresponding metabolite References: 1. Ling, L. L. et al.: Nature 2015, 517: 455-459. 2. Erol, O. et al.: ChemBioChem. 2010, 11(9): 1253-65. 3. Lohr, F. et al.: Chem. Sci. 2013, 4: 4175-4180. 4. Zocher, G. et al.: Chem. Sci. 2015, DOI: 10.1039/c5sc02488a. DPhG Annual Meeting 2015 Conference Book • 95 96 • DPhG Annual Meeting 2015 Conference Book 3 SHORT POSTER LECTURES DPhG Annual Meeting 2015 Conference Book • 97 SHORT POSTER LECTURES SPL.001 / POS.018 Antibiotics from predatory bacteria – From discovery to MOA studies Nett, M.1; Schieferdecker, S.1; König, S.2; Korp, J.1; Werz, O.2 1 Leibniz Institute for Natural Product Research and Infection Biology/Hans-Knöll-Institute, Beutenbergstr. 11a, 07745 Jena, Germany and Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University, Philosophenweg 14, 07743 Jena, Germany 2 Pharmaceutical Bacteria, which prey on other microorganisms, are commonly found in the environment. While some predatory bacteria act as solitary hunters, others are known to band together in large, wolf pack-like consortia [1]. Anecdotal evidence suggests that the wolf pack-forming bacteria utilize antibiotics as part of their feeding strategy and, in fact, genome sequencing projects unveiled a rich and diverse secondary metabolome in these organisms [2,3]. We recently started to investigate the biosynthetic potential of selected predatory bacteria [4,5]. A Pyxidicoccus fallax strain, which had previously been isolated from the shores of the river Saale, was found to produce a new class of macrolide antibiotics with potent activity against staphylococci [6]. The gulmirecins did not exhibit cytotoxic effects against human cells and they also showed no cross-resistance with established antibiotics. Further analyses involving label incorporation into DNA, RNA, protein and cell wall biosynthesis revealed the molecular target of gulmirecin A. Subsequently, we isolated an antiproliferative compound with preferential activity against leukemic cells from the gulmirecin-producing P. fallax strain. The metabolite was identified as the known natural product myxochelin A and its antileukemic properties were traced to an inhibition of human 5-lipoxygenase [7]. Acknowledgments: Financial support by the Deutsche Forschungsgemeinschaft within the SFB 1127 “Chemical Mediators in Complex Biosystems” is gratefully acknowledged. References: 1. Nett, M.; König, G. M.: Nat. Prod. Rep. 2007, 24(6): 1245–1261. 2. Kiss, H. et al.: Stand. Genomic Sci. 2011, 5(3): 356–370. 3. Nett, M.: Progress in the Chemistry of Organic Natural Products (Springer) 2014. 4. Schieferdecker, S. et al.: J. Antibiot. 2014, 67(7): 519–525. 5. Schieferdecker, S. et al.: Eur. J. Org. Chem. 2015, (14): 3057–3062. 6. Schieferdecker, S. et al.: Chem. Eur. J. 2014, 20(48): 15933–15940. 7. Schieferdecker, S. et al.: J. Nat. Prod. 2015, 78(2): 335–338. 98 • DPhG Annual Meeting 2015 Conference Book SHORT POSTER LECTURES SPL.002 / POS.031 Targeting excipients for individual radiation therapy of cancer – Surface modification of PLGA polymer and liposome nanoparticles entrapping lanthanides by cholesterol- and PLA-bound ligands Nawroth, T.1; Krebs, L.1; Johnson, R.1; Langguth, P.1; Hellmann, N.2; Decker, H.2, Schmidberger, H.3; Goerigk, G.4; Boesecke, P.5a; Le Duc, G.5b; Bravin, A.5c; Schweins, R.6 Pharmacy & Biochemistry Institute, Pharmaceutical Technology, Johannes Gutenberg University, Staudingerweg, D-55099 Mainz, Germany 2 Molecular Biophysics Institute, Johannes Gutenberg University, Jakob Welder Weg 26, D-55128 Mainz, Germany 3 Department of Radiooncology and Radiotherapy (Clinics), University Medical Center; Langenbeckstr.1, D-55131 Mainz, Germany 4 HZB, Institute of Soft Matter and Functional Materials , BESSY Synchrotron, ASAXS, D-14109 Berlin, Germany 5 ESRF, European Synchrotron Radiation Facility, a)ID01, b)BioMedical Facility,c)ID17, 71 Avenue des Martyrs, F-38043 Grenoble, France 6 ILL, Institut Laue Langevin, DS / LSS, 71 Avenue des Martyrs, F-38042 Grenoble CEDEX 9, France 1 Nanoparticles can concentrate millions of drug molecules per unit and specifically target them to cancer cells, if they are recognized by cellular receptors [1,2]. Enhancer radiotherapy drugs increase the radiation cross section by Lanthanides for photon therapy PT, or Boron and Gadolinium for neutron capture therapy NCT. Case- and person-specific cell targeting results from surface modifications by signal lipids and anchor-ligands, recognized by disease specific cellular receptors. Our development, shall deliver a case- and person-specific cancer therapy. Our therapeutic nanoparticles of 100nm size are liposomes [1,5], polymers [7], including optionally bio-Ferrofluids [2]. The ligand proteins are bound by a fast click-link technique to anchor-linker constructs, which we synthetize bearing a terminal binding pre-activated S-group [6]. The ligand SH-group bearing (modified) proteins were bound in the last step resulting in artificial membrane proteins at the nanoparticle surface. We studied particle structure [4], drug load and release [5], and surface modifications with SANS, neutron reflectometry (ILL), ASAXS (ESRF, BESSY), DLS. Radio-therapy tests [3,5,7] were done with photons (PT) at the radiooncology clinics Mainz (linear accelerator) and the ESRF-synchrotron (ID17 biomedical facility), and with neutrons (NCT) at the ILL, and the TRIGA reactor Mainz [5]. For therapy tests we developed the EPN-test with kinetic cell cultures as tumor model [3]. Cell-specific nanoparticles for radiation therapy bear target excipients at the surface (artificial membrane protein) Acknowledgments: We are grateful for the funding by the German Ministry of Science and Education BMBF, grant 05KS7UMA. References: 1. Nawroth, T.; Rusp M.; May R.: Physica B 2004, 350(1-3): E635–638. 2. Alexiou, C. et al.: Eur. Biophys. J. 2006, 35: 446–450. 3. Buch, K. et al.: Radiation Oncology 2012, 7(1): 1–6. 4. Tenzer, S. et al.: ACS Nano 2011, 5(9): 7155–7167. 5. Peters, T. et al.: Radiation Oncology 2015, 10: 52–64. 6. Krebs, L.: 12/2014, Diploma thesis, Mainz (Bio. Medical. Chemistry BMC / in AK Pharmaceutical-Technology) 7. A particulate system for use in diminishing cell growth / inducing cell killing” EU-Patent 11 007 401.0 ; PCT 13 07 12; Johannes Gutenberg-Universität Mainz: Buch, K. et al.: (2012) / publication (2014) DPhG Annual Meeting 2015 Conference Book • 99 SHORT POSTER LECTURES SPL.003 / POS.036 Examination of homogeneity of stent coatings produced via fluidized bed process Wentzlaff, M.1; Senz, V.2; Grabow, N.2; Weitschies, W.1; Seidlitz, A.1 1 Institute 2 Institute of Pharmacy, C_DAT, Ernst-Moritz-Arndt University of Greifswald, Felix-Hausdorff-Straße 3, 17487 Greifswald, Germany for Biomedical Engineering, University of Rostock, Friedrich-Barnewitz-Straße 4, 18119 Rostock, Germany Stents were coated using a Mini-Glatt® fluidized bed apparatus (Mikro-Kit® product container) with the fluorescent model substance triamterene suspended in a dispersion of ammonium methacrylate copolymer (Eudragit® RS 30 D). Further components of the spray liquid were formic acid, triethylcitrate and purified water. Using the same process parameters 9 batches of 50 stents each (length 15 mm, unexpanded diameter 1.6 mm) were coated under addition of 625 steel springs per batch of comparable measures to fill the product container. The coating time and used volume of spraying liquid was varied to achieve 3 different coating masses (low, intermediate, and high coating mass with theoretical model substance deposition of 2.5, 5.0 or 12.5 µg/mm2 surface area) with 3 batches per mass. The obtained stents were examined regarding the coating process yield (mass gain of complete batch relative to mass of solid content of the sprayed coating liquid), stent body defect rate (percent deformed during the process), coating layer mass (differential weighing of coated stents and stents after complete coating removal), and the model substance content (fluorimetric determination after elution with methanol). Furthermore, stents were examined via fluorescence and scanning eletron microscopy. All coating processes were successfully completed with the intended process parameters. The process yield was 42±3% for the high coating mass, 41±3% for the intermediate coating mass, and 47±1% for the low coating mass. The defect rate of the stent bodies rose with increasing coating time but was overall low with a maximum defect rate of 8% for the high coating mass (mean processing time 138 min). Results of the determination of coating mass and model substance content as well as fluorescent microscopic images of stent sections are depicted in Figure 1. Smooth and form fitting coatings without polymer bridges or webbings were obtained for all batches. Even upon stent expansion to a diameter of 3 mm with a catheter pump only very few coating defects in the zones of deformation were observed. Coating split-off or delaminations were not observed during these experiments. Scanning electron microscopic images revealed a slightly smoother appearance of the coatings with low coating mass. Standard deviations for the coating mass as well as the model substance content were all below 10% and an acceptable reproducibility between the batches was obtained. Mean model substance loads of 58±3 µg (low coating mass), 113±6 µg (intermediate coating mass), and 271±15 µg (high coating mass) were detected. Drug-eluting stents are often coated individually using spray coating techniques. Such processes have been reported to yield 5 - 10 coated stents per hour and a deposition rate < 5% [1]. In the process reported here 675 units were coated simultaneously in a time ranging from 30 min (low coating mass) to 138 min with an average process yield of > 40% and a low defect rate. The variation regarding the model substance content was < 10% of the mean load which is comparable to drug content variation of commercially available drug-eluting stents [2]. Acknowledgments: Financial support by the federal ministry of education and research (BMBF) within REMEDIS is gratefully acknowledged. References: 1. Grabow, N. et al.: Biomed. Tech. (Berl.) 2013, 58(S1): DOI 10.1515/bmt-2013-4383. 2. Seidlitz, A. et al.: Biomed. Tech. (Berl.) 2012, 57(S1): DOI: 10.1515/bmt-2012-4120. 100 • DPhG Annual Meeting 2015 Conference Book SHORT POSTER LECTURES SPL.004 / POS.098 Reversing cisplatin resistance in ovarian carcinoma cells by inhibition of protein disulfide isomerase 1 Kalayda, G. V.1; Kullmann, M.1; Hellwig, M.1; Kotz, S.2; Hilger, R. A.3; Metzger, S.2,4; Jaehde, U.1 1 Institute of Pharmacy, Clinical Pharmacy, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany Biocenter, University of Cologne, Zülpicher Str. 47b, 50674 Cologne, Germany 3 Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Hufelandstraße 55, 45147 Essen, Germany 4 IUF-Leibniz Research Institute for Environmental Medicine, Auf’m Hennekamp 50, 40225 Düsseldorf, Germany 2 Cologne The clinical success of the widely used antitumor drug cisplatin is limited due to the frequently observed development of resistance in the course of the chemotherapy. Cisplatin resistance is a multifactorial event, which amongst others includes enhanced intracellular deactivation of the reactive cisplatin species. Recently, several intracellular binding partners of the fluorescent cisplatin analogue CFDA-cisplatin have been identified, among them the members of protein disulfide isomerase (PDI) family PDIA1 and PDIA3 [1]. The aim of our work was to elucidate the contribution of PDIA1 and PDIA3 to cisplatin resistance and to enhance cisplatin sensitivity by modulation of these proteins in A2780 human ovarian carcinoma cell line and its cisplatinresistant variant A2780cis. Knockdown of PDIA1 performed using the respective small interfering RNA increased cytotoxicity of cisplatin in resistant A2780cis cells as assessed by the MTT assay. Sensitivity of the parent A2780 cell line to cisplatin was slightly, however not significantly increased. On the contrary, PDIA3 knockdown had no influence on cisplatin cytotoxicity in both cell lines. A tendency for enhanced apoptosis (evaluated using the FITC Annexin V Apoptosis Detection Kit with PI®) was observed after PDIA1 knockdown in the A2780 and A2780cis cell lines. This was not the case after PDIA3 knockdown. Pharmacological inhibition of PDIA1 with PACMA31 [2] re-sensitized A2780cis cells to cisplatin treatment. In A2780 cells, the cytotoxicity of cisplatin was only marginally increased. Determination of the combination index revealed that cisplatin and PACMA31 act synergistically in both cell lines. However, synergism was much more pronounced in the cisplatin-resistant cells. Our results warrant further evaluation of PDIA1 as promising target for chemotherapy, and its inhibition by PACMA31 as a new therapeutic approach. Furthermore, combination of cisplatin with PACMA31 may help to reverse resistance in ovarian cancer. Acknowledgments: This project was supported by the Deutsche Forschungsgemeinschaft (JA 817/4-1). References: 1. Kotz, S. et al.: Electrophoresis 2015, in revision. 2. Xu, S. et al.: Proc. Natl. Acad. Sci. 2012, 109: 16348–16353. DPhG Annual Meeting 2015 Conference Book • 101 SHORT POSTER LECTURES SPL.005 / POS.101 Target-sensitive, vascular directed liposomes with entrapped chemokine receptor antagonists for a local interference with tumor cell metastasis Schlesinger, M.1; Roblek, M.2; Calin, M.3; Stan, D.3; Zeisig, R.4; Simionescu, M.3; Bendas, G.1; Borsig, L.2 1 Department of Pharmacy, Rheinische Friedrich-Wilhelms-University Bonn, 53121 Bonn, Germany of Physiology, University of Zürich and Zürich Center for Integrative Human Physiology, CH-8057 Zurich, Switzerland 3 Institute of Cellular Biology and Pathology ‘‘N. Simionescu’’ of the Romanian Academy, Bucharest, Romania 4 Experimental Pharmacology & Oncology Berlin Buch GmbH, 13125 Berlin, Germany 2 Institute Increased levels of chemokines are associated with augmented number of metastases and poor prognosis of cancer patients. The inflammatory chemokine CCL2 and its receptor CCR2 are known to initiate the early metastatic niche through recruitment of inflammatory myeloid cells and activation of the endothelium in proximity to cancer cells [1-4]. Both steps enable tumor cell transmigration and proliferation in the subendothelial tissue. A systemic blockade of the CCL2-CCR2 axis is demanding due to side effects and poor target specificity. Here we present PEGylated target-sensitive liposomes (TSL) encapsulated with a CCR2 antagonist (Teijin compound 1) coupled with a specific peptide recognized by endothelial VCAM-1, which enables a specific delivery to cancer cell-activated endothelial cells. Binding of TSL to endothelial VCAM-1 triggers the release of the CCR2 antagonist which leads to a subsequent blockade of endothelial CCR2. A local CCR2 blockade reduces an opening of tight junctions and finally the vascular permeability. Thus, tumor cells reveal a reduced transmigration and consequently the number of pulmonary metastatic foci in mice is attenuated. Nevertheless, the number of recruited inflammatory myeloid cells to the metastatic niche is not reduced. Application of VCAM-1 targeted TSL, loaded with Teijin compound 1, mitigated the number of pulmonary metastases in a murine (MC-38GFP cells) and a human xenograft (patient’s derived cells) model. Also in vitro, the transmigration of patient´s derived melanoma cells was strongly decreased. Hence, here we present a potential therapeutic approach for treatment of pulmonary metastasis without affecting homeostatic and immune functions. Acknowledgments: This work was supported by the frame of EuroNanoMed funded under the ERA-NET scheme of the Seventh Framework Programme of the European Commission. References: 1. Kitamura, T. et al.: J. Exp. Med. 2015, 212: 1043. 2. Qian, B.-Z. et al.: Nature 2011, 475: 222. 3. Zhao, L. et al.: Hepatol. Baltim. 2013, Md 57: 829. 4. Wolf, M. J. et al.: Cancer Cell 2012, 22: 91. 102 • DPhG Annual Meeting 2015 Conference Book SHORT POSTER LECTURES SPL.006 / POS.106 Rational design and diversity-oriented synthesis of peptoid-based selective HDAC6 inhibitors with potent anticancer activity Diedrich, D.1; Syntschewsk, V.1; Hamacher, A.1; Alves Avelar, L. A.1; Gertzen, C. G. W.1; Reiss, G. J.2; Kurz, T.1; Gohlke, H.1; Kassack, M. U.1; Hansen, F. K.1 Institut für Pharmazeutische und Medizinische Chemie, Heinrich Heine Universität Düsseldorf, Universitätsstr. 1, 40225 Düsseldorf, Germany 2 Institut für Anorganische Chemie und Strukturchemie, Heinrich Heine Universität Düsseldorf, Universitätsstr.1, 40225 Düsseldorf, Germany 1 Histone deacetylases (HDACs) catalyze the cleavage of acetyl groups from N-acetyl-lysine residues of histones and non-histone proteins. These posttranslational modifications (PTMs) are important for the regulation of gene transcription and protein function [1-5]. Human HDACs have been classified into four groups based on their homology to yeast histone deacetylases and cofactor dependence [1]. HDAC classes I (HDACs 1-3,8), IIa (HDACs 4,5,7,9), IIb (HDACs 6,10) and IV (HDAC 11) contain zinc-dependent deacetylase domains and are considered as “classical” HDACs [1-5]. Most of the currently used HDAC inhibitors (HDACi) target multiple HDAC isoforms and their clinical use may thus cause serious unwanted side effects [1]. Notably, there is increasing evidence that isoform-selective inhibitors are less toxic than pan-inhibitors [1,2]. Epigenetic drug discovery is therefore shifting towards the development of class- or isoform-selective HDACi. HDAC6, a class IIb enzyme, is structurally and functionally unique among the eleven human zinc-dependent histone deacetylases [5]. It is the only HDAC with two independent functional catalytic domains and a C-terminal zinc finger motif responsible for binding ubiquitinated proteins. The enzyme was initially described as tubulin deacetylase; however, it also modulates the function of other non-histone proteins implicated in regulatory processes, including cortactin, peroxiredoxins and Hsp90 [5]. Due to the large number of substrates, HDAC6 is involved in numerous diseases such as autoimmune disorders, inflammation, neurogenerative diseases and cancer [5]. Consequently, HDAC6 has emerged as an attractive target for the treatment of the above-mentioned diseases and the search for novel potent and selective HDAC6 inhibitors is of high importance [5]. We report here on the rational design and diversity-oriented synthesis of a series of selective HDAC6 inhibitors utilizing peptoid-based cap groups. A synthetic protocol based on an efficient and straightforward multicomponent approach allowed the rapid generation of a HDACi mini library. The biological evaluation of the target compounds included cellular HDAC and MTT assays on sensitive and chemoresistant cancer cell lines. The most active peptoid-based HDACi were investigated for their activity against selected HDAC isoforms and enhancement of cisplatin-induced cytotoxicity. Molecular modelling, MD simulations and docking studies allowed rationalization of the observed selectivity profile. Taken together, our data indicate that the peptoid-based HDACi are a new class of potent and selective HDAC6 inhibitors with remarkable activity against a panel of cancer cells of different chemosensitivity and tissue origin. Acknowledgments: This work was supported by funds from the Fonds der Chemischen Industrie (FCI). References: 1. Witt, O. et al.: Cancer Lett. 2009, 277(1): 8–21. 2. Marek, L. et al.: J. Med. Chem. 2013, 56(2): 427–436. 3. Hansen, F. K. et al.: ChemMedChem 2014, 9(3): 665–670. 4. Hansen, F. K. et al.: Eur. J. Med. Chem. 2014, 82: 204–213. 5. Kalin, J. H.; Bergman, J. A.: J. Med. Chem. 2013, 56(16): 6297–6313. DPhG Annual Meeting 2015 Conference Book • 103 SHORT POSTER LECTURES SPL.007 / POS.118 Inhibition of endothelial Cdk5 reduces tumor growth by promoting non-productive angiogenesis Merk, H.1; Zhang, S.1; Lehr, T.2, Bibb, J. A.3; Adams R. H.4,5; Zahler, S.1; Vollmar, A. M.1; Liebl, J.1 Department of Pharmacy, Pharmaceutical Biology, Ludwig-Maximilians-University, 81377 Munich, Germany Clinical Pharmacy, Saarland University, 66123 Saarbrücken, Germany 3 Department of Psychiatry and Neurology and Neurotherapeutics, The University of Texas Southwestern Medical Center, Dallas, Texas 75390-9070, USA 4 Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, 48149 Münster, Germany 5 Faculty of Medicine, University of Münster, 48149 Münster, Germany 1 2 Therapeutic success of VEGF-based anti-angiogenic tumor therapy is limited due to resistance. Thus, new strategies for anti-angiogenic cancer therapy based on novel targets are urgently required. Our previous in vitro work suggested that small molecule Cdk5 inhibitors affect angiogenic processes such as endothelial migration and proliferation. Moreover, we recently uncovered a substantial role of Cdk5 in the development of lymphatic vessels. Here we pin down the in vivo impact of endothelial Cdk5 inhibition in angiogenesis and elucidate the underlying mechanism in order to judge the potential of Cdk5 as a novel anti-angiogenic and anti-cancer target. By the use of endothelial-specific Cdk5 knockout mouse models and various endothelial and tumor cell based assays including human tumor xenograft models, we show that endothelial-specific knockdown of Cdk5 results in excessive but nonproductive angiogenesis during development but also in tumors, which subsequently leads to inhibition of tumor growth. As Cdk5 inhibition disrupted Notch function by reducing the generation of the active Notch intracellular domain (NICD) and Cdk5 modulates Notch-dependent endothelial cell proliferation and sprouting, we propose that the Dll4/Notch driven angiogenic signaling hub is an important and promising mechanistic target of Cdk5. In fact, Cdk5 inhibition can sensitize tumors to conventional anti-angiogenic treatment as shown in tumor xenograft models. In summary our data set the stage for Cdk5 as a drugable target to inhibit Notch-driven angiogenesis condensing the view that Cdk5 is a promising target for cancer therapy. 104 • DPhG Annual Meeting 2015 Conference Book SHORT POSTER LECTURES SPL.008 / POS.141 Design and optimization of N-benzyl benzamides: A novel fused scaffold for orally available dual sEH/PPARγ modulators for treatment of metabolic syndrome Proschak, E.1 1 Institute of Pharmaceutical Chemistry, Goethe-University Frankfurt, Max-von-Laue-Strasse 9, D-60438 Frankfurt a. M., Germany. The metabolic syndrome (MetS) is a multifactorial disease cluster consisting of dyslipidemia, cardiovascular disease, type 2 diabetes mellitus and obesity. Pharmacological intervention in the MetS is dependent on numerous drugs, thus polypharmacy is an obvious problem in the treatment of MetS patients. This study focuses on the dual target approach to accomplish a more efficient therapy for MetS. The two targets addressed by dual ligand design are the soluble epoxide hydrolase (sEH) and the peroxisome proliferator-activated receptor type γ (PPARγ). In vivo studies could demonstrate that even though an inhibitor of sEH or PPARγ agonist have benefits when used individually, the combination is more beneficial for the multidisease features in cardiometabolic syndrome [1]. Using a split-and-combine strategy we designed a library of dual sEH/PPARγ modulators and proved that both targets can be simultaneously addressed by a merged pharmacophore [2]. In a follow-up study, we designed lead-like merged N-benzyl benzamides which were able to modulate she and PPARγ. Structure activity relationship studies on both targets were performed resulting in an equipotent submicromolar (sEH IC50 = 0.3 µM / PPAR EC50 = 0.3 µM) propionic acid benzylbenzamide derivative. Evaluation in vitro and in vivo displayed good ADME properties qualifying the novel dual modulator as pharmacological tool compound for long term animal models of MetS. 8week evaluation in spontaneously hypertensive obese rats (SHROB), a rat model of MetS, demonstrated excellent efficacy including simultanious reduction of blood pressure and improvement of glucose tolerance. References: 1. Imig, J .D. et al.: Exp. Biol. Med. 2012, 237(12):1402-12. 2. Blöcher, R. et al.: J. Med. Chem. 2012, 55(23):10771-5. DPhG Annual Meeting 2015 Conference Book • 105 SHORT POSTER LECTURES SPL.009 / POS.167 Development and preclinical characterization of partial farnesoid X receptor agonists for metabolic disorders Merk, D.1 1 Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt, Germany; As a member of the nuclear receptor superfamily, farnesoid X receptor (FXR) acts as cellular regulator of bile acid homeostasis [1]. In this role, FXR controls the expression of several genes involved in bile acid, lipid and glucose homeostasis [2-4]. The involvement of FXR in several metabolic pathways offers a novel attractive drug target for the treatment of metabolic disorders, i.e. dyslipidemias and diabetes mellitus and first beneficial effects of pharmacological FXR activation on hyperglycemia have yet been reported from the clinical development of obeticholic acid which is the most developed FXR agonist so far [5]. However, the orchestra of nuclear receptors and their target genes is a very complex network that also bears the risk of severe side-effects as it has e.g. been observed for the glitazones that are agonists on the peroxisome proliferator-activated receptor γ (PPARγ). A potential way to reduce the risk of such undesired effects of nuclear receptor over-activation is the development of partial agonists that only activate the respective receptor to moderate amplitude [6]. Starting from a virtual screening program, we have developed a set of highly potent FXR partial agonists by systematic structure-activity-relationship studies and repeated rounds of structural optimization [7-9]. Preparation of co-crystal structures of selected representatives helped the structural optimization process and improved our knowledge on the molecular mechanism of partial FXR activation. The resulting agents are selective for FXR, display EC50 values for partial FXR activation in the low nanomolar range and activate the receptor to an amplitude of around 40% compared to the physiological agonist chenodeoxy cholic acid. Intensive in vitro pharmacological characterization revealed a direct interaction with the FXR-LBD and partial agonistic potency on all investigated FXR target genes. Moreover, the compounds showed good metabolic stability and low toxicity [7-9]. First in vivo data is very promising and warrants further exploration of the strategy and the compound class. The current optimization addresses the improvement of drug-likeness, stability and bioavailability to obtain a further optimized model agent for intensive in vivo investigation of partial FXR agonism as novel strategy to treat metabolic disorders. Acknowledgments: Financial support by the Else-Kröner-Fresenius-Stiftung and the Vereinigung der Freunde und Förderer der GoetheUniversität is gratefully acknowledged. References: 1. Makishima, M. et al.: Science 1999, 284(5418): 1362–1365. 2. Kuipers, F.; Bloks, V.; Groen, A.: Nat. Rev. Endocrinology 2014, 10(8): 488–498. 3. Düfer, M. et al.: Diabetes 2012, 61(6): 1479–1489. 4. Fang, S. et al.: Nat. Med. 2015, 21(2): 159–165. 5. Mudaliar, S. et al.: Gastroenterology 2013, 145(3): 574–582. 6. Merk, D.; Steinhilber, D.; Schubert-Zsilavecz, M.: Future Med. Chem. 2012, 4(8): 1015–1036. 7. Merk, D. et al.: Bioorg. Med. Chem. 2014, 22(8): 2447–60. 8. Merk, D. et al.: J. Med. Chem. 2014, 57(19): 8035–8055. 9. Merk, D. et al.: Bioorg. Med. Chem. 2015, 23(3): 499–514. and unpublished data 106 • DPhG Annual Meeting 2015 Conference Book SHORT POSTER LECTURES SPL.010 / POS.174 Time-resolved in situ assembly of the 5-lipoxygenase / 5-lipoxygenase-activating protein complex in primary human leukocytes Garscha, U.1; Gerstmeier, J.1; Werz, O.1 1 Chair of Pharmaceutical/ Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller University Jena, Philosophenweg 14, 07743 Jena, Germany 5-Lipoxygenase (5-LOX) catalyzes the first two steps in the biosynthesis of pro-inflammatory leukotrienes (LTs) from arachidonic acid (AA) [1]. Upon cell activation, 5-LOX translocates to the nuclear membrane where cytosolic phospholipase A2 (cPLA2) liberates AA that is subsequently transferred by 5-LOX-activating protein (FLAP) to 5LOX for efficient metabolism. Although biochemical or fluorescence microscopy data propose an association of 5LOX with FLAP [2,3], the in situ assembly of native 5-LOX/FLAP complexes in primary human cells remains elusive. The main objective of the present study was the time-resolved visualization of in situ 5-LOX/FLAP interaction at the nuclear membrane of human primary leukocytes in relation to 5-LOX activity. Immunofluorescence microscopy and proximity ligation assays [4] were applied to monitor 5-LOX/FLAP co-localization and interaction, respectively. We show for the first time an in situ 5-LOX/FLAP interaction in human blood monocytes and neutrophils. FLAP inhibitors (MK886, BAY X-1005) abolished 5-LOX/FLAP complex assembly but failed to block 5-LOX translocation and colocalization with FLAP. Inhibition of cPLA2 by RSC-3388 prevented 5-LOX/FLAP interaction but exogenously added AA restored it. Interestingly, 5-LOX/FLAP complex assembly appeared delayed at time points when 5-LOX product formation was already terminated. Though neutrophils and monocytes possess comparable quantities of catalytically active 5-LO enzyme, 5-LO activity in intact neutrophils is pronounced due to prolonged cellular 5-LO reaction, accompanied by delayed 5-LO nuclear membrane translocation. Conclusively, our data suggest that FLAP regulates 5-LOX product synthesis in two ways: by inducing an initial flexible association for efficient 5-LOX product synthesis, but also by formation of a tight 5-LOX/FLAP complex that terminates 5-LOX activity. Acknowledgments: Financial support was provided by Deutsche Forschungsgemeinschaft (DFG) within the SFB1127: Chemical Mediators in complex Biosystems. References: 1. Rådmark, O. et al.: Biochim. Biophys. Acta. 2015, 1851(4): 331-339. 2. Mandal, A.K. et al.: Proc. Natl. Acad. Sci. U. S. A. 2008, 105(51): 20434-9. 3. Bair, A.M. et al.: Mol. Biol. Cell. 2012, 23(22): 4456-64. 4. Söderberg, O. et al.: Nat. Method. 2006, 3(12): 995-1000. DPhG Annual Meeting 2015 Conference Book • 107 SHORT POSTER LECTURES SPL.011 / POS.203 Defined immobilisation of interleukin-4 (IL-4) for spatial controlled M2 macrophage polarization Lühmann, T.1; Spieler, V.1; Werner, V.1; Meinel L.1 1 Institute for Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany Introduction: Regulation of macrophage (Mφ) plasticity has broad implications in the treatment of a variety of inflammatory diseases, including, obesity, impaired wound healing, arteriosclerosis or rheumatoid arthritis. In the course of the inflammatory process, Mφ are the dominant infiltrating cells into the injured tissue and mediate temporally defined actions, by polarizing toward a spectrum of different phenotypes, including a classical (proimflammatory M1) and an alternative (anti-inflammatory, M2) polarization state [1]. Interleukin 4 (IL-4) is one of the major cytokines to trigger Mφ polarization to the M2 activation state, leading to cellular responses and an environment that stimulate endogenous tissue repair mechanisms. The therapeutic potential of human recombinant IL-4 has been previously investigated by subcutaneous administration in Phase II clinical trials but the treatment was associated with severe toxic side-effects [2]. Therefore novel delivery options for this interesting potent cytokine need to be explored. In this study, we aimed to circumvent the current challenges of IL-4 by deploying genetic codon extension to integrate the non-natural amino acid propargyl-L-lysine (Plk) into the protein backbone at position 42 during protein translation in E. coli. [3,4]. This approach enables site-specific immobilisation of Plk-IL-4 by biorthogonal chemistry, avoiding wandering species but rather addressing its Mφ polarization efficiency at specific sites. Methods: Human IL-4 and human plk-IL4 were expressed in E.coli BL21 (DE3) and were purified using cationic exchange chromatography as previously described [5]. Characterisation of plk-IL-4 was performed by MALDI-MS, ESI-LC-MS/MS and HPLC. Bioactivity of IL-4 and Plk-IL-4 was determined by proliferation of TF-1 cells and by using a secreted alkaline phosphatase (SEAP) STAT-6 reporter gene assay. NHS modified agarose particles were decorated with azide functionalities and Plk-IL-4 was immobilized using CuAAC chemistry. Monocytes were isolated from human peripheral blood mononuclear cells obtained from blood buffy coats by a two-step density gradient centrifugation and by magnetic associated cell sorting. Monocytes were differentiated into Mφ with M-CSF1 (unpolarized, M0) and further treated with a combination of M-CSF-1 and Plk-IL-4 or wild-type IL-4 for M2 polarization or with a cocktail of LPS and IFNγ for M1 polarization, respectively. Degree of Mφ polarization was assessed by RT-PCR with gene markers described in [6]. Results and Discussion: Plk-IL4 was successfully expressed in the presence of 3 mM Plk and was purified by cationic exchange chromatography in an analogue manner to wild-type IL-4. The correct incorporation of Plk at position 42 was confirmed by MALDI-MS and ESI-MS analysis and peptide mapping after trypsin digest. The soluble Plk-IL-4 was as active as the wild type analogue in respect to TF-1 cell proliferation and SEAP stimulation. M2 polarization of Mφ as analysed by M2 marker gene upregulation was similarly in the presence of soluble PlkIL-4 compared to the wild type analogue. Plk-IL-4 was successfully immobilized onto azide-functionalized agarose particles using CuAAC click chemistry. Proliferation of TF-1 cells was stimulated by plk-IL-4 decorated agarose beads in a dose (particle number) dependant manner and in comparison to controls, in which the immobilisation was performed without copper sulphate. The potential of immobilized Plk-IL-4 decorated agarose particles on Mφ plasticity is currently explored, aiming for lasting M2 Mφ polarization. Site-directed immobilisation of IL-4 is a powerful tool for decoration of material surfaces targeting tissue repair and wound healing by controlling Mφ plasticity in a spatial controlled manner. References: 1. Mosser, D.M.; Edwards, J. P.: Nat. Rev. Immunol. 2008, 8(12): 958–69. 2. Whitehead, R. P. et al.; J. Immunother. 2011, 25(4): 352–358. 3. Eger, S. et al.: Methods Mol. Biol. 2012, 832: 589–596. 4. Nguyen, D. P. et al.: J. Am. Chem. Soc. 2009, 131: 8720. 5. Kimmenade, A.V. et al.: Eur. J. Biochem. 1988, 173: 109–114. 6. Jaguin, M. et al.: Cell. Immunol. 2013, 291: 51–61. 108 • DPhG Annual Meeting 2015 Conference Book SHORT POSTER LECTURES SPL.012 / POS.210 Understanding Plasmodium falciparum’s exploitation of the innate immune system aids the identification of potential novel intervention strategies Schmidt, C. Q.1; Kennedy, A. T. 2; Harder, M. J. 1; Lim, N. Y. T. 2; Tham, W. H. 2 1 Ulm University, Institute of Pharmacology of Natural Products & Clinical Pharmacology, Ulm, Germany University of Melbourne, Department of Medical Biology and The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia. 2 The Introduction: Malaria remains one of the world’s deadliest diseases. Of the five human malarias, Plasmodium falciparum inflicts the most mortality and severe form of human malaria. From initial entry into the human body P. falciparum parasites face the innate immune system’s first line of defence against invading pathogens, the complement system. This ancient defence system can directly lyse pathogens via formation of the membrane attack complex or lead to phagocytosis via opsonisation of the pathogen with C3b. Malaria parasites are exposed to complement when merozoites, the invasive form of the life cycle, are released from a spent erythrocyte into circulation. Recently, complement receptor 1 (CR1) on erythrocytes has been identified as an alternative entry receptor for invasion utilized by merozoites during P. falciparum blood stage infection [1,2]. CR1 regulates and protects erythrocytes from self-damage of the host’s complement system, but during merozoite invasion the parasite adhesin PfRh4 interacts directly with the conserved N-terminal regulatory region of CR1 to mediate successful invasion [3]. Upon engagement of CR1 by PfRh4 some complement regulatory functions of CR1 are inhibited exposing the merozoite and invaded red cells to the danger of complement mediated destruction [4]. Objectives: This study aims (i) to identify the amino acids in CR1 that mediate binding to the parasite adhesin PfRh4 and (ii) to produce soluble domain fragments of CR1 that inhibit the PfRh4 invasion pathway. The third objective (iii) investigates how the merozoites evade the destruction by the human complement system. Methods: The first two N-terminal domains of CR1, as well as a panel of rationally designed mutants (homologous substitution) were expressed recombinantly and tested for biophysical binding characteristics for PfRh4 interaction as well as for their activity to inhibit erythrocyte entry of merozoites in growth culture. In regards to immune evasion merozoites obtained from parasite culture were exposed to human serum or complement proteins for subsequent binding studies and functional analysis to identify routes of complement evasion. Results: We have produced wildtype CR1 complement control protein (CCP) module pairs 1-2 (CCP1-2) and CCP1-2 variants bearing specific mutations. All proteins were expressed in the host Pichia pastoris, purified to homogeneity and validated by mass spectrometry. Using surface plasmon resonance (SPR) we determined affinity constants for PfRh4 binding, thus shedding more light onto the bimolecular interaction between CR1 and the malaria adhesion PfRh4. We show how these mutations affect malaria parasite recognition of red blood cells and how they affect parasite growth in a CR1-dependent manner. We identified the CR1 amino acids 18 and 20 (sequence numbers) as crucial binding partners for PfRh4. In terms of inhibiting the CR1-dependent invasion pathway, we discovered that the recombinant CR1 protein consisting of CCP1-2 efficiently blocks parasite invasion, in vitro. Finally, we investigated how the merozoites resist clearance by the complement system. Merozoites were exposed to human non-immune serum and were found to specifically recruit the host complement regulators Factor H (FH) and FHL-1 protein (FHL-1). Both human complement regulators retained their functional activity when bound to the merozoite surface. By employing a recombinant FH CCP-domain fragment library, the domains 4 to 6 in FH were identified to be responsible for the recruitment by the malaria parasites. Co-immunoprecipitation experiments with soluble parasite lysate identified one merozoite surface protein as the interaction partner for FH/FHL-1 recruitment. Preventing FH/FHL-1 binding to merozoites enhanced complement-mediated parasite lysis. Conclusion: We have identified the binding patch on CR1 that mediates erythrocyte invasion via the parasite adhesion protein PfRh4 and showed that this invasion pathway can be inhibited in vitro with small recombinant protein fragments of CR1. We also show that FH recruitment to merozoites protects the parasites from complement-mediated lysis and have identified the merozoite protein responsible for mediating FH/FHL-1 recruitment. These findings have important implications for the future design of vaccine strategies or the development of peptide inhibitors. References: 1. Tham, W-H. et al.: Proc. Natl. Acad. Sci. 2010, 107(40): 17327–32. 2. Spadafora, C. et al.: PLoS Pathog. 2010, 6(6): e1000968. 3. Park, H. J. et al.: J. Biol. Chem. 2014, 289(1): 450–63. 4. Tham, W-H.; Schmidt, C.Q. et al.: Blood 2011, 118(7):1923–33. DPhG Annual Meeting 2015 Conference Book • 109 110 • DPhG Annual Meeting 2015 Conference Book 4 POSTERS DPhG Annual Meeting 2015 Conference Book • 111 POSTERS 4.1 Biopharmaceutics POS.001 Comparison of two binding kinetic assays in long term studies for performance qualification using Biacore X100 Steinicke, F.1; Oltmann-Norden, I.1; Wätzig, H.1 1 Institute of Medicinal and Pharmaceutical Chemistry, Technical University of Brunswick, Beethovenstraße 55 in 38106 Braunschweig Surface Plasmon Resonance (SPR) is a dominant tool for biomolecular interaction characterization. This technique facilitates label-free binding analysis studies of biomolecules such as affinity, kinetic, thermodynamics and specificity analysis in real-time. Therefore SPR is an important application in drug discovery and proteomics. In this study the model system for measuring binding kinetics was β2 microglobulin from human urine and the antibody anti-β2 microglobulin produced in mouse. The antibody was covalently bound by amine coupling on the surface of the gold chip and the antigen was flowed over the chip. Two different assay designs were implemented to determine kinetic rate constants (kd, ka), dissociation constants (KD), residuals from the experimental to the optimal fitted curve (RU) and the maximal Response Units (Rmax) as an indicator for ageing of the chip/antibody. The first method was a titration series called Single Cycle Kinetic (SCK) where subsequently five different concentrations of β2 microglobulin (2, 4, 8, 16 and 32 nM) were injected. One regeneration step followed subsequently to loose off the β2 microglobulin from the antibody. The second method was a Multi Cycle Kinetic (MCK) where all concentrations were injected in their own cycle with two regeneration steps after each. As a further variation the randomization of the injection sequence was evaluated in MCK. The dilution series was prepared freshly every day with a HEPES-buffer pH 7.4 and the regeneration solution was glycineHCl 10 mM pH 2.5. The investigation of this antibody/antigen system by using both SCK and MCK methods reveals that the parameter that represents best the aging of the chip was Rmax. In SCK the Rmax was decreasing slightly after 20 runs without affecting the quality of the other kinetic parameters. Interestingly in contrast to SCK in MCK Rmax was stable over 30 cycles although there are five times more regeneration steps for one data set than in SCK. That implies that the ageing of the chip is not only caused by the harsh regeneration steps but mainly by the time period it is used. The most stable parameter measured was the kd with a relative percental standard deviation (RSD%) of 2.5 to 6.2% over a minimum of 30 measurements. In contrast the ka was more unstable within the range of 6.5 to 30%. Acknowledgments: We thank GE Healthcare Life Sciences for excellent technical support References: 1. Rich, R. L.; Myszka, D. G.: J. Mol. Recognit. 2011, 24(6): 892-914. 2. GE Healtcare: Biacore X100 Getting Started 2009 3. Olaru, A. et al.: Crit. Rev. Anal. Chem. 2015, 45(2): 97-105. targeting ligand for folate receptor-mediated uptake and as an anticancer agent as it is toxic to the target cells by blocking de novo thymidylate and purine synthesis and consequently DNA and RNA synthesis [4]. Besides, we exploit a therapeutic siRNA against Eg5 that blocks mitosis to cause death of rapidly dividing cancer cells. Together we formulate Eg5 siRNA and MTX-conjugated polymer as a nanosized tissue-specific siRNA polyplex with synergistic antitumor effect (Figure A). The sizes of siRNA polyplexes were estimated by atomic force microscope. The cellular uptake was determined by flow cytometry, and knockdown of a luciferase reporter gene was used to monitor gene silencing efficiency. To evaluate the efficacy of Eg5 siRNA, we measured toxicity by cell viability assay, mRNA expression by qRT-PCR, and aster formation of cellular DNA. In NMRI-nude mice bearing KB xenograft tumors, the intratumoral retention of Cy7-labeled siRNA polyplexes was determined by NIR fluorescence bioimaging to investigate the tissuespecific targeting, and tumor size as well as aster formation in vivo after the treatments with EG5 siRNA polyplexes were monitored to assess the therapeutic potency. The siRNA polyplexes were homogeneous spherical nanoparticles with 6.5 nm of hydrodynamic diameter. These polyplexes were taken up by KB cells in a MTX-dependent manner, and this attributed to association with folate receptor. Transfections induced significant silencing of luciferase expression in KB/eGFPLuc cells. Treatments with MTXconjugated polyplexes containing Eg5 siRNA in KB cells triggered knockdown of Eg5, resulted in typical aster formation, and caused enhanced cytotoxicity. in vivo studies indicated that the MTX-based 640 polyplexes showed significantly enhanced intratumoral retention (168 h) compared to non-targeted alanine-substituted 188 polyplexes (48 h) (Figure B), and mediated the longer survival time (56 days) than untreated controls (24 days). We developed a specific and efficient siRNA carrier system with dualfunctional ligand for cellular delivery and antitumor effect. siRNA polyplexes successfully delivered cargo into the target cells mainly via folate receptor. When combined with therapeutic Eg5 siRNA and MTX, the siRNA polyplexes carried out synergistic cytotoxic activity. This highly functionalized and molecule-defined carrier system for siRNA delivery could be a potential strategy for RNAi-based cancer therapeutics. Acknowledgments: DFG Excellence Cluster Nanosystems Initiative Munich (NIM), Bavarian Research Foundation, Dr. Rong Zhu References: 1. Lächelt, U.; Wagner, E.: Chem. Rev. 2015, DOI: 10.1021/cr5006793. 2. Lee, D. J.; Wagner, E.; Lehto, T.: Methods in Molecular Biology (Humana) 2015. 3. Dohmen, C. et al.: ACS Nano. 2012, 6(6): 5198-208. 4. Lächelt, U. et al.: Mol. Pharm. 2014, 11(8): 2631-9. POS.002 Targeted Co-delivery of Bifunctional GlutamylMethotrexate and Eg5 siRNA Using Nanoplexes for Combined Antitumoral Potency Lee, D. J.1,2; Kessel, E.1; He, D.1,2; Klein, P.1; Lächelt, U.1,2; Lehto, T.1; Wagner, E.1,2 1 Department of Pharmacy and Center for NanoScience, Ludwig Maximilian University, Butenandtstraße 5-13, Munich, 81377, Germany 2 Nanosystems Initiative Munich, Schellingstraße 4, Munich, 80799, Germany The novel strategy that synthetic small interfering RNA (siRNA) can invoke RNAi responses is expected to be an excellent option for treating many incurable diseases such as cancer. However, efficient tissuespecific delivery of siRNA remains the major limitation in the development of RNAi therapy [1]. By solid phase supported synthesis, we have synthesized a series of sequence-defined polymers which include a cationic (oligoethanamino)amide core, cysteines (as bioreversible disulfide-forming units), and polyethylene glycol chain (for shielding surface charges) coupled to a terminal ligand [2,3]. To recognize the target cells, the antifolate drug methotrexate (MTX) is employed as both 112 • DPhG Annual Meeting 2015 Conference Book POS.003 2D separation of proteins: Combining the strengths of SAX and CGE Maul, K. J.1,2; Hahne, T.1; Wätzig, H.1,2 1 Institute of Medicinal and Pharmaceutical Chemistry, Technische Universität Braunschweig, Beethovenstraße 55, 38106, Germany 2 PVZ – Center of Pharmaceutical Engineering, Franz-Liszt Straße 35a, 38106, Germany The two techniques HPLC with a strong anion exchanger (SAX) column and capillary gel electrophoresis (CGE) are both commonly used in protein analysis. In this work a combination of both techniques is used to characterize a complex protein mixture such as biopharmaceuticals. This is still mostly done by using classic two dimensional gel electrophoresis (2D-GE). To achieve comparable results the combination of SAX and CGE was chosen. In the first step the proteins were separated according to their charge at a certain pH using SAX-HPLC. Afterwards, in the second dimension CGE was used. The separation of this technique is based on BIOPHARMACEUTICS POS.004 A modified disintegration tester for solid dosage forms: Influence of motion and velocity profiles on disintegration behaviour Rach, R.1; Kindgen, S.1; Nawroth, T.1; Abrahamsson, B.2; Langguth, P.1 Institute of Pharmacy and Biochemistry, Department of Pharmaceutical Technology and Biopharmacy, Johannes Gutenberg University, 55128 Mainz, Germany 2 Pharmaceutical Development, AstraZeneca R&D, 43183 Mölndal, Sweden 1 The disintegration of solid oral dosage forms is determined by the USP/PhEur disintegration test device. In this compendial method a basket-rack assembly containing six dosage forms is raised and lowered in immersion fluid over a distance of 55 mm for 29 to 33 times per minute. There is no appreciable horizontal motion or movement of the axis from the vertical and no possibility of velocity adjustment [1]. There are no investigations whether this direction of movement respectively its velocity imitates physiological conditions. In addition the physiological conditions vary over the gastrointestinal (GI) tract. For example the stomach, where immediate release dosage forms mainly disintegrate, is divided into several parts with different stress conditions. In the proximal part, the fundus, the mechanical stress conditions are significantly lower in comparison to the distal part, the antrum [2]. In this project a new designed modified disintegration tester is used to investigate the influence of movement directions and velocities on disintegration of tablets. Acknowledgments: This work has received support from the Innovative Medicines Initiative Joint Undertaking (http://www.imi.europa.eu) under Grant Agreement No. 115369, resources of which are composed of financial contribution from the European Union’s Seventh Framework Program and EFPIA companies' in kind contribution. 100 100 80 80 60 40 Sugar-based 0.1 N HCl Isomalt-based 0.1 N HCl 20 Isomalt-based SSF 0 10 20 30 60 40 Sugar-based 0.1 N HCl Isomalt-based 0.1 N HCl 20 Sugar-based SSF 0 Drug release [%] Acknowledgments: We thank PolymicroTM for providing the capillaries. To evaluate in vivo parameters relevant for drug release in the oral cavity, we performed a study in 6 healthy volunteers. The study “medication” consisted of different types of commercial candies that were either sugarbased or sugar-free or differed in taste, respectively. The volunteers had to suck a candy at 3 different times of the day over 3 different days and the following parameters were recorded: overall sucking time, temperature and pH in the oral cavity before and immediately after sucking, saliva osmolality before and after sucking. In a second series of tests, the saliva secretion rate during sucking was recorded. The same candy formulations were then subject to dissolution experiments with compendial test methods for lozenges, i.e. experiments were performed in the paddle apparatus using 500 mL 0.1 N hydrochloric acid or water as test media. In the final set of in vitro experiments, we wanted to address essential in vivo parameters such as the presence of small fluid volumes, continuous saliva flow, saliva composition, agitation and tongue pressure acting on the dosage form. We thus designed a new test apparatus consisting of a custom-made flow through cell that allows for using small media volumes, and an “artificial tongue” which can be agitated to slide over the dosage form at a predetermined agitation rate. Simulated Saliva Fluid [1] was used as the test medium. Drug release [%] the molecular size. These two steps are very similar to 2D-GE, where isoelectric focussing is combined with SDS-PAGE. For this approach a mixture of five proteins was used to determine the suitability and precision of the combination. These proteins were namely Myoglobin, β-Lactoglobulin, Ovalbumin, bovine serum albumin and a monoclonal antibody. The first dimension is conducted at pH 8.5 with gradient elution. The sodium chloride concentration of the eluent was increased from 0.0 M to 0.75 M over 40 min. 20 fractions were collected. As second dimension CGE was used with a SDS-gel buffer. This whole analysis takes less than 24 hours to get a 2D separation. In both dimensions a DAD was used to collect the data. The advantages compared to 2D-GE are the short analysis time, the possibility to detect in both dimensions and the higher precision. 40 Sugar-based SSF Isomalt-based SSF 0 0 2 Time [min] 4 6 8 10 12 Time [min] Figure: Release profiles obtained from two lozenge formulations obtained in 0.1 N HCl and SSF [1] with the paddle apparatus (500 mL, 75 rpm) (A) and with the new dissolution apparatus for lozenges (9 mL/min, 180 dpm) (B), mean of n= 3 ± S.D., the grey arrows indicate average in vivo sucking time. Results obtained with the compendial dissolution methods available for testing drug release from lozenge formulations might be useful for quality control but, as indicated by the time required for total dissolution of the formulations, did not relate to the real sucking process. It was thus clear that when the aim is to estimate in vivo sucking time and drug release in the oral cavity, a different test setup would be required. Results from the experiments performed with the new dissolution apparatus, in contrast, are very promising. The new setup will thus be applied to a range of additional lozenge formulations and with different simulated saliva fluids with the aim of proposing a set of test parameters that can be applied for predicting in vivo drug release and potentially also be used for quality control of lozenges. References: 1. Mashru, R. C. et al.: Drug Dev. Ind. Pham. 2005, 31(1): 25-34. References: 1. The International Pharmacopeia, Methods of Analysis, http://apps.who.int/phint/en/p/docf/ (22.05.2015) 2. Koziolek, M. et al.: Eur. J. Pharm. Sci. 2014, 57: 250-256. POS.006 Application of patient-specific test methods in estimating in vivo drug release of oral mesalazine formulations POS.005 Development of a new test methodology for predictive in vitro dissolution testing of lozenges Tietz, K.1; Gutknecht, S.1; Klein, S.1 1 Institute of Biopharmaceutics and Pharmaceutical Technology, Department of Pharmacy, Ernst Moritz Arndt University, Felix Hausdorff Street 3, Center of Drug Absorption and Transport, 17489 Greifswald, Germany A variety of lozenges for local drug delivery to the oral cavity is on the market since quite a long time. However, when the objective is to compare drug release from these formulations in a predictive way, no guideline is available to assist in establishing an appropriate in vitro dissolution test. The purpose of the present set of experiments was thus to evaluate critical in vivo parameters for drug release in the oral cavity, to screen the applicability of compendial dissolution methods in terms of obtaining results being predictive for the in vitro performance of lozenge formulations, and finally to evaluate the applicability of a new methodology in terms of predicting drug release of lozenges in the oral cavity. Karkossa, F.1; Krüger, A.1; Klein, S.1 1 Institute of Biopharmaceutics and Pharmaceutical Technology, Department of Pharmacy, Ernst Moritz Arndt University, Felix Hausdorff Street 3, Center of Drug Absorption and Transport, 17489 Greifswald, Germany There is an ongoing discussion on - apart from clinical trials - how to demonstrate therapeutic equivalence for locally applied and locally acting products in the gastrointestinal tract. Possibly, among other alternatives, in vitro drug release models could be considered surrogates of drug release and availability at the site of action. However, to date the conditions in which in vitro models provide valid surrogates of in vivo release and availability would have to be defined. To demonstrate the potential applicability of in vitro test methods for screening therapeutic equivalence of locally acting oral mesalazine formulations and also to get an idea of which would be the right dosage form for an individual patient, recently a series of in vitro studies was performed comparing a variety of in vitro release methods ranging from pharmacopoeial methods through methods addressing average gastrointestinal transit times and pHconditions to first experiments simulating gastrointestinal conditions in individual subjects [1]. The objective of the present series of tests was to further by applying detailed individual pH-gradients as obtained in study DPhG Annual Meeting 2015 Conference Book • 113 POSTERS performed with a pH-sensitive capsule [2] and also a set of novel bicarbonate-based biorelevant media to simulate intestinal passage of the dosage forms. Figure: Release profiles obtained when simulating the gastrointestinal passage of Salofalk 500 mg and Claversal 500 mg tablets in three different individuals, paddle apparatus, 500 mL, 75 rpm, mean of n= 3 ± S.D., the shaded area represents small intestinal residence time. For this purpose, we selected three individual fasted gastrointestinal transit profiles representative for essentially different small intestinal passages [2], i.e. relatively low, average, and relatively high small intestinal pH conditions and different gastrointestinal transit times. Experiments were performed with Salofalk 500 mg tablets and Claversal 500 mg tablets in the Paddle apparatus. After simulating gastric residence in SGFsp, the pHysio-grad [3] device was used to simulate small and large intestinal transit in bicarbonate-based biorelevant media. Our results clearly indicate that the two mesalazine formulations will show an essentially different in vivo performance in individual subjects. Whereas in the patients with low and high small intestinal pH, drug release of both formulations is expected to take place in the mid or distal small intestine, respectively, in the patient with average small-intestinal pH conditions only the Salofalk formulation will release the active in the (terminal) small intestine. Drug release from the Claversal tablet will, in contrast, initiate in the (proximal) colon. Based on the observations made in the present series of tests we conclude that with a set of physiologically-based dissolution models taking into account the particular features in gastrointestinal physiology and typical dosing scenarios in the target patient group, it should be possible to estimate the in vivo performance of oral formulations, to discriminate between formulations and finally to contribute to a safe and effective drug therapy for the individual patient. References: 1. Klein, S.: Pharmazie 2015; 70(8). 2. Koziolek, M. et al.: J. Pharm. Sci. 2014, 19 (2014) EPub. 3. Garbacz, G. et al.: Eur. J. Pharm. Sci. 2014, 51: 224-31. 114 • DPhG Annual Meeting 2015 Conference Book PHARMACOLOGY 4.2 Pharmacology POS.007 Influence of nuclear receptor ligands on the expression of metabolizing enzymes and transporter proteins in the human intestine Brueck, S.1; Busch, D.1; Martin, P.2; Haenisch, S.2; Cascorbi, I.2; Siegmund, W.1; Oswald, S.1 1 Department of Clinical Pharmacology, Center of Drug Absorption and Transport (C_DAT), University Medicine Greifswald, Felix- Hausdorff-Str. 3, 17487 Greifswald, Germany 2 Institute of Experimental and Clinical Pharmacology, University Hospital Schleswig-Holstein, Arnold-Heller-Str.3, 24105 Kiel, Germany Background: Drug transporters and drug metabolizing enzymes located in the human intestine are crucial determinants in the pharmacokinetics of orally administered drugs. Nuclear receptor ligands (NRL) like rifampicin and carbamazepine are known to influence expression of these proteins and can cause thereby unwanted drug-drug interactions (DDI). However, the available knowledge is rather limited and was predominantly focused on gene expression data. Moreover, little is known about the intestinal regulation of enzymes and transporters beyond activation of nuclear receptors (e.g. miRNA). Thus, it was the aim of this study to comprehensively analyse the impact of NRL on gene and protein expression of clinically relevant enzymes and transporters and the potential regulatory role of miRNAs in the human intestine. Methods: Tissue specimen (~2-5 mg) from lower duodenal mucosa were taken before and after treatment with rifampicin (600 mg, 8 days) [1] or carbamazepine (600 mg, 18 days) [2], respectively. Protein abundance of clinically relevant intestinal transporters was quantified using a validated LC-MS/MS-based targeted proteomics assay. In parallel, gene expression (mRNA and miRNA) analysis was conducted using TaqMan® real-time RT-PCR assays. Results: Rifampicin treatment led to a significant increase in gene expression (over 1.5-fold) of ABCB1, ABCC2, CYP2B6, CYP2C19, CYP2C8, CYPC9, CYP3A4, CYP3A5 and UGT1A family and in protein abundance of ABCB1, CYP3A4, CYP2C9 and UGT1A1. In contrast to this, pre-treatment with carbamazepine led to no significant changes on protein levels of the investigated transporters and enzymes, whereas mRNA expression of ABCB1, ABCC2, ABCG2, SLCO2B1, CYP2C19, CYP2C9, CYP2D6, CYP3A4, CYP3A5, SULT1A family and UGT1A family were found to be significantly increased. The observed changes in gene expression and protein content were correlated to intestinal miRNA expression. Conclusion: This study investigated for the first time the impact of rifampicin and carbamazepine on the gene and protein expression of clinically relevant enzymes and transporters in the human intestine in a comprehensive manner. Rifampicin was found to be a strong inducer of several enzymes and transporters which is in good agreement to profound DDI observed in clinical studies. In addition to the activation of nuclear receptors, the expression of intestinal enzymes and transporters may be influenced by miRNAs. References: 1. Giessmann, T. et al.: Clin. Pharmacol. Ther. 2004, 76(3): 192-200. 2. Oswald, S. et al.: Clin. Pharmacol. Ther. 2006, 79(3): 206-17. hyperalgesia after topical application of high-concentration (40%) menthol in comparison to its solvent ethanol [1,2]. Method: The study was a single-center, randomized, placebo controlled, double-blind, 2 period cross-over trial in the “balanced placebo design” (BPD) [3], in a cohort of 16 healthy subjects. The randomization was twofold: firstly, subjects were assigned to the order of substance application and, secondly, subjects were assigned to the order of information about the applied substance given by the investigator during the two applications of each substance. Treatments were topical menthol (40%) in ethanol as model for cold and mechanical hyperalgesia and topical ethanol as control condition. Cold and mechanical hyperalgesia and allodynia were determined by selected parameters of quantitative sensory testing (QST) [4]. The sensational thresholds and area of pinprick-hyperalgesia were quantified. Results: This study was performed with reliable, valid and repeatable QST measurements. For all parameters no suggestion effect could be detected in the multivariable models. Menthol reliably induced cold hyperalgesia. The cold pain threshold (CPT) was decreased statistically significant (p<0.0001) and a distinct effect was found for treatment condition and drug condition vs. placebo and control condition. Minor suggestion effects on cold pain thresholds could be seen in a subgroup of subjects that have not received menthol before. This subgroup showed decreased cold pain thresholds in the placebo condition compared to control condition. However, cold hyperalgesia seemed to be the most robust parameter. Effect sizes were similar to those previously published [1,2,5]. Pinprick hyperalgesia was less reliable. Mechanical pain threshold (MPT) and mechanical pain sensitivity (MPS) only changed marginally after application of menthol in treatment as well as in drug condition. Effect sizes differed from previously published data [2,5]. The area size of pinprick-hyperalgesia was not influenced statistically significant. Parameters like mechanical pain threshold (MPT) and mechanical pain sensitivity (MPS) seemed to be influenced by treatment sequence effect and period effect. Conclusion: There is no statistically significant effect of suggestion or expectation on the measured parameters. The menthol model is therefore a reliable, non-suggestible model to induce cold hyperalgesia. Pinprick hyperalgesia is not as reliable to induce and may be influenced by treatment sequence and periodic effect. Performing the model, future studies should consider a prior demonstration of menthol to familiarize the subject with the expected effect. Acknowledgments: The research leading to these results is a part of the Europain Collaboration, which has received support from the Innovative Medicines Initiative Joint Undertaking, under Grant Agreement No. 115007, resources of which are composed of funding from the European Union’s Seventh Framework Programme (FP7/2007–2013) and the kind contribution of EFPIA companies. References: 1. Wasner, G. et al.: Brain 2004, 127(5): 1159-1171. 2. Binder, A. et al.: J Pain 2010, 12(7): 764-773. 3. Enck, P.; Klosterhalfen, S.; Zipfel, S.: BMC Med Res Methodol 2011, 11: 90. 4. Rolke, R. et al.: Pain 2006, 123(3): 231-243. 5. Mahn, F. et al.: Eur J Pain 2014, 18(9): 1248-1258. POS.009 POS.008 Impact of Suggestion and Expectation on a Human Experimental Model of Cold and Mechanical Hyperalgesia after Topical Application of High-Concentration Menthol in Comparison to Ethanol Barnscheid, L.1; Helfert, S. M.2; Reimer, M.2; Rengelshausen, J.1; Baron, R.2; Binder, A.2 1 Early Clinical Science, Translational Science & Strategy, Grünenthal GmbH, 52099 Aachen, Germany 2 Division of Neurological Pain Research and Therapy, Department of Neurology, University Hospital Schleswig-Holstein,24105 Kiel, Germany Background: Human experimental pain models play an important role in studying neuropathic pain mechanisms. The objective of the present study was to evaluate the impact of suggestion and expectation on a human experimental model of cold and mechanical (pinprick) Characterization of the expression and function of endogenous transporters in frequently used cellular models Otter, M.1; Eriksson, P. O.1; Keiser, M.1; Oswald, S.1 Department of Clinical Pharmacology, Center of Drug Absorption and Transport (C_DAT), University Medicine Greifswald, Felix-Hausdorff-Str. 3, 17487 Greifswald, Germany 1 Background: Drug transporters are known to be important determinants in the pharmacokinetics and efficacy of many drugs. In order to characterize the affinity or inhibitory properties of drugs to transporter proteins in preclinical drug development, transporter-overexpressing cellular models based on MDCKII and HEK293 cells are widely used and recommended by EMA and FDA guidelines. However, there is evidence that these cells possess a considerable endogenous (“background”) transporter expression which might affect in vitro transport. In this study, we compared the expression profile of frequently used cellular models with the respective tissue of origin. Furthermore, we investigated the endogenous transporter expression in different MDCKII and HEK293 cell lines. On this basis, functional consequences of endogenous ABCB1 were examined in cellular uptake studies. DPhG Annual Meeting 2015 Conference Book • 115 POSTERS Methods: Renal tissue from human or dog and the following cell lines have been investigated: HEK293 wild-type cells or stably transfected with control vector, OATP1A2, OATP1B1, OATP1B3 or OATP2B1; MDCKII wild-type cells or stably transfected with control vector or ABCB1. The mRNA expression of clinically relevant human ABC and SLC transporters (HEK293 cells) and the respective canine transporters (MDCKII cells) were determined by validated TaqMan® gene expression assays. Protein abundance was quantified by LC-MS/MS-based targeted proteomics. The functional study was performed with talinolol and trospium chloride as probe substrate of OATP1A2 and ABCB1 using wild-type, stably transfected vector control and OATP1A2overexpressing HEK293 cells. Intracellular accumulation of [3H]-talinolol and [3H]-trospium chloride was measured by liquid scintillation counting after cell lysis. Results: The observed gene expression and protein abundance data were mostly not correlated. For only few transporters such as ABCB1, the expression in cellular models and tissue of origin was very similar while expression of the most transporters (e.g. typical renal transporter PEPT2) was markedly different in both cellular models. Several drug transporters could be identified as endogenous transporters in the different cell lines. In HEK293 cells, the endogenous ABCB1 protein content was considerably lower in vector control-, OATP1A2-, OATP1B1-, OATP1B3and OATP2B1-transfected cells compared to wild-type cells. On the contrary, markedly higher expression has been observed for endogenous ABCC2 in HEK-OATP1B1 and for endogenous ABCC3 in MDCK-ABCB1 cells. Uptake studies regarding the functional consequences of endogenous transporters showed that higher protein abundance of endogenous ABCB1 in wild-type cells resulted by trend in reduced intracellular accumulation of ABCB1 probe substrates compared to vectortransfected cells. Conclusion: There are markedly differences in expression pattern of endogenous transporters in the investigated HEK293 and MDCKII cells. These endogenous transporters may affect drug transport and the estimated transporter affinity to the focused transporter protein. POS.010 Modulation of the l-arginine-nitric oxide pathway by apelin in an in vitro model of pulmonary arterial hypertension Glatzel, A.1; Lüneburg, N.1; Klose, H.2; Böger, R. H.1; Harbaum, L.2 1 Institute of Clinical Pharmacology and Toxicology, University Medical Center HamburgEppendorf, Germany ² Section Pneumology, II. Department of Medicine, University Medical Center HamburgEppendorf, Germany Background: Idiopathic pulmonary arterial hypertension (IPAH) is a difficult-to-treat rare lung disease characterised by endothelial dysfunction with increased vasoconstriction and occlusive vascular remodeling [1-3]. A dysfunctional endothelial L-arginine-nitric oxide (NO) pathway is a key pathomechanism of IPAH and can be provoked by hypoxia in in vitro models [4-5]. The small peptide apelin is involved in the maintenance of pulmonary vascular homeostasis [6]. However, its precise mechanism of action is still unclear. Asymmetric dimethylarginine (ADMA) is an endogenous inhibitor of endothelial NO synthase and is associated with clinical parameters in IPAH and other cardiovascular diseases [7-9]. ADMA is degraded by dimethylarginine dimethylaminohydrolase 1 and 2 (DDAH) enzymes [10]. Objective: To determine the influence of apelin on the L-arginine/NO pathway in human microvascular pulmonary endothelial cells (HPMECs). Methods: HPMECs were cultured under normoxic and hypoxic conditions and treated with apelin. The expression of regulators of the Larginine/NO pathway were analysed using real-time PCR. In addition, the effect of apelin on the the phosphoinositide-3 kinase (PI3K)/Akt signalling pathway was determined using an immunoassay and specific inhibitors. Apelin and ADMA concentrations were measured in cell culture supernatants and IPAH patients’ serum using an enzyme-linked immunosorbent assay (ELISA) and a validated high throughput liquid chromatography–tandem mass spectrometry (LC-MS/MS) assay. Results: Circulating Apelin was lower in patients with IPAH compared to age-matched healthy subjects. Its receptor (APLNR) expression was reduced on HPMECs following treatment with apelin due to a negative feedback mechanism. Apelin directly influenced the L-arginine/NO 116 • DPhG Annual Meeting 2015 Conference Book pathway by increasing the expression of DDAH2, but not DDAH1. Thus, the concentration of ADMA was decreased in HPMECs supernatant by apelin. This effect was observed under normoxic conditions as well as under PAH-related hypoxic conditions. Furthermore, apelin induced the phosphorylation of several proteins playing a role in the PI3K/Akt signalling pathway. Foremost the glycogen synthase kinase 3 alpha and beta (GSK-3α/ß) was inactivated as a result of apelin treatment. An inhibition of GSK-3α/ß by GSK-3 inhibitor I mimicked the effect of apelin on DDAH2 expression. Conclusion: Apelin directly modulates the L-arginine/NO pathway and mediates enhanced degradation of ADMA, which may increase NO synthesis. The effect of apelin on DDAH2 expression is caused by inhibition of GSK-3α/ß. An Apelin receptor agonist might be a novel and promising therapeutic option for treatment of IPAH. References: 1. Rabinovitch, M. et al.: J. Clin. Invest. 2012, 122(12): 4306-13. 2. Galiè, N. et al.: Eur. Respir. J. 2009, 34(6): 1219-63. 3. Humbert, M. et al.: J. Am. Coll. Cardiol. 2004, 43(12): 13S-24S. 4. Cooper, C. J. et al.: Circulation 1996, 93(2): 266-71. 5. Lüneburg, N. et al.: Biomed. Res. Int. 2014, 2014(2014):501612 6. Andersen, C. et al.: Pulm. Circ. 2011, 1(3): 334-46. 7. Vallance, P. et al.: J. Cardiovasc. Pharmacol. 1992, 20(12): 60S-62S. 8. Kielstein, J. T. et al.: Arterioscler. Thromb. Vasc. Biol. 2005, 25(7): 1414-8. 9. Zoccali, C. et al.: Lancet 2001, 358(9299): 2113-7. 10. Ogawa, T. et al.: Arch. Biochem. Biophys. 1987, 252(2): 526-37. POS.011 Regulation of mitogen-activated protein kinase kinase kinase DLK by calcineurin Duque Escobar, J.1,3; Lemcke, T.2; Hasenpusch, D.2; Oetjen, E.1,2,3 1 Institut für Klinische Pharmakologie und Toxikologie, Universitätsklinikum Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany 2 Institut für Pharmazie, Bundesstr. 45, 20146 Hamburg, Germany 3 DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck Loss of beta-cell mass is the most important factor for the pathogenesis of type 1 and type 2 diabetes. It is unknown which mechanisms induce beta-cell apoptosis though different hypotheses are postulated. Inhibition of the calcium-calmodulin dependent phosphatase calcineurin has been shown to reduce beta-cell function and induce beta-cell apoptosis. Our previous results showed that inhibition of calcineurin by the structurally distinct immunosuppressant drugs cyclosporin A and tacrolimus decreased insulin gene transcription [1]. Furthermore, both drugs stimulated the catalytic activity of the mitogen-activated protein kinase kinase kinase 12 (DLK Dual Leucine zipper kinase) and induced betacell apoptosis [2]. In the present study, the regulation of DLK by calcineurin was investigated. In silico analysis revealed two putative calcineurin interaction domains within DLK [3]. The consensus motifs 273-PNMLIT-278 and 362-LPVP365 were mutated to PNRLKT, APAP and a double mutant, respectively, by primerless PCR. The expression vectors for these mutants and DLK wild-type were transiently transfected into the beta-cell line HIT. Immunoblot analysis using an antibody against the COOH-terminus of DLK showed similar expression levels of the single mutants, whereas the double mutant was barely expressed. Both single mutants exhibited no catalytic activity, measured as phosphorylation of DLK on Ser-302 and phosphorylation of the downstream kinase JNK. A DLK homology model was generated, using the crystal structures of MLK1 and B-Raf as templates: The model confirmed that 273-PNMLIT-278 interfered with the conformation of the catalytic domain. Mutation of L362 to A and V364 to PHARMACOLOGY A within the second putative calcineurin interaction domain 362-LPVP365 did not reduce DLK protein expression. The L362A DLK mutant showed no catalytic activity in relation to Ser-302 phosphorylation of DLK and JNK phosphorylation, whereas the V364A DLK mutant resulted in 3fold higher JNK phosphorylation than DLK wild-type. In addition, in reporter gene assays this mutant was more potent to inhibit KCl/Forskolin-induced CRE-dependent gene transcription in a dosedependent manner, than DLK wild-type. Considering that LxVP represents a calcineurin interaction domain, our results suggest that the interaction of DLK with calcineurin via its LPVP motif dephosphorylates and inhibits DLK activity. These findings also show the importance of calcineurin for the maintenance and survival of beta-cell function and mass. The compounds were further investigated at the CB-like orphan G protein-coupled receptors GPR18 and GPR55, both of which are known to interact with certain CB receptor ligands [6]. β-Arrestin recruitment assays were employed [7]. Only few of the studied compounds displayed a moderate inhibition of GPR55 activation, while none of the compounds showed any activation or inhibition of GPR18. References: 1. Pertwee, R. et al.: Pharmacol. Rev. 2010, 62(4): 588-631. 2. Howlett, A. et al.: Pharmacol. Rev. 2002, 54(2): 161-202. 3. Lindigkeit, R. et al.: Forensic. Sci. Int. 2009, 191(1-3): 58-63. 4. Banister, R. et al.: Chem. Neurosci. 2015, DOI: 10.1021/acschemneuro.5b00107. 5. Rempel, V. et al.: J. Med. Chem. 2012, 55, 7967-7977. 6. Henstridge, C. et al.: Mol. Endocrinol. 2011, 25(11): 1835-1848. 7. Rempel, V. et al.: Med. Chem. Comm. 2002, 54(2): 161-202. References: 1. Oetjen, E. et al.: Mol. Pha. 2003, 63(6): 1289-95. 2. Plaumann, S. et al.: Mol. Pha. 2008, 73(3): 652-9. 3. Rodríguez, A. et al.: Mol. Cell. 2009, 33(5): 616-26. POS.013 POS.012 Pharmacological evaluation of synthetic cannabinoids identified as constituents of spice Schoeder, C.1; Hess, C.2; Madea, B.2; Müller, C. E.1 1 Pharma-Zentrum Bonn, Pharmazeutisches Institut, Pharmazeutisches Chemie I, An der Immenburg 4, D-53121 Bonn, Germany 2 Universitätsklinikum Bonn, Institut für Rechtsmedizin, Forensische Toxikologie, Stiftsplatz 12, D-53111 Bonn, Germany The cannabinoid (CB) receptor CB1 mediates the main psychoactive effects of the natural product Δ9-tetrahydrocannabinol (THC) which acts as a partial agonist at the receptor. The second CB receptor CB2 is mainly expressed in the immune system [1]. Both CB receptors are Gi proteincoupled mediating inhibition of adenylate cyclase and thereby reducing intracellular cAMP levels. In the past decades a wide range of potent synthetic CB receptor agonists and antagonists has been developed [2]. Natural and synthetic CB1 agonists are being abused due to their psychoactive, euphoric and analgesic effects, e.g. as ingredients of products commercialized as incense called “spice”. Spice has been declared to be a herbal product, but potent synthetic CB receptor agonists have been found in many spice preparations, which are not just used as incense, but typically abused by smoking the dubious mixture of unknown composition. Due to severe side effects many of the synthetic CB agonists found in spice preparations are now on the list of controlled substances [3]. However, the drug market is steadily flooded with new synthetic CB receptor agonists that are not yet subject to control by the authorities. To obtain compounds with CB1 receptor-agonistic effects, typical bioisosteric variations of known potent CB receptor agonists are observed, e.g. replacing hydrogen by fluorine atoms, indole by indazole residues and naphthyl by quinolone ring systems [4]. The pharmacological profiles of these compounds are often unknown, and therefore forensic consequences for producers, traders and consumers are not enforceable since a scientific basis is lacking. In the present study we investigated a series of compounds collected by the Institute of Forensic Medicine and suspected to act as CB1 receptor agonists. Neuroprotective effects of monoacylglycerol lipase inhibition in a 6-hydroxydopamine model of Parkinson’s disease in mice Porazik, C.1,2; Witting, A.2; Ferger, B.1 1 CNS Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach/Riß, Germany 2 Department of Neurology, Ulm University, Germany Monoacylglycerol lipase (MAGL) is the principal enzyme for 2arachidonylglycerol (2-AG) metabolism and plays an important role in the endocannabinoid system. Here, we investigate the effects of the commercially available MAGL inhibitors KML29 and JZL184 in comparison with the cyclooxygenase 2 (COX2) inhibitor rofecoxib in an animal model of Parkinson’s disease (PD). 6-Hydroxydopamine (6-OHDA) (3.5 µg/µl) was administered in the left striatum in male C57BL/6JRj mice. KML29, JZL184 and rofecoxib (30 mg/kg/day p.o.) were administered daily for one week starting one day before 6-OHDA surgery. Motor behaviour indicated by rotarod measurement was assessed at day 7. Post mortem (day 7), striatal dopamine depletion and nigral tyrosine hydroxylase positive (TH+) cell loss were evaluated. Furthermore, the effect of MAGL and COX inhibition on mechanistic biomarkers was assessed. 6-OHDA led to a 70% dopamine depletion in the striatum and 40% loss of TH + neurons in the substantia nigra. KML29 treatment significantly attenuated the 6-OHDA-induced dopamine depletion by 50% and TH+ neuron loss by 30%. Motor behavior was impaired by 6-OHDA but not restored by MAGL inhibition. Overall, the effects of JZL184 and rofecoxib treatment were less pronounced. The MAGL inhibitors significantly increased 2-AG levels and decreased arachidonic acid and prostaglandins in the brain. Rofecoxib decreased prostaglandins but had no effect on 2-AG and arachidonic acid levels. In conclusion, targeting the endocannabinoid system by MAGL inhibition was found to be superior over COX2 inhibition in the 6-OHDA PD model and could be a valuable new therapeutic concept for PD in the future. Acknowledgments: The study was funded by the Boehringer Ingelheim Ulm University BioCenter (BIU) and supported technically by Johannes Hanselmann and Noemi Pasquarelli. POS.014 Targeting cardiomyocyte proliferation for heart regeneration using phenotypic screens in neonatal mouse cardiomyocytes. Carrillo García, C.1; Raulf, A.2; Hesse, M. 2; Fleischmann, B. 2; Schade, D.1 Department of Chemistry and Chemical Biology, TU Dortmund, Otto-Hahn-Str. 6, D-44227 Dortmund, Germany. 2 Intitut für Physiologie 1 - Life and Brain Center, Universität Bonn, Sigmund-Freud Str. 25, D53105 Bonn, Germany. 1 The compounds were investigated in radioligand binding assays for their interaction with both CB receptor subtypes, CB1 and CB2. Subsequently they were investigated for their functional properties in cAMP accumulation assays [5]. Almost all of the investigated compounds were found to be highly potent CB receptor agonists - some compounds showing even subnanomolar affinities for the CB1 and CB2 receptor. There is a great medical need for innovative therapies for the treatment of heart failure and a true paradigm shift is required which aims at treating the actual cause instead of rather coping with the symptoms. In this regard, replacing lost heart tissue with new cardiomyocytes after myocardial infarction is of particular interest. In contrast to the adult DPhG Annual Meeting 2015 Conference Book • 117 POSTERS mammalian heart, the developing heart (embryonic and neonatal) exhibits a pronounced regenerative capacity. Hence, there is a pressing need for a detailed understanding of the underlying physiological mechanisms of cardiomyocyte development during this neonatal period (e.g. mitotic arrest) to reveal the biological basis of potential regenerative mechanisms and for successful therapeutic translation [1]. Here, we present a ‘forward chemical genomics’ approach to achieve this goal. The basis is the establishment of a phenotypic assay that quantifies cell cycle activity and cytokinesis (along with other phenotypic features) of neonatal cardiomyocytes via high-throughput-compatible fluorescence microscopy in 384- and 1536-well plates (high-content analysis). For this, we isolate and dissect transgenic neonatal murine hearts which express H2B-mCherry under the control of a cardiac specific promoter (=Myh6), allowing us to unambiguously detect and quantify cardiomyocyte nuclei [2]. To date, we could show general feasibility of this primary cells-based approach for high-throughput application and have characterized the very few described pro-proliferative compounds and growth factors. Several key phenotypic endpoints have been addressed including proliferative activity, cardiomyocyte cell number, cytokinesis and nuclearity, the latter being of particular importance for cardiomyocytes and their ability to proliferate. Therefore, next steps will be to systematically screen focused small molecule libraries with annotated targets and signaling pathways to rapidly draw conclusions regarding involved biological factors, pathways and mechanism. Later, evaluation of compound libraries that cover a large chemical space will provide “hits” that possibly reveal completely new biological mechanisms. References: 1. Schade, D.; Plowright, A.T.: J. Med. Chem. 2015, accepted. 2. Raulf, A. et al.: Basic Res. Cardiol. 2015, 110(3): 33. 118 • DPhG Annual Meeting 2015 Conference Book NATURAL COMPOUNDS 4.3 Natural compounds POS.015 Xysmalobium undulatum everything began (Uzara®) research – how Helmstädter, A.1 1 Institute of Pharmaceutical Chemistry, Goethe University, Max-von-Laue-Str. 9, 60438 Frankfurt, Germany Xysmalobium undulatum (Uzara®) is a traditionally used medicinal plant from South Africa. It found its way into European research in the early 20th century and has been widely used as a remedy against diarrhoea since then. Circumstances of knowledge transfer and early research into phytochemistry and therapeutic potential of X. undulatum have been largely unknown so far. As could be shown, the drug was brought to Europe by a former soldier, Wilhelm Heinrich Adolph Hopf (1887-1929). At the age of 16, W. H. A. Hopf joined the German navy, but some years later he moved to South Africa, where he was engaged in the Boer War. He married Mary Annie Langham Thomson (1876-1967) December 16, 1906. Most probably in 1909, the family returned to Melsungen (along with the Uzara roots) and stayed until the end of World War I. Then, Hopf and his wife moved to England, lived in Rugby and changed their name from Hopf to Hopford on 19 November 1918. They had two children. He died december 27, 1929 in Rugby. Details about Hopf’s life are provided here the first time, as well as some aspects of early research on the drug, which has mainly been done at the University of Marburg, Germany. POS.016 The acetyl-CoA carboxylase (ACC) inhibitor soraphen A blocks the proliferation and migration of primary endothelial cells Glatzel, D.1; Müller, R.2; Koeberle, A.3; Werz, O.3; Fürst, R.1 1 Institute of Pharmaceutical Biology, Biocenter, Goethe-University, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany 2 Department of Microbial Natural Products, Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarland University, Campus Building C2.3, 66123 Saarbrücken, Germany 3 Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, University Jena, Philosophenweg 14, 07743 Jena, Germany Acetyl-CoA carboxylase catalyzes the first step in the biosynthesis of fatty acids in bacterial and eukaryotic cells, i.e. the conversion (carboxylation) of acetyl-CoA into malonyl-CoA. ACC-generated malonylCoA functions as a substrate for de novo lipogenesis and acts as an inhibitor of mitochondrial fatty acid β-oxidation. Because of its role in lipid metabolism this enzyme has become an interesting target in drug discovery in the field of metabolic diseases and cancer. Despite the high interest in ACC as pharmacological target, no attention has as yet been given to the role of ACC in endothelial cells. We aimed to investigate the role of ACC in two functional key aspects of angiogenesis: endothelial cell proliferation and migration. To inhibit the function of ACC, we used the ACC inhibitor soraphen A, a polyketidic natural compound isolated from the myxobacterium Sorangium cellulosum, as well as an RNAi-based approach. Primary human umbilical vein endothelial cells (HUVECs) were used as in vitro model. First, we analyzed the action of soraphen A on cell viability. The compound did neither lower the metabolic activity of HUVECs up to a concentration of 100 µM after 24 and 48 h (CTB assay) nor increase in the apoptosis rate after 24, 48, or 72 h up to 100 µM. Measuring adenosine triphosphate (ATP) levels revealed that 30 µM soraphen A does not alter the ATP levels in HUVECs after 24 h treatment. In contrast, a 48 h treatment significantly lowers the ATP levels by 12%. Also gene silencing of ACC1 in HUVECs attenuated the ATP levels by 11%. Mitochondrial membrane potential (MMP) assays showed 10% decreased MMP levels in soraphen A-treated cells after 24 h. Interestingly, the compound inhibited the proliferation of endothelial cells with an IC50 value of 34 µM. Cell cycle analysis showed that soraphen A decreases the amount of cells in the G0/G1 phase by 26% and increases the number of cells in the G2/M phase by 50%. In a wound healing/scratch assay, 30 µM soraphen A lowered the migration of endothelial cells by 65%. Also gene silencing of ACC1 in HUVECs strongly decreased endothelial migration. Furthermore, Boyden chamber assays revealed that soraphen A can also lower chemotactic migration by 34%. Since actin rearrangement is necessary for migratory processes, we analyzed the F-actin cytoskeleton (microscopy) and found that soraphen A decreased the number of filopodia by 60% but did not influence stress fiber formation. Surprisingly, soraphen A-treated cells did not exhibit significant alterations in their capacity to form tube-like structures on Matrigel. In summary, we could gather first hints that inhibiting ACC has an immense impact on the proliferation and migration of primary endothelial cells. The mechanistic basis of this phenomenon will be investigated in future studies by analyzing the lipid profile and the transcriptome of endothelial cells. Acknowledgments: This work was supported by the German Research Foundation (DFG, FOR 1406, FU 691/9-2). POS.017 Does STW 5 have region specific effects in the intestine? Nieber, K.1; Voß, U.1; Abdel-Aziz, H.2; Okpanyi, S.2; Kelber, O.2 1 Institut für Pharmazie, Universität Leipzig, Brüderstr. 34, 04013 Leipzig, Germany Abteilung, Steigerwald Arzneimittelwerk GmbH, Havelstr. 5, 64295 Darmstadt, Germany 2 Wissenschaftliche STW 5 (Iberogast) is a fixed combination of nine plant extracts. It is successfully used for treatment of functional dyspepsia and irritable bowel syndrome (IBS). In this study we compared the effects of STW 5 with the effects of its component Iberis amara (STW 6) and another, related combination, STW 5-II, which does contain six components, on tone and acetylcholine (ACh)-induced contractions in intact and inflamed intestinal preparations. We used 1-1.5 cm long ileum and colon preparations of male Wistar rats to study region specific differences. The inflammation was induced by intraluminal instillation of 2,4,6trinitrobenzene sulfonic acid (TNBS, 10 mM, 30 min). Incubation with STW 5 (512 µg/ml) reduced the tone and decreased ACh-induced contractions of untreated ileal and colonic preparations concentration dependently (64-512 µg/ml). The effects of STW 5-II in a concentration of 533.2 µg/ml were comparable to those of STW 5. STW 6 in equivalent concentrations (3-24.1 µg/ml) neither affected the tone nor the contractility. TNBS-induced inflammation was accompanied by a significant reduction of ACh-induced contractions. Co-incubation of TNBS with STW 5 (512µg/ml), STW 5-II (533.3 µg/ml) or STW 6 (24.1 µg/ml) partially normalized the TNBS-induced attenuation of tone as well as of ACh-induced contractions in ileum preparations. In inflamed colon segments the co-incubation of TNBS with STW 6 in a high concentration (24.1 µg/ml) revealed protective effects whereas STW 5 as well as STW 5-II had no effects. In conclusion, STW 5 and the related combination STW 5-II influenced ACh-induced contractions and tone in untreated ileal and colonic preparations, whereas their component STW 6 did not contribute to these effects. In TNBS-inflamed ileum preparations STW 5, STW 5-II as well as STW 6 normalized contractile disturbances, while in colon preparations only STW 6 was effective. Our study therefore allows the conclusion that a region specific action might contribute to the clinically proven effects in irritable bowel syndrome (IBS). POS.018 Antibiotics from predatory bacteria – from discovery to MOA studies Nett, M. 1; Schieferdecker, S. 1; König, S. 2; Korp, J. 1; Werz, O. 2 1 Leibniz Institute for Natural Product Research and Infection Biology/Hans-Knöll-Institute, Beutenbergstr. 11a, 07745 Jena, Germany 2 Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University, Philosophenweg 14, 07743 Jena, Germany For abstract see Short Poster Lecture SPL.001 on page 98. DPhG Annual Meeting 2015 Conference Book • 119 POSTERS POS.019 Natural substances as inhibitors of the tumor target human hyaluronidase hyal-1 Lengers, I.1; Orlando, Z.1; Melzig, M. F.3; Buschauer, A.4; Hensel, A.2; Jose, J.1 1 Institute of Pharmaceutical and Medicinal Chemistry of Pharmaceutical Biology and Phytochemistry, PharmaCampus, Westfälische Wilhelms-Universität, Corrensstraße 48, 48149 Münster, Germany 3 Institute of Pharmacy, Pharmaceutical Biology, Freie Universität Berlin, Königin Luise Str. 2+4, 14195 Berlin, Germany 4 Institute of Pharmacy, Department of Pharmaceutical/Medicinal Chemistry II, University of Regensburg, Universitätsstr. 31, 93040 Regensburg, Germany 2 Institute The negatively charged polysaccharide hyaluronic acid (HA) assembled from repeating disaccharide units of D-glucuronic acid and N-acetyl-D-glucosamine. Its chain size triggers different physiological and pathophysiological functions. Space filling, anti-inflammatory and antiangiogenic effects are caused by high molecular weight HA (>20 kDa). HA hydrolization by hyaluronidases leads to low molecular weight HA (<20 kDa), resulting in inflammatory and angiogenic effects [1]. The degradation of HA is mainly catalyzed by human hyaluronidase Hyal-1. It has been demonstrated that in prostate or bladder tumour cells the expression level of Hyal-1 was elevated [2,3]. For this reason Hyal-1 is an interesting target for drug discovery. The surface display of active Hyal-1 on Escherichia coli, via Autodisplay, enables the screening for potential inhibitors in a whole cell system. Based on this technique we determined the inhibitory effect of different plant extracts and triterpenoid saponins on human Hyal-1. The IC50 values of the extracts of Malvae sylvestris flos, Equiseti herba and Ononidis radix were determined to lay between 1.4 and 1.7 mg/mL. The obtained IC50 values for the triterpenoid saponins, glycyrrhizinic acid (reference inhibitor), gypsophila saponin 2, SA1641 and SA1657 were 177 µM, 108 µM, 296 µM and 371 µM, respectively. These natural substances identified can be used as a starting point for the synthesis of new small molecule inhibitors targeting human Hyal-1. References: 1. Stern, R.: Semin. Cancer Biol. 2008, 18: 275-280. 2. Lokeshwar, V.B, et al.: J. Urol. 2000, 163: 348-356. 3. Lokeshwar V.B, et al.: J. Biol. Chem. 2001, 276(15): 11922-11932. POS.020 Archazolid A influences endothelial processes responsible for tumor cell adhesion and transmigration Luong, B.1; Menche, D.2; Müller, R.3; Fürst, R.1 1 Institute of Pharmaceutical Biology, Biocenter, Goethe-University, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany 2 Kekulé Institute for Organic Chemistry and Biochemistry, University of Bonn, GerhardDomagk-Str. 1, 53121 Bonn, Germany 3 Helmholtz Institute for Pharmaceutical Research Saarland, Department Microbial Natural Products, Campus, C2.3, Saarland University, 66123 Saarbrücken, Germany Metastasis is the major cause of death in cancer patients. Archazolid A is a v-ATPase inhibitor of myxobacterial origin, which has shown antimigratory and cell-death inducing effects on tumor cells. In vivo, archazolid A reduced metastases formation [1,2]. A key step during the formation of secondary tumors is the adhesion of tumor cells onto endothelial cells and their subsequent extravasation through the endothelium into the underlying tissue. The aim of this study is to characterize how the interaction between tumor cells and endothelial cells is influenced by archazolid A. First, we investigated the impact of archazolid A on the viability of primary human umbilical vein endothelial cells (HUVECs). Concentrations up to 1 nM did neither significantly influence the metabolic activity (CTB assay) nor the apoptosis rate (flow cytometry) of HUVECs. Treatment of HUVECs with 1 nM archazolid A did not affect the confluency of the endothelial monolayer, but led to a swelling of the cells. Interestingly, the compound increased the adhesion of MDA-MB-231, a highly invasive breast cancer cell line, onto the endothelium and decreased the transmigration of these cancer cells through the endothelium (Boyden chamber assay). Several adhesion molecules and also the CXCR4/CXCL12 chemokine system have been postulated to be involved in the interaction of MDAs and endothelial cells. Quantitative 120 • DPhG Annual Meeting 2015 Conference Book real-time PCR revealed that the mRNA levels of the potentially involved adhesion molecules ICAM-1, VCAM-1, E-selectin, N-cadherin, and galectin-3 were not upregulated in HUVECs upon archazolid A treatment (1 nM) for 12 h. Also the chemokine system CXCR4/CXCL12 was not affected by archazolid A. Interestingly, archazolid A exerted a strong influence on adherens junctions, which are largely composed of VEcadherin. Immunofluorescent stainings as well as a cell surface protein isolation assay showed that archazolid A leads to a loss of VE-cadherin at the cell surface, while the total amount of cellular VE-cadherin is not affected. Despite this breakdown of adherens junctions, neither endothelial permeability (Transwell assay) nor endothelial contractility (Western blot) were influenced by archazolid A (1 nM). Taken together, the anti-metastatic effect of archazolid A seems to be a consequence of the increased adhesion of tumor cells onto the endothelium and a reduced transmigration through the endothelium. The precise underlying mechanisms are still to be resolved. However, there is no influence on molecules typically involved in tumor-endothelial cell interaction (adhesion molecules, CXCR4/CXCL12 system). We speculate that the effect might be associated with an impaired trafficking of interendothelial VE-cadherin junctions. Acknowledgments: This work was supported by the German Research Foundation (DFG, FOR 1406, FU 691/9-2). References: 1. Wiedmann, R. et al.: Cancer Res 2012, 72(22): 5976-87. 2. von Schwarzenberg, K. et al.: J. Biol. Chem. 2012, 288(2): 1385-96. POS.021 Soraphen A – targeting the membrane composition as an innovative approach to fight cancer Stoiber, K.1; Vollmar, A. M.1; Braig, S.1 1 LMU Munich, Department of Pharmacy, Pharmaceutical Biology, Munich, Germany The fatty acid metabolism is found to play a key role in oncogenic transformation. First metabolic investigations of human tumors revealed that malignant cells were characterized by an overexpression of lipogenic enzymes like the fatty acid synthase and acetyl-CoA carboxylase (ACC), resulting in a high rate of de novo fatty acid synthesis. We aim to characterize the impact of Soraphen A, an acetyl-CoA carboxylase inhibitor isolated from the myxobacterial strain Sorangium cellulosum, on cancer cells and biophysically disclose its mode of action. In order to elucidate the influence of ACC inhibition on the phospholipid composition of cancer cells, phospholipids of Soraphen A treated MDAMB-231 breast cancer cells were extracted and analyzed by liquid chromatography ESI tandem mass spectrometry. Interestingly, whereas the total amount of phospholipid content is not modulated, the composition of phospholipids is strongly altered by Soraphen A treatment. As phospholipids are the main components of cellular membranes, we analyzed whether changes in phospholipid composition induced by stimulation with Soraphen A affect their biophysical characteristics. We were able to demonstrate that Soraphen A strongly enhances the rigidity of cellular membranes by generating giant plasma membrane vesicles (GPMVs). In addition, inhibition of ACC with the myxobacterial compound impedes the deformability of whole cells as shown by optical stretcher experiments. Next, to investigate whether Soraphen A induced alterations in the biophysical properties of cancer cells have a functional impact on metastasis, two highly metastatic cancer cell lines were treated for 2 hours with the acetyl-CoA inhibitor and migration and invasion assays towards FCS and EGF as chemoattractant were performed. Soraphen A dose-dependently abrogates migratory and invasive potential of MDAMB-231 breast cancer and T24 bladder carcinoma cells. By using Soraphen A as a chemical tool to target the rate-limiting step of the fatty acid synthesis, we are able to characterize the impact of modulated membrane properties in cancer progression. Furthermore, Soraphen A as a therapeutic lead substance might promote the development of new cancer targeting drugs affecting the tumorsupportive cellular machinery of membrane features and lipogenesis. NATURAL COMPOUNDS POS.022 Crataegus ssp. promotes late-stage cardiac differentiation and regeneration Halver, J.1; Carrillo García, C.1; Willems, E.2; Schade, D.1 Department of Chemistry & Chemical Biology, TU Dortmund, Otto-Hahn-Str. 6, D-44227 Dortmund, Germany Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA92037, USA 1 2 Background: The minimal and clearly insufficient ability of the adult heart to regenerate after ischemic injury is a great appeal for identifying biological mechanisms, substances and factors that improve this process [1]. Two main sources for cardiomyocyte renewal and regeneration have emerged in the field: a) Adult multipotent progenitor cells and b) preexisting cardiomyocytes [2]. Objective: Based on the many positive effects on the myocardium after infarction and the overall cardiovascular protective activity of Crataegus ssp. (extract WS®1442) [3], we aimed at studying whether also mechanisms of cardiac differentiation and regeneration could possibly play a role. Results: Here, we show that WS®1442 efficiently stimulated cardiomyocyte differentiation from murine and human ESCs in a dosedependent manner after mesoderm was formed. This activity was thoroughly validated in a mESC-based (CGR8-Myh6-GFP) spontaneous differentiation assay. First bioassay-guided fractionations of the extract suggested that this activity is reserved for specific compound classes. Conclusions: According to the observed activity profile, we hypothesize that the identified active fractions in WS®1442 could possibly target multipotent progenitors, stimulate their differentiation towards the cardiac lineage but also expand their pool (proliferation). Further elucidation of the underlying cellular and molecular mechanisms might lead to novel targets that can be exploited for ex vivo expansion of cardiac progenitor cells. Eventually, it will be interesting to see whether (and how) our in vitro findings translate to in vivo regeneration. References: 1. Schade, D.; Plowright A. T.: J. Med. Chem. 2015, in press. 2. Harvey, R. P.; Graham R. M.; Pu, W. T.: Stem Cell Res. 2014, 13: 521-714. 3. Koch, E.; Malek, F. A.: Planta Med. 2011, 77(11): 1123-1128. DPhG Annual Meeting 2015 Conference Book • 121 POSTERS 4.4 Pharmaceutical Technology and drug formulations POS.023 In vitro model of infected stratum corneum for the efficacy evaluation of novel formulations of antifungal ciclopirox olamine as well as differential scanning calorimetry and stability studies Täuber, A.1; Müller-Goymann, C. C.1 1 Institut für Pharmazeutische Technologie, Technische Universität Braunschweig, Mendelssohnstraße 1, 38106 Braunschweig, Germany Introduction: Superficial fungal infections are a common disease and affect 20-25% of the world’s population [1]. Mostly, the infections are caused by dermatophyte fungi with Trichophyton rubrum being the main trigger. In this contribution, the antifungal agent ciclopirox olamine (CPX) was incorporated into a variety of poloxamer 407-based formulations. These novel compositions consisted of poloxamer 407 (P407), double distilled water, propylene glycol (PG), isopropyl alcohol (IPA) and medium chain triglycerides (MCT) in given ratios. Antifungal efficacy evaluation against T. rubrum was analysed in a novel in vitro model of infected stratum corneum (SC). Moreover, stability and differential scanning calorimetry (DSC) studies were carried out to analyse the influence of the formulations on the SC. Methods: The P407-based formulations were automatically stirred at 1440 rpm for 1.5 min with an Unguator® e/s (GAKO Konietzko GmbH, Bamberg, Germany). Subsequent storage was done for 24 h at 20 ± 1 °C to ensure sufficient equilibration. All the formulations were given codes reflecting their quantitative composition, e.g. 1P1050 represented a formulation loaded with 1% CPX, while the vehicle itself contained 10% P407/MCT (4:1), 50% IPA/PG (1:1) and 40% double distilled water (all w/w). In vitro studies of infected SC were performed according to Lusiana et al. [2] with slight modifications. In the present contribution, isolated human SC was used instead of bovine hoof plates and keratin films, respectively. SC was hydrated in autoclaved, double distilled water. With a polycarbonate filter as backing, approximately 5 SC were placed in a potato glucose agar. DSC studies were carried out according to Lusiana and Müller-Goymann [3] with one modification. Instead of 37 °C in the previous publication, equilibration and incubation was done at 32 °C. For stability studies, samples were stored in air-tight, sealed vials at 30 °C under light exclusion. After 1, 3, 6 and 12 months, CPX contents were determined with high performance liquid chromatography (HPLC). Approximately 50 mg of the formulation were weighed in a volumetric flask and dissolved with mobile phase under vortexing (Vortex-Genie® 2, Scientific Industries, Inc., New York, United States). 20 µL of each mixture was injected into the HPLC column. Results: Regarding the infected SC studies, a marketed semi-solid formulation (Selergo® 1% cream) showed complete fungal growth (= highest score: 10) after 6 days of incubation. Liquid P407-based formulations e.g. 1P1050 as well as a marketed nail lacquer (Ciclopoli®) indicated scores of 0 (= no fungal growth). The semi-solid P407-based formulations achieved scores of 6-8. 4P4030 was the only P407-based formulation tested with incorporation of 4% CPX. It showed only slight fungal growth with a score of 0.5. The scores of all tested formulations were significantly lower than that of Selergo® 1% cream (p < 0.05). The DSC measurements gave precise information on the interaction between applied formulation and SC. Regarding the excipients, water and PG had only slight impact on the SC, whereas MCT highly influenced the SC indicated as transition shifts. IPA showed no peak transition shift. Selergo® 1% cream had slightly influenced the transitions similar to 1P1020 and 1P1030. Increasing IPA/PG concentrations from 1P1020 to 1P1070_21 resulted in higher transition shifts. Furthermore, higher incorporated CPX amounts such as in 1-4P4030 augmented the transition shifts. After 12 months of storage, almost all P407-based formulations still indicated CPX contents ≥ 95%. Several liquid P407-based formulations exhibited phase separation. Moreover, slight yellow discolouration occurred. Conclusion: An in vitro model of infected SC has successfully been developed for pretesting novel antifungal formulations. Several liquid P407-based formulations showed superior T. rubrum growth inhibition in comparison to a marketed semi-solid reference. DSC studies indicated rising influence on the SC with increasing CPX- as well as IPA/PG 122 • DPhG Annual Meeting 2015 Conference Book content. One year stability studies exhibited CPX contents ≥ 95% for the majority of the P407-based formulations. References: 1. Havlickova, B.; Czaika, V.A; Friedrich, M.: Mycoses 2008, 51 (Suppl. 4): 2–15. 2. Lusiana; Reichl, S.; Müller-Goymann, C.C.: Eur. J. Pharm. Biopharm. 2013, 84 (3): 599–605. 3. Lusiana; Müller-Goymann, C.C.: Am. Ass. Pharm. Sci. 2011, 12 (2): 496-506. POS.024 “Nanoparticle Tracking Analysis of Size and Concentration detection in Particle Suspensions: Influence of experimental and data evaluation parameters” Gross, J.1; Karow, A. R.2; Sayle, S.2; Bakowsky, U.1; Garidel, P.2 Department of Pharmaceutics and Biopharmacy, Faculty of Pharmacy, 1 Philipps-University Marburg, Ketzerbach 63, 35032 Marburg, Germany 2 Boehringer Ingelheim Pharma GmbH & Co.KG, Global Bioprocess & Pharmaceutical Development, Birkendorfer Straße 65, 88397 Biberach (Riss), Germany 1 Nanoparticle Tracking Analysis (NTA) is an emerging technique for detecting particle size distributions and particle concentrations in biologics. This study deals with the performance evaluation for the detection and characterisation of various particles by NTA. Our investigation focusses on the NTA measurement parameter set-ups. In order to achieve this, we used polystyrene standard particles (monodisperse and polydisperse) as well as protein particles. On one site, we showed the highly precise and reproducible detection of particle size and concentration in monodisperse polystyrene particle systems, under specified and constant parameter settings. On the other hand, our results exemplify potential risks and errors while setting parameters with regards to the results and thus interpretation thereof. Especially changes of the parameters camera level and detection threshold led to significant changes in the determined particle concentrations. We defined specified “optimal” camera levels for monodisperse particle suspension characterizations in the size range of 20 to 1000 nm. We illustrated that the results of polydisperse polystyrene standard particle solution measurements highly depend on the used parameter settings. Changes in the settings led to the appearance or disappearance of “peaks” for polydisperse systems. For the use of NTA in biopharmaceutical analysis proteinaceous samples were investigated. We analysed protein particle suspensions and compared unstressed and stressed protein samples similar to polysterene particle measurements. We also measured these samples in two measuring modes (general capture mode and live monitoring mode) that the commercially available analysis tool is offering. Our results stated the live monitoring mode as more suitable for protein samples, as the results were more reproducible and less operator-depending. In conclusion, NTA is a potential technique and unique in quantitative evaluation of particle suspensions in the subvisible size range, especially for monodisperse suspensions. We strongly urge on not underestimating the influence of the operator, i.e. setting the measuring parameters on the obtained results. From an industry perspective the documentation of the parameter settings for each experiment is crucial for data evaluation, data interpretation and for data comparison. Acknowledgments: Boehringer Ingelheim Pharma GmbH & Co.KG, Michaela Blech POS.025 The glass transition temperature as an underestimated parameter for drug release properties of polymer based nanoparticles Lappe, S.1; Langer, K.1 1 Institute of Pharmaceutical Technology and Biopharmacy, University of Muenster, Corrensstraße 48, 48149 Münster, Germany Polymeric nanoparticles are widely used in pharmaceutical research acting as drug delivery systems. Great efforts were made to understand the release behaviour of drug loaded nanoparticular delivery systems [1]. PHARMACEUTICAL TECHNOLOGY AND DRUG FORMULATIONS Nevertheless the subject of the glass transition temperature (Tg), a well defined parameter for the starting material of nanoparticle preparation, is underestimated when finished drug delivery systems are characterised. Therefore the aim of this study was to take a more detailed look on the correlation between Tg and the drug release behaviour of flurbiprofen from poly(D,L-lactic-co-glycolic acid) (PLGA), poly(L-lactic acid) (PLA) and poly(butyl methacrylate-co-(2-dimethylaminoethyl) methacrylate-comethyl methacrylate) (Eudragit® E) nanoparticles. Nanoparticle preparation was carried out by an emulsion diffusion method. For the determination of Tg a differential scanning calorimetry (DSC) method was used. The release kinetics were conducted in phosphate buffer pH 8.0 or bovine serum albumin solution (5% w/v) at different temperatures for 48 h. In comparison to unloaded PLGA and PLA nanoparticles the Tg of flurbiprofen loaded nanoparticles was lowered. Flurbiprofen forms a solid solution with the polymer during nanoparticle preparation leading to a shift in Tg acting as a plasticiser within the polymer matrix [2,3]. Tg of unloaded Eudragit® E nanoparticles and the flurbiprofen loaded system were nearly identical suggesting less interactions between drug and polymer chains. Drug release kinetics were analysed at temperatures below and above Tg of the respective nanoparticle system. For the flurbiprofen loaded PLGA and PLA nanoparticles a reduced release could be observed when the temperature of the release medium was kept below Tg. An instant release of almost the entire encapsulated drug was determined increasing the temperature above Tg [4]. Regarding the flurbiprofen loaded Eudragit® E nanoparticles a temperature independent burst release was observed suggesting that the major part of the drug was not incorporated into the polymeric matrix but was rather adsorbed to the large nanoparticle surface. The present study shows the Tg dependent release behaviour of drug loaded polymeric nanoparticles. A Tg higher than the temperature of the release medium leads to a reduced burst release of drug substance. However this assumption is not applicable to any drug-polymer combination since the type of drug loading is also important. Therefore the parameter Tg needs to be considered in nanoparticle characterisation during the development of new drug delivery systems. References: 1. Sant, S. et al.: J. Control. Release 2005, 107(2): 203-214. 2. Pamujula, S. et al.: J. Pharm. Pharmacol. 2004, 56(9): 1119-1125. 3. Blasi, P. et al.: AAPS PharmSciTech 2007, 8(2): Article 37. 4. Faisant, N. et al.: Int. J. Pharm. 2006, 314(2): 189-197. POS.026 Nano modified antioxidants for the prevention and treatment neurodegenerative diseases Braun, A.1; Law, J. K. Y.1; Scholz, P.2; Keck, C. M.2; Ingebrandt, S.1; Schaefer, K. H.1 1 Department of Informatics and Microsystem Technology, University of Applied Sciences, Zweibrücken, 66482, Germany 2 Department of Applied Logistics and Polymer Sciences, University of Applied Sciences, Pirmasen, 66482, Germany Neurodegenerative diseases, including Alzheimer‘s and Parkinson‘s diseases, are commonly occurring diseases for elderly. Oxidative stress due to amyloid β (Alzheimer's disease) or α-synuclein (Parkinson‘s diseases) leads to neuronal death [1]. The symptoms of this diseases usually take years to develop, unfortunately, there is no effective solution for the diseases by far. An early stage of identification and prevention of such diseases will be beneficial. However, it is challenging to obtain biopsy from the central nervous system (CNS) over time. Therefore the aim of this project is to develop an effective nutritional supplement for preventing neurodegenerative diseases. Herbal substances have high antioxidant contents and studies have shown promising neuroprotective potential [2]. Moreover, most of these natural substances have low solubility in water and consequently poor absorption in human body. In order to enhance the solubility, nanocrystals of these antioxidants are produced [3]. To study the effect of nano antioxidants on the prevention of the neurodegenerative diseases, cell culture experiments were performed. Different kind of nano and micro particles (rutin, hesperidin, β-carotene) were incubate together with α-synuclein. In the DPPH radical scavenger assay the nano particles showed much higher antioxidant capacity than the micro particles. In vito experiments demonstrated that the nanomodified particles have a higher impact on neuronal survival in pathological condition in comparison to the micro particles. Acknowledgments: This work is supported by the Federal German Ministry of Education and Research: “IngenieurNachwuchs2013: Nanomodifizierte Antioxidantien zur Prävention und Behandlung neurodegenerativer Erkrankungen”. The authors would like to thank Mr. Rainer Lilischkis for his support for the SEM imaging. References: 1. Wei, X. et al.: Mol. Cell. Neurosci. 2013, 54: 71-83. 2. Kumiko, I. et al.: Free Radic. Biol. Med. 2001, 30(4). 3. Rachmat, M. et al.: Eur. J. Pharm. Sci. 2009, 36: 502–510. POS.027 A Slow-Release System of Bacterial Nanocellulose for Octenidine as Wound Dressing Alkhatib, Y.1; Dewaldt, M.1; Moritz, S.1; Kralisch, D.1,2; Fischer, D.1 1 Department of Pharmaceutical Technology, Friedrich-Schiller-University, Jena, 07745, Germany 2 JeNaCell, Jena, 07745, Germany The biopolymer bacterial nanocellulose (BNC) has raised a substantial interest in medical and pharmaceutical research and development due to its outstanding physicochemical and biological characteristics. Although BNC has an identical chemical formula like plant cellulose, its unique structure consisting of a three-dimensional network of nanosized fibres, a high purity, excellent biocompatibility, and unique mechanical stability provide an excellent basis as a biomaterial for artificial blood vessels, scaffolds for tissue engineering and wound dressings. Octenidine was introduced for skin, mucous membrane, and wound antisepsis more than two decades ago [1]. The antiseptic exhibits a broad antimicrobial activity against Gram-positive and Gram-negative, plaque-forming bacteria, and fungi. In this study, BNC fleeces were functionalized with octenidine to develop an active wound dressing for infected wounds over one week. Bacterial nanocellulose fleeces were prepared by strains of Komagataeibacter xylinus (DSM 14666) in Hestrin-Schramm medium under static conditions in a 24-well plate, harvested and purified in alkaline solution [2]. After autoclaving, BNC fleeces, were loaded with octenidine solution (0.5%, Schülke & Mayr) with and without the addition of the Poloxamers P338 and P407 (BASF SE) in different concentrations under shaking (70 rpm) for 48 hours. Release studies were performed in PBS buffer at pH 7.4 for up to 192 h at 32 °C using the Franz cell diffusion system (SES GmbH). Octenidine was quantified via UV/Vis spectrophotometry at 281 nm. Size and zeta potential were measured using a Zetasizer Nano ZS (Malvern Instruments). Compression and tensile tests as well as water absorption and water retention values of the BNC fleeces were determined. Antimicrobial efficacy against S. aureus and P. aeruginosa was investigated by an agar diffusion test and fluorescence staining method using the LIVE/DEAD® Bacterial Viability Kit (Molecular Probes). The biocompatibility of the BNC was tested on an ex ovo hen’s egg model. BNC fleeces were successfully loaded by the post synthesis technique. The addition of 5% Poloxamer P407 or 10% Poloxamer P338 induced the formation of octenidine loaded micelles (approx. 2-5 nm, zeta potential -0.8 to 4.3 mV) that retarded the release of octenidine by 2629% in comparison to the release profile of octenidine loaded in BNC without Poloxamers. The increase of the concentration of Poloxamer P407 to 18.5% or Poloxamer P338 to 22% resulted in an octenidine containing thermoreversible hydropolymer gel. The gel formation in the BNC slowed down the drug release with a continuous profile over up to 192 h. Whereas for octenidine loaded BNC no changes of the mechanical characteristics could be observed, the incorporation of the Poloxamers increased the compression stability and decreased the water absorption and retention values. As demonstrated in agar diffusion and LIVE/DEAD® tests, octenidine loaded BNC with and without Poloxamers demonstrated high antimicrobial activity against S. aureus and P. aeruginosa. DPhG Annual Meeting 2015 Conference Book • 123 POSTERS In conclusion, bacterial nanocellulose could be loaded effectively with octenidine. In the presence of different types of Poloxamers in various concentrations a controlled release over up to one week could be achieved. Therefore, this system could be used as a wound dressing with sustained drug release. Acknowledgments: The authors would like to thank R. Brabetz and E. Pfaff for their excellent technical assistance. We acknowledge the Thuringian Ministry of Education, Science and Culture, the EFRE (B714-10032) and the FAZIT Foundation, Gemeinnützige Verlagsgesellschaft mbH (S. M.) for financial support. References: 1. Hübner, N.O. et al.: Skin Pharmacol. Physiol. 2010, 23(5): 244-258. 2. Moritz, S. et al.: Int. J. Pharm. 2014, 471(1–2): 45-55. Figure: SEM image of fleece structure (A), TEM image of nanowhiskers (B) Acknowledgments: We would like to thank the Deutsche Forschungsgemeinschaft DFG (FI 899/2-1) for funding of the project. Additionally, F.A. Müller and F. Wesarg (IMT, FSU Jena) are acknowledged for SEM analysis and E. Pfaff and J. Thamm for technical assistance. References: POS.028 1. Moritz, S. et al.: Int. J. Pharm. 2014, 471(1–2): 45–55. 2. Müller, A. et al.: J. Pharm. Sci. 2013, 102(2): 579–592. Bacterial Nanocellulose as a Drug Delivery System for Nucleic Acids Pötzinger, Y.1; Rabel, M.1; Ahrem, H.1; Rahnfeld, L.1; Kralisch, D.1,2; Klemm, D.3; Fischer, D.1 1 Department of Pharmaceutical Technology, Friedrich-Schiller-University, Otto-Schott-Straße 41, 07745 Jena, Germany 2 JeNaCell GmbH, Winzerlaer Str. 2, 07745 Jena, Germany 3 Polymet Jena e.V., Wildenbruchstraße 15, 07745 Jena, Germany In the steadily emerging field of natural and renewable materials, the biopolymer bacterial nanocellulose (BNC) has attracted increasing importance due to its outstanding physicochemical and biological properties. BNC is characterized by its high purity, excellent biocompatibility, and unique mechanical stability favouring the application as tissue scaffold or wound dressing. Beside this, the nanosized three-dimensional network (Figure A) offers a huge interface for the local delivery of antiseptic drugs [1] or larger molecules like proteins [2]. By acidic hydrolysis, it is also possible to disintegrate the native BNC fibres in order to generate high-crystalline needle-like nanowhiskers (Figure B). The anionic surface of these nanowhiskers allows ionic interactions with charged molecules and opens up a new field for drug delivery. In the present study the preparation of non-viral carrier systems for gene therapy based on BNC fleeces on the one hand and on nanowhiskers on the other was investigated for the first time. BNC fleeces were produced by strains of Komagataeibacter xylinus (DSM 14666, DSMZ, Braunschweig) in static culture, harvested and purified. Resulting nanocellulose hydropolymers were used as neverdried (nd) or freeze-dried (fd) BNC and loaded with nucleic acids using three different strategies. Loading of nd-BNC with an injection technique or fd-BNC by reswelling increased the amount of loadable material and the ease and speed of loading in comparison to a conventional adsorption technique [2]. In vitro release studies were performed under agitated conditions. The cumulative release of adsorption loaded fleeces was highly varying and not reproducible. In contrast, a prolonged release exhibited a biphasic release profile with a rapid release in the initial 24 hours followed by a slower release rate up to 200h could be accomplished with fd-BNC loaded by reswelling. Injection loaded ndBNC showed an even more extended release profile represented by an almost linear curve. For the preparation of nanowhiskers nd-BNC was disintegrated and hydrolysed with sulfuric acid, purified and fractionated. By modifying the hydrolysis parameters it is possible to prepare nanowhiskers with a controllable size and surface charge, low polydispersity and high purity. No cytotoxicity of the nanowhiskers was observed in vitro up to a concentration of 1 mg/mL and 72 h incubation time using L929 mouse fibroblasts. Hemocompatibility was demonstrated quantitatively and qualitatively by testing hemoaggregation and hemolysis up to a concentration of 1 mg/mL. Additionally, no toxic effects of the particles towards the vascular system of developing chick embryos could be detected using a shell-less hen’s egg model, neither after topical nor after systemic administration. An effective DNA loading of the nanowhiskers was facilitated by modifying the surface with poly(ethylene imine) using a layer-by-layer technique which could be demonstrated by measuring size and zeta-potential, performing agarose gel electrophoresis and DNA quantification using AccuBlueTM. In conclusion, the results confirmed the suitability of BNC for gene delivery applications. Both three-dimensional BNC fleeces and BNC based nanoparticles are highly biocompatible and could be loaded effectively with nucleic acids. Depending on loading techniques, release rates could be controlled and release profiles selectively adjusted. 124 • DPhG Annual Meeting 2015 Conference Book POS.029 Impact of cationic coatings on PLGA nanoparticle characteristics and cellular uptake Kunschke, N.1; Loretz, B.1; Lehr, C. M.1,2 1 Helmholtz-Institute for Pharmaceutical Research Saarland, Department of Drug Delivery, Saarland University Saarbrücken, 66123, Germany 2 Biopharmaceutics and Pharmaceutical Technology, Department of Pharmacy, Saarland University Saarbrücken, 66123, Germany The delivery of therapeutic nucleic acids into the deeper lung implies certain challenges: to reach the alveolar region, to overcome the noncellular barrier, to avoid the phagocyte clearance and to deliver the cargo into the cytoplasm to its target. At the same time the drug delivery system (DDS) has to be non-toxic, biodegradable and biocompatible [1]. A combined system of PLGA and a cationic coating reduces the required total amount of cationic material [2], which is leading to a lower cytotoxicity of the DDS and remains the ability to bind siRNA on the particle surface. The cationic polymers were selected by differences in molecular weight and number of primary amine groups which is significant for transfection efficiency. This may surely influence the entire particulate system and their ability to enter cells. Particle preparation was performed by a modification of the emulsiondiffusion-evaporation technique [3]. The model siRNA (21 nucleotides), provided from GlaxoSmithKline (UK), was loaded on the nanoparticles via electrostatic attraction. Protamine-, Chitosan- and CatStarch [4]coated PLGA nanoparticles were characterized by Dynamic light scattering (DLS) and showed a mean diameter from 125 nm to 170 nm. This value is depending on the MW of the used coating material. With a higher the MW, the nanoparticles offered an increased size. A positive ζpotential from + 15 mV to + 40 mV supported the assumption of an outer cationic polymer shell. The monodispersity (DLS: polydispersity index (PdI) lower 0.200) and spherical shape of the nanoparticles was confirmed via scanning electron microscopy (SEM). The colloidal stability could be indicated for at least 21 days at storage conditions (4 °C). Chitosan-PLGA NPs and CatStarch NPs showed an acceptable viability (MTT assay) on Calu-3 and A549 cells (> 80%). Protamine-PLGA NPs could reach at most 60% viability, after 4 h incubation of particle concentrations lower than 250 mg/ml. Differences in cellular uptake and localization after a 24h incubation period could be visualized via confocal laser scanning microscopy (CLSM). It was shown that a variation of the cationic coating of PLGA nanoparticles can easily be performed via the emulsion-diffusionevaporation technique. And the resulting particulate systems represent a promising approach in gene delivery to the lung. Acknowledgments: COMPACT (Collaboration on the Optimization of Macromolecular Pharmaceutical Access to Cellular Targets) References: 1. Kumar, R. et al.: Biomaterials 2004, 25(10): 1771-7. 2. Naffee, N. et al.: Nanomedicine 2007, 3(3): 173-183. 3. Beisner, J. et al.: Lung Cancer 2010, 68(3): 346-54. 4. Yamada, H. et al.: Biomacromolecules 2014, 15(5): 1753-1761. PHARMACEUTICAL TECHNOLOGY AND DRUG FORMULATIONS POS.030 Protein Corona Formation on Polymeric Nanoparticles and its Impact on Cellular Uptake, Cytotoxicity, and Activation of Endothelial, Epithelial, or Immune Cells Obst, K.1; Charbaji, N.1; Miceli, E.2; Dimde, M.2; Balzus, B.3; Bodmeier, R.3; Calderón, M.2; Haag, R.2; Hedtrich, S.1 Institute for Pharmaceutical Sciences, Freie Universität, Königin-Luise-Str. 2-4, 14195 Berlin, Germany 2 Institute of Chemistry and Biochemistry, Freie Universität, Takustr. 3, 14195 Berlin, Germany 3 Institute for Pharmaceutical Sciences, Freie Universität, Kelchstr. 31, 12169 Berlin, Germany 1 Nanoparticles have gained great significance in different medical applications such as drug delivery, diagnostics or therapy. Nanomaterials, however, undergo a substantial change as soon as they come in contact with biological fluids such as human blood which leads to the formation of a protein corona that covers the particle. About 1,000 different proteins are present in blood plasma which readily interact with the nanoparticles’ surface depending on the particles’ size, surface charge and composition and which gives the nanomaterial a new biological identity [1-3]. Comprehensive and systematic studies on protein corona formation, however, and the impact on interactions between nanoparticle and biological environment are rather scarce. Hence, in this project, we systematically evaluated the protein corona formation (identity and quantity) and subsequently studied its impact on cellular uptake, cytotoxicity, and activation of endothelial, epithelial or immune cells. Therefore, nanoparticles (Eudragit RS nanocarriers, ethyl cellulose nanocarriers, core-multishell (CMS) nanotransporters, dendritic polyglycerol nanogels, and polyglycerol nanogel with 10%, 30% and 90% amination) differing greatly in composition, particle size polarity and surface charge were prepared and subsequently analysed. Initially, the particle size and zeta potential were assessed. Afterwards, the nanoparticles were incubated with human plasma for 1 h or 24 h and afterwards purified with a 0.7 M sucrose cushion and washing steps. Next, the isolated protein corona was subjected to SDS-Page. Subsequently, to identify the attached proteins, clear protein bands were cut out, the gel was digested using trypsin and the samples were ultimately analysed by mass spectrometry. For protein quantification, a bicinchoninic acid assay was performed. To study the effects of the protein corona on biological systems, we used primary macrophages, keratinocytes and human umbilical vein endothelial cells (HUVEC) to investigate cell viability, induction of apoptosis and necrosis, release of pro-inflammatory cytokines, cellular uptake and the respective uptake mechanism. The nanoparticles’ size ranged from 30 nm to 200 nm and zeta potential was between -24 mV and +30 mV. 1 h incubation with blood plasma already initiated protein corona formation. Incubation for 24 h did not change the type of attached proteins but significantly increased the amount. Hereby, most pronounced protein corona formation was observed with Eudragit RS nanoparticles, whereas with ethyl cellulose, CMS and nanogels only one dominant adsorbed protein such as apolipoprotein A-I for ethyl cellulose or serum albumin for CMS and nanogels was identified. About 10 to 15 proteins adsorbed on Eudragit RS nanoparticles ranging from 16 to 215 kDa including several apolipoproteins, inter-alpha-trypsin inhibitors and complement factors. In terms of cytotoxicity, overall protein corona-coated nanoparticles showed reduced toxicity and the cellular uptake was slightly diminished. Preliminary results indicated no change in the cellular uptake mechanism. In conclusion, our data suggest that proteins strongly adsorb on cationic Eudragit RS nanoparticles and significantly less protein corona is formed on ethyl cellulose and polyglycerol based nanomaterial which might be advantageous in terms of drug delivery. Moreover, the results indicate that protein corona formation leads to reduced cytotoxicity, which might be due to a reduced cellular uptake. Acknowledgments: This work was supported by a grant from the Helmholtz Virtual Institute. We thank Dr. Christoph Weise (Institute of Chemistry and Biochemistry, Freie Universität Berlin) for his help with the mass spectrometry analyses. References: 1. Rahman et al.: Biophysics (Springer Series) 2013, 15. 2. Tenzer et al.: Nature Nanotechnology 2013, 8: 772-781. 3. Docter et al.: Nat. Protoc. 2014, 9(9): 2030-2044. POS.031 Targeting Excipients for Individual Radiation Therapy of Cancer – Surface Modification of PLGA Polymer and Liposome Nanoparticles entrapping Lanthanides by Cholesterol- and PLAbound Ligands Nawroth, T. 1; Krebs, L. 1; Johnson, R. 1; Langguth, P. 1; Hellmann, N. 2; Decker, H. 2; Schmidberger, H. 3; Goerigk, G. 4; Boesecke, P. 5a; Le Duc, G. 5b; Bravin, A. 5c; Schweins, R. 6 Pharmacy & Biochemistry Institute, Pharmaceutical Technology, Johannes Gutenberg University, Staudingerweg, D-55099 Mainz, Germany 2 Molecular Biophysics Institute, Johannes Gutenberg University, Jakob Welder Weg 26, D55128 Mainz, Germany 3 Department of Radiooncology and Radiotherapy (Clinics), University Medical Center; Langenbeckstr.1, D-55131 Mainz, Germany 4 HZB, Institute of Soft Matter and Functional Materials , BESSY Synchrotron, ASAXS, D-14109 Berlin, Germany 5 ESRF, European Synchrotron Radiation Facility, a)ID01, b)BioMedical Facility,c)ID17, 71 Avenue des Martyrs, F-38043 Grenoble, France 6 ILL, Institut Laue Langevin, DS / LSS, 71 Avenue des Martyrs, F-38042 Grenoble CEDEX 9, France 1 For abstract see Short Poster Lecture SPL.002 on page 99. POS.032 Perfluorocarbon-Nanoemulsions: Evolution of a marker agent for in vivo 19F Magnetic Resonance Imaging Grapentin, C.1; Krämer, W.1; Temme, S.2; Flögel, U.2; Schrader, J.2; Wang, X.3; Peter, K.3; Schubert, R.1 Department of Pharmaceutical Technology and Biopharmacy, University of Freiburg, Hermann Herder Str. 9, 79104 Freiburg, Germany 2 Department of Cardiovascular Physiology, University of Düsseldorf, Universitätsstr. 1, 40225 Düsseldorf, Germany 3 Baker IDI Heart and Diabetes Institute, 75 Commercial Road, Melbourne, Australia 1 Perfluorocarbon nanoemulsions (PFC-NE) are disperse systems consisting of nanoscale liquid perfluorocarbon droplets stabilized by an emulsifier, usually phospholipids. Perfluorocarbons are chemically inert and non-toxic substances that are exhaled after in vivo administration. The manufacture of PFC-NE requires high energy input, suitable techniques being high pressure homogenization or microfluidization. Originally investigated as oxygen carriers for cases of severe blood loss, the application of PFC-NE nowadays is more focused on using them as marker agents in 19F Magnetic Resonance Imaging (19F MRI). 19F is scarce in organisms and thus PFC-NE are a promising tool for highly specific and non-invasive imaging via 19F MRI [1]. Combining 19F images with classic 1H images, the fluorine signal is set into an anatomic context. One field of application utilizes the capability of neutrophils, macrophages and monocytes to phagocytize PFC-NE and the subsequent migration of these innate immune cells to inflamed tissues. This technique has proven practicability in the unambiguous visualization of inflammatory foci in numerous disease models in mice [2]. Recently, the successful imaging of myocardial infarction in mini pigs has been reported [3]. The translation to clinical trials in human seems potential, but requires the development of a stable nanoemulsion whose droplet size is well characterized over a long storage time. In a study, we used dynamic light scattering in comparison to a second method, analysis of transmission electron microscopy images of cryo-fixed samples, to evaluate stability of PFC-NE [4]. Besides a passive targeting approach towards inflammatory tissue, site specific delivery of PFC-NE to other pathological foci unaffected by phagocytic immune cells is of interest. Combining 19F MRI with site specific targeting of PFC-NE, pathological sites might be visualized specifically before alterations are observed in classic 1H MRI. Tissue specific targeting needs the decoration of the nanoemulsion with a homing ligand, e.g. a peptide or antibody, whose antigen is overexpressed in the target tissue. One approach for surface modification of PFC-NE utilizes cholesterolPEGs bound to tissue specific peptides. The cholesterol moiety inserts into the phospholipid layer of preformed droplets, allowing the preparation of highly specific marker agents in small scales principally on demand. This post insertion technique has recently proven feasibility in the visualization of thrombi using PFC-NE equipped with an α2antiplasmin-based peptide in mice [5]. Small, robust homing ligands like RGD-peptide (bound to a phospholipid anchor) can be incorporated into NEs during the manufacturing process. DPhG Annual Meeting 2015 Conference Book • 125 POSTERS For larger ligands of proteinaceous nature, such as (single chain) antibodies, high shear forces and elevated temperatures might lead to a degradation of the protein. A platform technology of PFC-NE bearing a reactive group for covalent coupling of a targeting ligand is highly desirable. Such nanoemulsions can be manufactured via microfluidization using maleimide-PEG2000-phospholipids in a mixture with standard phospholipids. Our results show that the reactive maleimide groups stay intact during microfluidization and that ligands containing a thiol group can be successfully coupled. In an in vitro attempt, such PFC-NEs are directed to activated platelets via a singlechain variable fragment binding to glycoprotein IIb/IIIa [6]. Ongoing research focuses on the advancement of this platform technique, decorating PFC-NE with ligands of different sizes, and thus facilitating site specific targeting in a variety of disease models both in vitro and in vivo. Acknowledgments: The authors gratefully emphasize financial support by Deutsche Forschungsgemeinschaft (DFG). References: 1. Temme, S. et al.: WIREs Nanomed. Nanobiotechnol. 2012, 4: 329-343. 2. Floegel, U. et al.: Circulation 2008, 118: 140-148. 3. Boenner, F. et al.: Eur. Heart J. – Cardiovasc. Imaging 2015, 16: 612-620. 4. Grapentin, C. et al.: PLoS ONE 2015, 10(6): e0130674. 5. Temme, S. et al.: Circulation 2015, 131. 1405-1414. 6. Wang, X. et al.: Circulation 2012, 125: 3117-3126. POS.033 Entrapment of doxorubicin in different nanoparticle systems – An overview Pieper, S.1; Langer, K.1 Institute of Pharmaceutical Technology and Biopharmacy, Corrensstr. 48, 48149 Münster, Germany 1 Doxorubicin-loaded nanoparticles (DOX-NPs) are a promising approach regarding the therapy of carcinosis. Reported advantages of DOX-NPs over traditional medication are an improved drug targeting due to EPReffect (enhanced permeability and retention effect), a lower rate of side effects and good prospects to overcome multi drug resistance in cancer cells [1]. However, there are several different nanoparticle preparation techniques and starting materials, which can all influence essential properties of the nanoparticles in therapy. In order to come up with the most promising nanoparticle formulation, different materials and techniques of nanoparticle preparation have been analysed in terms of their physicochemical properties and their ability to entrap doxorubicin. The particle size and polydispersity were measured by photon correlation spectroscopy (PCS), while the amount of entrapped doxorubicin was calculated via an indirect quantification method. Therefore, the nanoparticles were centrifuged, the supernatants were collected and analysed via HPLC using a reversed phase column and a mobile phase consisting of water and acetonitrile (70%:30%, v/v) containing 0.1% trifluoroacetic acid [2]. The resulting amount of doxorubicin corresponds to the non-entrapped part of drug hence affecting the loading efficiency. The following nanoparticle materials have been investigated: human serum albumin (HSA), chitosan, poly(lactic-co-glycolic acid) (PLGA) and PLGA modified with polyethylenimine (PLGA-PEI). For particle preparation different techniques such as desolvation, emulsion-diffusion and nanoprecipitation were used. For example, HSA nanoparticle were formed according to a standard desolvation method, while PLGA and PLGA-PEI nanoparticles were manufactured by emulsion-diffusion and nanoprecipitation techniques [3,4]. With this work, we want to present promising nanoparticle systems, and furthermore, discuss problems and difficulties that have occurred. References: 1. Haley, B.; Frenkel E.: Urol. Oncol. 2008, 26: 57-64. 2. Dreis, S. et al.: Int. J. Pharm. 2007, 341: 207-214. 3. Langer, K. et al.: Int. J. Pharm. 2003, 155: 75-82. 4. Astete, C. E.; Sabliov, C. M.: J. Biomater. Sci. Polym. Ed. 2006, 17: 247-289. 126 • DPhG Annual Meeting 2015 Conference Book POS.034 Archaesomal enwrapped polyplexes (AePPs) for the expression of luciferase plasmid (pCMV-luc) in various cell lines Engelhardt, K. H.1; Pinnapireddy, S.1; Baghdan, E.1; Bakowsky, U.1 1 Department of Pharmaceutical Technology and Biopharmaceutics, Ketzerbach 63, 35037 Marburg, Germany Gene therapy is a relatively new and widely researched field with great potential in treating a wide range of human disorders. Nucleic acid-based biopharmaceuticals allow treatment of not only symptoms but also the very cause of an array of diseases by induction and/or inhibition of genes. Gene therapy is based on the principle of delivering an intact fragment of genetic material into the cytoplasm or nucleus of a desired cell. The wide array of non-viral vectors can be broadly classified into nanoparticles, polymers and lipid vectors. Lipid vectors are the only synthetic vectors currently used in clinical trials. However, a number of problems still need to be resolved before liposomes can be used in a clinical setting. One of the major problems is poor stability of liposomes, which is partially attributed to the hydrolysis of ester bonds and oxidation of unsaturated fatty acids [1]. To overcome these stability issues is the use of more stable lipids for the preparation of transfection reagents. A unique class of lipids, ether lipids, found exclusively in the cellular membranes of the third domain of life referred to as archaea, can be considered promising alternatives to common lipids. Members of the archaea are known to inhabit environments ranging from ordinary to ones characterised by extremes of salt, temperature and pH [2]. The presence of ether lipids in membranes of the archaea is believed to be responsible for their unusual temperature and pH stability. Lipids from Sulfolobus are characterized by two different backbones. The first one is called Glycerol-Dialkyl-GlycerolTetraethers (GDGT), where the glycerol is linked with two biphytanyl ether chains at both sides. The second one is called Glycerol-DialkylNonitol-Tetraethers (GDNT) containing an additional nonitol group [3]. Liposomes containing different kinds of tetraether lipids (archaeosomes) were first prepared by using the thin-film hydration method and characterized by AFM (Atomic Force Microscopy) and DLS (Dynamic Light Scattering). Polyplexes were formed by mixing 25-kDa branched polyethylenimine (25kDa-bPEI) with pDNA at N/P 15. Archaeosomal enwrapped polyplexes (AePPs) were finally obtained by mixing achaeosomes and polyplexes at increasing Lipid/DNA ratios. For transfection experiments COS-7 and SKOV-3 cells were seeded at a density of 1 x 104 cells/per well on a 96-well plate 24h before transfection experiments. After 4 h transfection with pDNA, cells were incubated 48 h before determination of gene expression efficiency using luciferase assay. Polyplexes at N/P 15 and archaeosomes had an overall charge of approximately +35 mV and -27 mV, respectively. Depending on the amount of archaeosomes incubated with polyplexes, AePPs had a mean diameter of 200-300 nm and zeta potential ranged from +35 to +8 mV. Transfection experiments of aforementioned AePPs revealed successful transfection of COS-7 and SKOV-3 cells at certain Lipid/DNA ratios. A reduction of 20-50% of transfection efficiency in comparison with bPEI alone would be acceptable, considering the much higher stability of AePPs. Polyplexes could be enwrapped with archaeosomes resulting in new transfection reagents that are nanoscaled and stable. AePPs could be easily prepared by adding positively charged polyplexes to negatively charged archaesomes. Due to the unique chemical structure of tetraether lipids, they can resist the acidic environment in the GIT and are less susceptible to enzymatic degradation. These properties make them more suitable as drug delivery systems, especially for sensitive compounds like DNA. Acknowledgments: The authors would like to thank the Deutsche Forschungsgemeinschaft (DFG) for the financial support. References: 1. Felgner, PL. et al.: Proc. Natl. Acad. Sci. 1987, 84: 413-7417. 2. Hanford, J.M.; Peeples, L.T.: Appl. Biochem. and Bio. 2002, 97(1): 45-62. 3. Jacquemet, A.: Biochimie. 2009, 91(6): 711-717. PHARMACEUTICAL TECHNOLOGY AND DRUG FORMULATIONS POS.035 POS.036 Birch bark extract particle deposition from supercritical solutions and use for stabilisation of semisolid systems Examination of homogeneity of stent coatings produced via fluidized bed process Armbruster, M.1; Wahl, M. A.1 Wentzlaff, M. 1; Senz, V. 2; Grabow, N2; Weitschies, W. 1; Seidlitz, A. 1 1 Department Germany of Pharmaceutical Technology, Auf der Morgenstelle 8, 72076 Tübingen, Introduction: Birch bark dry extract consisting of pentacyclic triterpenes like for example betulin can be used to stabilize semisolid systems such as creams and gels [1,2]. One of the crucial parameters for this function as a pickering emulsifier and a gelling agent is the extract´s surface [3]. Usually birch bark extract is produced by organic solvent extraction (OSE). Birch bark extraction with supercritical fluids can be used as an alternative extraction method [4], with different types of particle deposition. Therefore, this method could enable the creation of extract particles with different surface properties and thereby improve the production of semisolid systems. The obtained extracts were investigated by gas adsorption (BET) and inverse gas chromatography (iGC) to characterize the extract´s surface. To determine the extracts’ ability to stabilize semisolid systems, gels were made and their rheology studied by amplitude sweep method. Materials and Methods: The birch bark (Birken AG) extraction was performed on a high pressure lab scale extraction unit (Sitec) at 350 bar and 60 °C with supercritical (sc) carbon dioxide (CO2) as extraction solvent. Three different types of particle deposition were tested (Figure 1). One way of particle deposition is the generation of a liquid CO2 phase in a separator, in which the sc solution is expanded at 40 bar. In this liquid phase the extract accumulates and when the pressure is released particles deposit and can be removed as a dry extract (SCF-E) (Figure 1a). Another option is the RESS (rapid expansion of supercritical solutions) method. Thereby a sc solution is sprayed through a nozzle and particles deposit directly into the gaseous phase (R-E) (Figure 1b). This method has been modified by spraying directly into a lipophilic phase (jojobaoil) to create a gel in a single step production, including extraction, particle deposition and gel formation (Figure 1c). Other gels were produced by dispersing 6% extract in jojobaoil with an Ultra-Turrax® (Ika Labortechnik) for two minutes at 8000 rpm. The amplitude sweep (AS) was performed at 23 °C with a 25 mm plate-plate setup and a gap of 0.8 mm at a Physika MCR 501 rheometer (Anton Paar). For the AS a log ramp (deformation 0.1-1000%; frequenzy 1/s) was used. Figure 1a-c: different types of sc particle deposition; Figure 2: gel formation of OS-E gel and SCF-E gel Results: The first 6 hours of gelling were investigated for gels made out of OS-E and SCF-E by performing an AS every 30 min. The mean storage modulus (G’) in the linear viscoelastic region is used as an indicator for gel strength and viscosity (Figure 2). For the SCF-E gel the G’ values start and remain at a very high level compared to the OS-E gel. Another AS after 24 hours acts as a reference for progressing gelling process. The SCF-E gel easily reaches this point within the 6 hours whereas the OS-E gel comes up to only 28% of the reference. Thus, the SCF-E is able to stabilize the gels faster and better. One possible explanation is the SCF-E´s higher BET surface area (36.2 ± 2 m2/g) compared to the OS-E (29.4 ± 0.7 m2/g). Conclusion: Birch bark particle deposition from supercritical CO2 can provide extracts with superior gelling properties, compared to organic solvent extract. Most likely, this is caused by different deposition conditions and therefore different extract surface properties. References: 1. Scheffler, A.: United States Patent US2003087789-2003-05-08, 2003. 2. Laszczyk, M.: Triterpentrockenextrakt aus Birkenkork 2007. 3. Grysko, M.: Herstellung und Charakterisierung von halbfesten Systemen auf der Basis vonTriterpentrockenextrakt aus Birkenkork 2011. 4. Krasutzky, P.: Nat. Prod. Rep. 2006, 23(6): 919-942. 1 Institute of Pharmacy, C_DAT, Ernst-Moritz-Arndt University of Greifswald, Felix-HausdorffStraße 3, 17487 Greifswald, Germany 2 Institute for Biomedical Engineering, University of Rostock, Friedrich-Barnewitz-Straße 4, 18119 Rostock, Germany For abstract see Short Poster Lecture SPL.003 on page 100. POS.037 Lipoproteins as new protein delivery system Thoma, F.1; Langer, K.1 Institute of Pharmaceutical Technology and Biopharmacy, University of Muenster, Corrensstraße 48, 48149 Muenster, Germany 1 Proteins are very important pharmaceuticals for the treatment of several diseases, but in some cases the therapeutic application is limited by insufficient delivery options. Currently in the field of protein delivery many promising drug delivery systems such as nanoparticles or liposomes enter the focus of research. But especially polymer based nanoparticles have been found to be inadequate for protein delivery due to the high protein adsorption on hydrophobic surfaces and low entrapment efficiency [1]. Hence, the aim of this work was the establishment of a new kind of protein delivery system. Oriented on physiological carriers micelle-like lipoproteins were built based on soy lecithin, triglycerides, and cholesterol by an emulsification process using human serum albumin (HSA) as model protein. The resulting lipoproteins were purified by extensive dialysis. Furthermore, different mass ratios of the ingredients were used to allow an enhanced protein loading. The resulting lipoproteins were characterized with regard to their physicochemical properties such as hydrodynamic diameter, zetapotential, and polydispersity index (PdI) by photon correlation spectroscopy (PCS). In addition, the stability of lipoproteins under physiological conditions was confirmed by titration with NaCl solution up to the physiological concentration of 150 mmol/L. To determine the protein integrity after lipoprotein preparation dissolved lipoproteins were applied on a sodium dodecylsulfate polyacrylamide gel electrophoresis (SDS-PAGE). Despite the occurrence of strong shear forces during emulsification process no additional bands of protein fragments or aggregates were detectable. In conclusion we were able to establish lipoproteins as an appropriate protein delivery system with respect to physical stability and protein loading. References: 1. Barichello, J. M. et al.: Drug Dev. Ind. Pharm. 1999, 25(4): 471-476. POS.038 In vitro dissolution behavior of extended-release theophylline capsules in media containing alcohol Knop, K.1; Kremer, M.1; Ridder, S.1; Kleinebudde, P.1 1 Institute of Pharmaceutics and Biopharmaceutics, Heinrich Heine University Duesseldorf, Universitaetsstr. 1, 40225 Duesseldorf, Germany In 2005 severe interactions were reported for extended-release hydromorphone capsules taken together with alcohol (Palladone case, FDA alert 7/2005 [1]). Therefore the FDA implemented in vitro dissolution test media containing up to 40% ethanol in their 'product-specific recommendations for generic drug development' guidelines for several drugs [2]. The potential appearance of dose dumping is important for all drugs with a narrow therapeutic index and can be detected by in vitro dissolution tests. Aim of the present study was to evaluate the influence DPhG Annual Meeting 2015 Conference Book • 127 POSTERS of alcohol on the drug release from extended-release theophylline capsules purchased on the German market. Four extended-release capsule preparations (Bronchoretard 350, Euphylong 300 mg, Theophyllin AL 300 retard, Theophyllin Hexal 300 mg) containing different retardation excipients were purchased in a local pharmacy. The dissolution tests were performed using the basket apparatus (Ph.Eur. 2.9.3) and the test conditions described for 'Theophylline Extended-Release Capsules' Test 6 in the USP 37 (0.05 M pH 6.6 phosphate buffer, 1000 mL, 100 rpm). 10, 20 and 40% of the buffer solution were substituted with ethanol. Theophylline concentration was determined spectroscopically at 271 nm. Only the content of the capsules (pellets) was used for dissolution testing. Disintegration tests (Ph.Eur. 2.9.1) with the complete capsules were performed in the different media to ensure that the capsule shell didn't have an influence. The influence of capsule disintegration on dissolution can be neglected because all capsules disintegrated within 1 to 2 min in buffer solution and within 2 to 3 min in buffer with 40% ethanol. All preparations exhibited extended release behavior in phosphate buffer with MDT (mean dissolution time) values between 3 h (Euphylong) and 10 h (Theophyllin Hexal). In ethanolic media all preparations showed dose dumping but to a different extent. While Bronchoretard released the total dose in the first two hours in media with 40, 20 and even 10% ethanol (see Figure), Euphylong and Theophyllin AL underwent dose dumping only in media with 40% ethanol. Theophyllin Hexal represented a central position with dose dumping in 40 and 20%, but not in 10% ethanol. The more ethanol sensitive preparations Bronchoretard and Theophyllin Hexal contain copolymers of ethyl acrylate and methyl methacrylate or ammonio methacrylate as retardation excipients. Euphylong and Theophyllin AL comprise cellulose derivatives and were less affected by alcohol in lower concentrations. In comparison to extended release theophylline matrix tablets [3] the capsule preparations were more vulnerable to ethanol in the dissolution media. As a consequence, extended release theophylline capsules should not be taken together with alcohol in higher concentrations. References: 1. FDA: FDA Alert [7/2005]: Alcohol-Palladone interaction, http://www.fda.gov/Drugs/DrugSafety/PostmarketDrugSafetyInformationforPatientsandProvi ders/ucm129288.htm (11.06.2015). 2. FDA: Product-specific recommendations for generic drug development, http://www.fda.gov/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/ucm07520 7.htm (11.6.2015). 3. Knop, K.; Kleinebudde, P.: In vitro dissolution behavior of extended release theophylline tablets in ethanolic media, 1st European Conference on Pharmaceutics, Reims 2015. POS.039 Oral carbon monoxide release system Steiger, C.1; Meinel, L.1 1 Institute for Pharmacy and Food Chemistry, University of Würzburg, Germany Introduction: Carbon monoxide (CO) has therapeutic effects in various gastrointestinal diseases [1] yet clinical use today is challenged by inappropriate delivery modes [2]. Consequently, we developed a tablet referred to as oral carbon monoxide release system (OCORS) providing precise, controlled and targeted CO delivery for the treatment of gastrointestinal injury and inflammation, respectively. The model was downsized to 1 mm and 0.5 mm in diameter for preclinical models (Figure B). Methods: OCORS is an oral tablet based on sulfite induced CO release from the CO releasing molecule 2 (CORM-2) [2]. OCORS performance was detailed as a function of the presence of buffer within the tablet core and characteristics of a water-insoluble cellulose acetate coating, forming a semipermeable shell around the tablet core. Amperometric detection was deployed for recording CO release profiles throughout 20 hours. Results and Discussion: OCORS was tuned for environmental pH insensitivity by appropriate buffer systems blended within the tablet core. 128 • DPhG Annual Meeting 2015 Conference Book CO release kinetic of 1 mm OCORS is tailorable from 25 min up to 20 hours by variation of coating thickness and coating hydrophobicity (Figure A). OCORS is a readily available tablet for oral use in preclinical models. The controlled release system reliably delivered CO independent of environmental pH, such that the therapeutic gas can be safely generated at gastric, intestinal or colonic sites. In vivo experiments of OCORS are required to demonstrate the pharmacokinetics and clinical potential of this oral delivery platform for therapeutic gases. Figure: A) Fast, intermediate, and slow CO release pattern of the 1mm oral carbon monoxide release system (OCORS). B) 0.5 mm diameter OCORS along with a human hair for size comparison. References: 1. Motterlini, R.; Otterbein, L. E.: Nat. Rev. Drug Discov. 2010, 9(9): 728-U724. 2. Steiger, C. et al.: J.Control Release 2014, 189C: 46-53. POS.040 Vinyl sulfone-poly(vinyl alcohol)-stabilized PLAnanoparticles as reactive particle system for various surface modifications Raudszus, B.1; Langer, K.1 Institute of Pharmaceutical Technology and Biopharmacy, University of Muenster, Corrensstr. 48, 48149 Muenster, Germany 1 The functionalization of the surface of nanoparticles plays an important role as an approach for targeted drug delivery since a specific ligand may lead to the ability to overcome several physiological barriers like the blood-brain barrier [1]. The aim of the present project is to obtain nanoparticles, which feature a surface modification with Apolipoprotein E in order to allow the particles to cross the blood-brain barrier via receptormediated transcytosis [2]. Poly(vinyl alcohol) (PVA) is a polymer often used for the preparation of poly(lactic-co-glycolic acid) (PLGA) or poly(lactic acid) (PLA) nanoparticles, because of its ability to stabilize nanoparticles successfully because of sterical effects [3]. Due to the high presence of PVA on the particle surface, the carboxyl groups of the polymers PLGA and PLA are less accessible for coupling reactions. Therefore the PVA was replaced by the more reactive vinyl sulfone-poly(vinyl alcohol) (VS-PVA). The vinyl sulfone-poly(vinyl alcohol) was synthesized by adding an excess of divinyl sulfone to a solution of PVA in 0.1 M NaOH. The reaction was stopped by addition of HCl. Afterwards the synthesized vinyl sulfone-poly(vinyl alcohol) was purified by dialysis, in order to remove excess divinyl sulfone. The purified product was analysed by 1H-NMR and IR spectroscopy to prove the covalent linkage of the reaction partners. The introduced vinyl sulfone-group is amine- and thiol-reactive, which makes further modifications with molecules, containing these functional groups, possible. The investigated nanoparticles were based on PLA and prepared by emulsion-diffusion method with VS-PVA as stabilizer. The obtained nanoparticles had a size of about 230 nm, PDI below 0.1, and a negative zetapotential of about -47 mV. After nanoparticle preparation different amines like tyramine or PEG derivatives were coupled by adding a solution of these substance to the nanoparticle suspension. In order to increase the nucleophilicity of the amine-containing reagent, the reaction was performed in borate buffer of pH 9. In conclusion the obtained particle system offers a starting point for an effective functionalization of PLGA or PLA particle surfaces. Attached PEG chains can be used as a linker to introduce further ligands like proteins, peptides or other macromolecular compounds, which cannot be attached to the surface directly because of steric hindrance. References: 1. Kreuter, J.: Adv. Drug Deliv. Rev. 2013, 71: 2-14. 2. Zensi, A. et al.: J. Control. Release 2009, 137: 78-86. 3. Astete, C.; Sabliov C.: J. Biomater. Sci. Polymer Edn. 2006, 17: 247-289. PHARMACEUTICAL TECHNOLOGY AND DRUG FORMULATIONS POS.041 Polyarginine modified human serum albumin (HSA) nanoparticles for effective cell transfection sulphate was extracted from the sprayed material with sulphuric acid and quantified utilising derivatisation with para-dimethylaminobenzaldehyde followed by photometric determination. Mesken, J.1; Langer, K.1 Institute of Pharmaceutical Technology and Biopharmacy, University of Muenster, Corrensstraße 48, 48149 Muenster, Germany 1 Non viral gene therapy has entered the focus of many research groups in the field of gene therapy in humans. By using a non-viral approach the safety risks of viral gene delivery systems such as carcinogenicity, immunogenicity or inflammation can be avoided. However in many cases an efficient gene transfer to cells is solely achieved by viral vectors. Hence the aim of the present work was the preparation of biodegradable, nontoxic, plasmid-loaded human serum albumin (HSA)-based nanoparticles with a promising transfection efficiency. Therefore the nanoparticle-surface was modified by the arginine-rich cell-penetrating peptide nona-arginine. The plasmid-loaded nanoparticles based on HSA were prepared by desolvation technique and were stabilised by glutaraldehyde as described previously [1]. As a model plasmid a vector encoding the mCherry fluorescent protein was used. The modification of the surface was performed in a three-step strategy. After purification of the nanoparticles followed by the introduction of thiol groups onto the surface [2], nona-arginine was activated by a bifunctional PEG-based crosslinker, followed by coupling to sulfhydryl groups of the particle surface. Dynamic light scattering (DLS) analysis showed a monodisperse size distribution and the surface modification worked successfully which could be revealed by C18-RP-HPLC analytics. Cell culture studies with a focus on transfection efficiency were conducted in HEK293 and HT-29 cells using fluorescence microscopy and flow cytometry. Cytotoxicity was evaluated using WST assay and cellular uptake experiments were performed using fluorescent labelled HSA-nanoparticles. In conclusion it was possible to develop a biodegradable, nontoxic particle system with a potential to transfect cells. Figure: microscopic image of foam sample generated from a an HFA suspension containing 4% PLA and 8% solid particles Generally, samples solidify in minutes after actuation and show increased material density at specimen’s edges. Microscopic observations elucidate interconnected pores with a diameter > 200 µm (Figure). Solutions of 4 to 7% PLA in HFA are emptied to more than 97.5% (standard deviation 0.8%) from the canister, whereas higher concentrations are not completely expelled due to high viscosity. XRPD results indicate conversion of calcium phosphates to hydroxyapatite in < 24 h. This finding is confirmed by scanning electron microscopy which shows the appearance of typical needle-like crystals. Polymer-containing samples typically show a decreased brittleness which finds its expression in an increased deflection. Acknowledgments: This study is part of a project being funded by the Business Development and Technology Transfer Corporation of Schleswig-Holstein and Stryker Trauma GmbH. References: 1. Ginebra, M.-P. et al: Adv. Drug Deliv. Rev. 2012, 64(12): 1090–1110. 2. Salgado, A. J. et al.: Macromol. Biosci. 2004, 4(8): 743–765. POS.043 References: The increase of dermal bioavailability of the antioxidant rutin by new smartCrystal technology 1. Steinhauser, I. et al.: J. Drug Target. 2009, 17 (8): 627-637. 2. Weber, C. et al.: Int. J. Pharm. 2000, 211: 67-68. Pyo, S. M.1; Keck, C. M. 2; Müller, R. H.1 1 Institute of Pharmacy; Pharmaceutics, Pharmaceutical Nanotechnology & NutriCosmetics, Freie Universität Berlin, Kelchstr. 31, 12169 Berlin, Germany 2 PharmaSol GmbH, Stubenrauchstr. 66, 12161 Berlin, Germany POS.042 Novel foamable cement formulation for application of antibiotics to bone voids Arntz, P.1; Nassut, R.2; Scherließ, R.1; Steckel, H.3 1 Department of Pharmaceutics and Biopharmaceutics, Kiel University, Grasweg 9a, 24118 Kiel, Germany 2 Stryker Trauma GmbH, Prof.-Küntscher-Strasse 1-5, 24232 Schönkirchen / Kiel, Germany 3 Deva Holding A.S., Halkalı Merkez Mah. Basın Ekspres Cad. No:1, 34303 Küçükçekmece/İstanbul, Turkey Calcium phosphate cements are recognised as biocompatible and bioactive materials for bone replacement and are discussed as drug delivery systems [1]. Disadvantages of currently marketed products are a laborious manual application procedure and their low flexural strength. Also, cellular penetration should be granted in an ideal formulation to allow for vascular ingrowth and remodelling to native bone [2]. These issues are addressed with a novel foam formulation. Tetra-calcium phosphate, di-calcium phosphate and tri-sodium citrate were milled (Labstar, Netzsch, Germany) in ethanol (99.9% V/V) to an average particle size in the upper nanometre range (Zetasizer Nano ZS, Malvern, UK) and filled into glass bottles containing poly-D,L-lactic acid (Resomer R 205) and 1% gentamicin sulphate. Ethanol content is reduced to 35% (m/m) of dry residue by vacuum drying to minimise the amount of ethanol per dose but avoid agglomerate formation. After crimping using continuous valves, PLA is dissolved in 1,1,1,2,3,3,3heptafluorpropane (HFA). Samples derived from actuation were investigated regarding macro- and microscopic appearance of foamed specimen, hydroxyapatite formation using x-ray powder diffraction (XRPD) (STUDI P, STOE & Cie GmbH, Germany), suspension stability and content delivery. Three-point bending (BZ2.5/TN1S, Zwick/Roell AG, Germany) was conducted and compared to results of samples without PLA. Finally, sprayed gentamicin Although the flavonoid rutin has excellent antioxidant properties on cellular level, it shows too poor skin penetration and thus an insufficient dermal bioavailability due to its low solubility and slow dissolution rate. To enable the dermal use of rutin its solubility was increased by smartCrystal technology where the µm-sized rutin raw drug powder was reduced in size to submicron-size range. Whether rutin smartCrystals bring a real improvement in penetration and antioxidant activity compared to raw drug powder was investigated by the performance of an ex vivo penetration study and an in vitro antioxidant activity study. Rutin smartCrystal suspension was kindly provided by PharmaSol GmbH. As reference rutin raw drug powder suspension was produced with identical composition. Particle size distributions of both suspensions were compared by laser diffraction (LD, Mastersizer 2000, Malvern Instruments, UK). For the ex vivo penetration study tape stripping test was performed on pig ears with two hydrogels containing either rutin smartCrystals or raw drug powder both at 5%. DPPH (2,2-diphenyl-1picrylhydrazyl) assay was performed to compare the antioxidant activity of rutin smartCrystal contained in intense lifting eye serum (Dr, JK Cosmeceuticals, Germany) with eight other anti ageing products on the market having rutin or its derivatives as active. The maximum discoloration of a methanolic DPPH solution with an absorption of 1.0 at a wavelength 517 nm was measured over 60 minutes with a PharmaSpec UV-1700 photometer (Shimadzu Corporation, Japan). LD diameters 50% of 240 nm and 90% of 860 nm confirmed the submicron size of the rutin smartCrystals. The penetration depths and strengths of rutin applied as smartCrystal and raw drug powder hydrogel were compared. Until the 7th tape strip 1.5 fold higher amount of rutin could be detected for the raw drug powder hydrogel. This shows a remaining of active in the upper layers of the stratum corneum standing for low penetrated amount of active into the deeper layers. Starting from the 8th tape strip the penetration behaves inversely. In the deeper and therefore more relevant layers of the stratum corneum rutin DPhG Annual Meeting 2015 Conference Book • 129 POSTERS smartCrystal hydrogel shows 2.2 and 2.5 times higher rutin amount for the 14th and 23th tape strip, respectively, proving the significant penetration enhancement of rutin smartCrystals compared to the raw drug powder. The DPPH assay allowed the division of tested products into three antioxidant activity classes from very strong discoloration of the methanolic DPPH solution (> 80%, class I) representing very strong antioxidant activity to medium (60 to 20%, class II) and low (< 10% class III) discoloration standing for almost no antioxidant activity. Only the intense lifting eye serum (Dr, JK Cosmeceuticals, Germany) with a discoloration of more than 85% within 15 minutes counted to class I. The highest discoloration reached from the reference products was under 60% and three of them even did not shown any discoloration effects on the DPPH solution. Thus, the antioxidant activity of intense lifting eye serum (Dr, JK Cosmeceuticals, Germany) with rutin as smartCrystals is superior towards the tested dermal anti ageing products with rutin or its derivatives. Rutin smartCrystals lead to a distinctly deeper and up to 2.5 times higher penetration into the skin of active compared to raw drug powder formulation. Also the antioxidant activity of rutin smartCrystals in intense lifting eye serum (Dr, JK Cosmeceuticals, Germany) proved to be superior in comparison to other marketed products with rutin formulated in standard manner. So, the combination of deeper and stronger penetration with higher antioxidant activity will automatically lead to an improved dermal bioavailability. Thus, the smartCrystal technology enables the effective use of poor soluble plant extracts such as rutin in cosmetic and dermal formulations. in Milli-Q water. The samples were stored at 25 °C shaking with 100 rpm in an Innova 4230 shaker for some hours. Then the formulations were incorporated into 5% hydroxypropyl cellulose (HPC) gel, and were studied in the pig ear penetration test via tape stripping. High performance liquid chromatography (HPLC) was performed to determine the drug amount. Different actives have different maximum loading in smartPearlsTM, the range changed from 41% of coenzyme Q10 to 15% of betulin. All of the loaded smartPearlsTM expressed superior ability in dermal delivery. For example, the 32% azithromycin loaded Syloid 3D showed 14 times higher Cs than RDP and 7 times higher Cs than 190 nm sized nanocrystals. According to the pig ear study, the smartPearls® demonstrated a higher penetration than their comparison, especially in deeper skin layers. For instance, the cyclosporine loaded Syloid 3D gel (1% active in gel) revealed 6 times higher penetration than 5% RDP and 5% 300 nm sized amorphous nanoparticles in the 20th - 30th layer. The 1% rutin Syloid 3D gel represented up to 35 times higher penetration than 250 nm sized nanocrystals (5% in gel) in the 17th - 19th layers. The smartPearlsTM proved to be an effective dermal delivery system for poorly soluble actives for cosmetics and pharmaceuticals. smartPearlsTM are superior due to the increased Cs and skin penetration, and preserve the amorphous state. In view of the discussion about nanoparticles, they are not nanoparticles but a micrometer sized delivery system (e.g. 10 to 40 μm porous particles). Acknowledgments: PharmaSol GmbH Berlin, Germany, Grace GmbH & Co. KG, Worms, Germany and China Scholarship Council (CSC). References: 1. Wei, Q.; Keck, C. M.; Müller, R.H.: Int. J. Pharm. 2015, 482(1-2): 11-20. 2. Monsuur, F.; Höfer, H.H.; Keck, C. M.: US patent application 2014. POS.044 smartPearlsTM: Novel dermal amorphous delivery system based on porous particles Keck, C. M.1; Jin, N.2; Du, W. J.2; Staufenbiel, S.2; Monsuur, F.3; Höfer, H. H.3; Müller, R. H.2 1 PharmaSol GmbH, Stubenrauchstr. 66, 12161 Berlin, Germany of Pharmacy, Freie Universität Berlin, Kelchstr. 31, 12169 Berlin, Germany GmbH & Co. KG, In der Hollerhecke 1, 67547 Worms, Germany 2 Institute 3 Grace Poorly soluble cosmetic and pharmaceutical actives have often a very poor dermal penetration and thus low bioavailability, often not reaching the effective concentration. Approaches such as penetration enhancers have problems such as tolerability by the skin and lack of regulatory accepted status. A smarter approach is to increase the saturation solubility Cs. This leads to an increased concentration gradient, and thus increased penetration, e.g. by using amorphous µm-sized actives, drug nanocrystals (small size creating increased dissolution pressure), or the combination of both, i.e. amorphous nanoparticles (e.g. NanoMorph concept). However, the problem of the amorphous state is its low stability, tendency to re-crystallize. In the last decade, increasing interest focussed in oral drug delivery on porous materials loaded with drug in the amorphous state (e.g. CapsMorph), using e.g. porous silica. The confinement of the drug in small pores prevents re-crystallization, which makes the amorphous state stable up to 5 years [1]. The small size dimension of the actives in the pores increases solubility, on top of the effect of the amorphous state. This delivery principle was transferred to the dermal administration route (smartPearls) [2]. For example, mesoporous silica Syloid® SP53D-11920 (Syloid 3D, company W. R. Grace & Co., Worms, Germany) was used for loading with various actives. Both drugs (e.g. azithromycin, cyclosporine) and cosmetic and consumer care agents (e.g. betulin, rutin, hesperidin, coenzyme Q10) have been used as model actives to investigate the performance of smartPearls® as dermal carriers. For comparison, raw drug powder (RDP), the “gold standard” nanoparticles, commercial products or effective clinical products were tested in skin penetration studies. Each active was dissolved in suitable organic solvent and was sprayed manually by a spraying nozzle onto certain amounts of silica (impregnation method). Subsequently, the solvent was evaporated in a compartment dryer at suitable temperatures. The solvent should evaporate completely under the temperature in which the active can keep its activity. In addition, the solvent should be safe in dermal use. The theoretical maximum loading is relative to the density of the drug, the density of the solvent and the pore volume of the silica. The practical maximum loading of silica was monitored by x-ray diffraction (XRD, Philips PW 1830). To get Cs, the maximum loaded silicas were dispersed 130 • DPhG Annual Meeting 2015 Conference Book POS.045 Polymorph formation during non-sink dissolution testing of co-amorphous indomethacin-arginine: spray dried powder vs. tablet formulation Lenz, E.1; Jensen, K. T.2; Knop, K.1; Grohganz, H.2; Löbmann, K.2; Rades, T.2; Kleinebudde, P.1 1 Institute of Pharmaceutics and Biopharmaceutics, Heinrich Heine University, Universitaetsstrasse 1, 40225 Duesseldorf, Germany 2 Department of Pharmacy, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark Co-amorphous drug-amino acid formulations are a promising alternative to common amorphous formulations which contain polymers as stabilizers. The co-amorphisation of indomethacin with arginine results in a physically stable preparation due to ionic interactions, and furthermore the intrinsic dissolution rate is improved compared to the pure amorphous drug [1]. Recently, spray dried indomethacin-arginine was successfully formulated into tablets [2]. In this study, the non-sink dissolution behaviour of pure spray dried powder was investigated and compared to the dissolution behaviour of tablets. Indomethacin-arginine (IND-ARG, 1:1 molar ratio, 1:0.49 weight ratio) was prepared from a 4% w/v solution in acetone/water (70:30; Büchi B290) [1]. For preparation of tablets (TAB IND-ARG), mannitol (Parteck® M200), croscarmellose sodium (Ac-Di-Sol® SD-711), colloidal silicon dioxide (Aerosil® 200) and magnesium stearate (Parteck® LUB MST) were used. Flat faced tablets were pressed with a drug load of 50 mg using a rotary die press with 82 MPa. Non-sink dissolution testing was performed in 900 mL KH2PO4 buffer (100 mM, pH 4.5, 37 °C) for 24 h with a paddle apparatus (n = 3; 50 rpm; HPLC analysis). After 24 h, media were filtered and the obtained precipitates were dried. They were characterized using XRPD and DSC. Scanning electron microscopy was performed after sputter-coating. Dissolution profiles of spray dried IND-ARG showed an immediate release of IND resulting in a maximum concentration (cmax) of 42.9±1.3 mg/L (tmax 7 min). The solubility of crystalline IND in the dissolution medium was thus exceeded by a factor of about 6. This supersaturation led to a fast crystallization of IND in a needle-like shape, whereas the stable γ-IND exhibits prism- and plate-like crystals. XRPD and DSC measurements showed that the precipitates consisted of α-IND, which is the most commonly observed metastable polymorphic form [3,4]. TAB IND-ARG slowly eroded within 40 min, resulting in a drug release with a cmax of 26.1±2.8 mg/L (tmax 35 min) and a supersaturation by a factor of about 4. The supersaturation was maintained for a longer PHARMACEUTICAL TECHNOLOGY AND DRUG FORMULATIONS period compared to IND-ARG, and as a consequence comparable areas under the curve were obtained after 24 h. The precipitates of TAB INDARG consisted also of needle-shaped crystals, but according to XRPD and DSC, another not yet identified polymorph was formed. In conclusion, co-amorphous IND-ARG crystallizes in different polymorphic forms in aqueous media depending on the rates of crystallization. References: 1. Jensen, K. T. et al.: J. Pharm. Pharmacol. 2015, (submitted). 2. Lenz, E. et al.: Tableting of spray dried co-amorphous indomethacin-arginine, AAPS Annual Meeting & Exposition 2014, San Diego. 3. Lin, S. Y.: J. Pharm. Sci. 1992, 81: 572-576. 4. Surwase, S. A. et al.: Mol. Pharmaceutics 2013, 10: 4472-4480. resulting in a higher densification factor. Therefore, the density of the resulting ribbon is higher and its porosity, consequently, lower. In conclusion, two scales of Freund-Vector compactors were investigated for different combinations of materials and pressures, and the porosity of the resulting ribbons was evaluated. It was observed that the increase in pressure reduces the porosity and higher fraction of MCC leads, in general, to higher porosities. It was confirmed that the higher the roll diameter, the lower the porosity of the resulting ribbons. Acknowledgments: This work was supported by the IPROCOM Marie Curie initial training network, funded through the People Programme (Marie Curie Actions) of the European Union’s Seventh Framework Programme FP7/2007-2013/ under REA grant agreement No. 316555. The authors would like also to thank the University of Surrey (Prof. Charley Wu and his group, especially Ms. Serena Schiano) and AstraZeneca in Macclesfield (Dr. Gavin Reynolds and Dr. Andreja Mirtic) both located in U.K., for the materials provided and for allowing the using of their pieces of equipment. POS.046 Effect of the scale of two Freund-Vector roll compactors on the porosity of ribbons Pérez Gago, A.1; Kleinebudde, P.1 Institute of Pharmaceutics and Biopharmaceutics, Heinrich-Heine-University, Universitätstrasse 1, 40225 Düsseldorf, Germany 1 In the pharmaceutical industry, the scale-up or transfer of a formulation from the lab scale to the pilot scale is a necessary step. Roll compaction is a continuous dry granulation process commonly used and therefore, understanding its scale-up is highly interesting. Several suppliers of roll compactors applied diverse design techniques when they developed their equipment in different scales. One of these manufacturers is the Freund-Vector Corporation which compactors are characterized by changing the roll diameter and roll width from one scale to other while other suppliers keep the roll diameter constant and only change the roll width. The aim of this work is to investigate how the change in scale affects to the porosity of the ribbons produced from MCC, mannitol and five binary mixtures of 15, 30, 50, 70 and 85% MCC using two different scales of roll compactors from the Freund-Vector Corporation. These excipients were chosen as they present opposite behaviours against compaction and they are widely used in the pharmaceutical industry. The TFC-Lab Micro and the TF-Mini (Vector Corporation, Marion, U.S.A.) were used to carry out the roll compaction process using a smooth roll surface and rim rolls as sealing system in both compactors, ensuring that only the scale has an effect on the products obtained. The TFC-Lab Micro is a small lab-scale compactor which roll diameter is 50 mm and width 24 mm while the TF-Mini has rolls of 100 mm diameter and 37 mm width. The pure materials together to the 50% MCC mixture were roll compacted in both scales under three different pressures (2, 5 and 8 MPa) while keeping the roll speed in 2 rpm and adjusting the feeding screw speed in order to reach a gap of 1.5 mm. The roll compaction of the 15, 30, 70 and 85% MCC mixtures was performed only at 5 MPa pressure. The resulting ribbons were characterized regarding porosity using an envelope and T.A.P. density analyser (GeoPyc® 1360, Micromeritics Instrument Corp., Norcross, U.S.A.). The porosity of the resulting ribbons was compared in the two scales considering on the one hand, the effect of the material properties and on the other hand, the pressure applied during the compaction process. If the different materials compacted at 5 MPa are compared, the porosity changes from 20.3% to a maximum of 33.2%, and although some differences between the two compactors were observed, in general, the porosity increases while the proportion of MCC rises. However, if the effect of the pressure is analysed for the pure materials and the 50% MCC which were compacted at 2, 5 and 8 MPa, as expected, a general decrease in porosity with higher pressures is observed, although the change from 5 to 8 MPa was less notable, probably because it was hard for both pieces of equipment to control this value of pressure. Again, the porosity tends to be greater as the proportion of MCC increases. Nevertheless, the most interesting behaviour, which was observed in both cases, was that the ribbons produced in the smaller-scale compactor TFC-Lab Micro showed higher porosity than those compacted on the TF-Mini, and this tendency was observed for all the compacts but the ones produced from mannitol and 15% MCC. This change on the porosity with the scale is expected as larger roll diameters result in higher densities. Two rolls presenting different diameters will have the same nip angle, which determines where the densification starts. However, for the larger roll, the volume included between the nip angle and the gap is higher than for the small roll. This means that if the same material is roll compacted for a given gap with a large roll, more volume is densified POS.047 Roll compaction of different grades of alpha-lactose monohydrate Grote, S.1; Kleinebudde, P.1 1 Institute Germany of Pharmaceutics and Biopharmaceutics, Heinrich-Heine-University, Duesseldorf, Roll compaction is an important process to improve properties of pharmaceutical formulations. As an intermediate step in tableting, properties of the resulting granules like flowability play an important role due to their effects on the subsequent steps [1]. In this study different grades of lactose (a milled grade, three primary crystal structures sieved to different sizes and one agglomerated) are investigated with regard to their resulting granules by roll compaction. The focus will be on the particle size and shape and their influence on the granules. Various formulations based on different grades of lactose (Granulac 200, Inhalac 250, Inhalac 230, Inhalac 120, Tablettose 80, Meggle, Wasserburg, Germany) were dry granulated by roll compaction (Minipactor, Gerteis, Jona, Switzerland) via binary mixtures with MCC (Vivapur 101, JRS Pharma, Rosenberg, Germany) containing 50% or 80% lactose, respectively. A specific compaction force of 5 kN/cm was applied and the gap size amounted 2 mm maintained by gap control. Smooth rolls were used combined with a rim roll sealing system and a roll speed of 3 U/min. To characterize the raw materials, particle size distribution (PSD) of the lactose was measured by laserlight scattering (Mastersizer 3000, Malvern Instruments, Malvern, Great Britain) and shape was identified by SEM (Phenom G2 pro, Phenom World, Eindhoven, Netherlands). Before determination of granules’ PSD via dynamic image analysis (Camsizer XT, Retsch GmbH, Haan Germany), samples were divided with a rotating sample divider (Retschmühle, Retsch GmbH, Haan, Germany). Finally, ffc-values were obtained by a ring shear tester to characterize flowability (RST-01, Dr. Schulze, Schüttguttechnik, Wolfenbüttel, Germany) Results show an influence of raw lactose structure especially on granules’ PSD. The primary crystals show in both cases, 50% and 80% Lactose, the highest fraction of fines followed by the milled grade. However the D(50) values of the 50% mixtures deviate from the fines and show a range from 599 µm for the milled grade over 608 to 657 µm for the primary structure up to 676 µm for the agglomerates. These results are not confirmed by the 80% mixtures where the D(50) of the milled grade, 553 µm, is placed in between the results of the primary structured grades (492 to 585 µm). Again the highest value is obtained for the agglomerated grade (619 µm). Summarising it seems that primary crystals of alpha-lactose monohydrate can only be roll compacted with an higher amount of fines although the raw material has higher D(50) values (54 to 131 µm) in comparison to the milled particles (33 µm). The agglomerated grade shows the biggest particles as raw material, D(50) 179 µm and also as granules. It may be assumed that a higher porosity of the starting material lead to harder ribbons and coarser granules. Evaluation of flowability by ffc-values shows an exceptional position of the granules produced with milled lactose. They are above the limit of 4, mixture of 50% 5.6 and mixture of 80% 4.5, which characterize a bulk material as easy flowing. All the other mixtures, except Inhalac 250 with an amount of 50% lactose (4.7), are below this limit and therefore characterized as cohesive. References: 1. Kleinebudde, P.: Eur. J. Pharm. Biopharm. 2004, 58(2): 317-326. DPhG Annual Meeting 2015 Conference Book • 131 POSTERS POS.048 Influence of different cationic lipids and DOPE on the cell uptake of lipoplexes Ziller, A.1; Hobernik, D.2; Bros, M.2; Langguth, P.1 Department of Biopharmaceutics and Pharmaceutical Technology, Johannes GutenbergUniversity, Staudingerweg 5, 55099 Mainz, Germany of Dermatology, University Medical Center of the Johannes Gutenberg- University, Langenbeckstrasse 1, 55131 Mainz, Germany 1 2 Department Personalised immunotherapies for cancer treatment are becoming more and more important. Protein structures which are only expressed on tumour cells are presented to the human immune system. Hence, the immune system generates a specific immune response, which leads to an attack against the tumour cells. One option for presentation of a protein is DNA, which codes for it and is translated in the target cells. The transport of DNA to the cells encounters several hurdles. DNases, which degrade DNA, are omnipresent in the human body. For endocytosis, DNA has to pass across negatively charged cell membranes. Therefore, carrier systems can be used to achieve a protected as well as a targeted transport of DNA [1]. In this study, a carrier system composed of liposomes and DNA, socalled lipoplexes, are tested. For preparation of lipoplexes the Dual Asymmetric Centrifuge (DAC) is applied [2]. They are composed of L-α-lysophosphatidylcholine (EPC) as basic ingredient and one kind of cationic lipid, while DNA concentration is set to 1mg/ml. Various cationic lipids and their influence on the uptake into DC-cells were studied: 20mol% of 1,2-dioleoyl-3-trimethylammonium-propane (DOTAP), 3ß-[N(N',N'-dimethylaminoethane)-carbamoyl]cholesterol (DC-Chol), Dimethyldioctadecylammonium (DDAB), 1,2-di-O-octadecenyl-3trimethylammonium propane (DOTMA) and 1,2-di-O-octadecenyl-3dimethylammonium propane (DODMA). Since 1,2-di-(9Z-octadecenoyl)sn-glycero-3-phosphoethanolamine (DOPE) as an additional component influences the uptake of DNA, DOPE is added in a molar ratio 1:1 to the cationic lipid as well [3]. Additionally, the proportion of different ingredients is varied. Size and zeta-potential of the formulations were characterised. The uptake of DNA was measured via the lucerifase assay at 24h and 72h in comparison to free DNA. Size and zeta-potential measurements show that formulation of DNA in lipoplexes is possible by achieving spherical vesicles. While the formulations with DOTAP, DC-Chol and DDAB show no significant effects, the formulations with DOTMA and DODMA cause a significant increase in the uptake in comparison to free DNA. The formulation with DOTMA and DOPE do not reach a significantly higher uptake than DOTMA without DOPE, whereas the formulation with DODMA and DOPE show even higher uptake rates. In summary, lipoplexes represent interesting delivery systems for the transport of DNA. Preparation with DAC has shown to be simple and fast, yielding applicable formulations. Working under DNase-free conditions is possible as well as very small batch sizes [2]. Hence, DAC supports the suitability of lipoplexes for individual immunotherapy. It has been shown that lipoplexes increase the uptake of DNA into cells. Moreover, the choice of cationic lipid as well as the addition of DOPE influence this uptake. Acknowledgments: BionTech AG, An der Goldgrube 12, 55131 Mainz, Germany; Lipoid GmbH, Frigenstraße 4, 67065 Ludwigshafen, Germany; Deutsche Forschungsgemeinschaft, Kennedyallee, 53175 Bonn, Germany References: 1. Wolff, J. A. et al.: Science 1990, 247(4949 Pt 1): 1465-1468. 2. Hirsch, M. et al.: J. Control. Release 2009, 135(1): 80-88. 3. Maitani, Y. et al.: Int. J. Pharm. 2007, 342(1-2): 33-39. POS.049 Impact of different drying and storage conditions on mechanical properties of orodispersible films Thabet, Y.1; Krainitzki, L.1; Dietzel, D.1; Preis, M.2 1 Institute of Pharmaceutics and Biopharmaceutics, Heinrich Heine University Duesseldorf, Universitaetsstraße 1, 40225 Duesseldorf, Germany 2 Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Artillerigatan 6A, 20520 Turku, Finland The use of orodispersible films (ODF) in pediatric therapy is gaining interest in the most recent years [1,2]. Mechanical robustness combined with rapid disintegration are crucial quality aspects of this innovative 132 • DPhG Annual Meeting 2015 Conference Book dosage form. The present study aimed to investigate whether different drying (room temperature (RT), 40 °C and 80 °C) and storage conditions (open, sealed in aluminum sachets, humid conditions) following the production of solvent-casting films influence the film properties. Hypromellose (HPMC), hydroxypropylcellulose (HPC) and polyvinyl alcohol (PVA) were used as film forming agents. Metamizole sodium salt, an analgetic, antipyretic and antispasmic drug, served as model drug. Films with and without drug load were prepared and subsequently their morphological, mechanical and disintegration properties were investigated. All produced HPMC films lead to transparent, flexible films. After storage, HPMC films containing metamizole show a temperature- dependent recrystallization. The higher the drying temperatures, the less crystals could be detected (by means of polarized light microscopy). HPC films dried at 80 °C lead to opaque, yellowish films, which has been described with regard to drying procedures applied to HPC and could be explained by the limited water solubility of HPC at higher temperatures [3]. ODFs with PVA as film forming polymer result in transparent, flexible films which also show a recrystallization after storage time. All films disintegrated rapidly within approximately 10-20 s using a modified disintegration tester with a particular film sample holder [4]. Mechanical testing determining the puncture strength and elongation of the samples, revealed the sensitivity to varying drying temperatures. The mechanical strength of the drug-loaded films decreased with increasing temperatures, e.g. for HPMC (RT: 1.67±0.41 N/mm2, 40 °C: 1.59±0.37 N/mm2, 80 °C: 0.21±0.06 N/mm2). Subsequent storage at 58.5% relative humidity for four weeks did not alter the mechanical properties (RT: 1.53±0.16 N/mm2, 40 °C: 1.55±0.19 N/mm2, 80 °C: 0.24±0.05 N/mm2). Furthermore, higher drying temperature lead to less flexible films. The drug-load on the one side increased the strength of the films, but decreased their flexibility on the other side. This study revealed that depending on polymer and drug properties certain drying conditions are favorable in the manufacturing of orodispersible films, but need to be carefully evaluated case by case. References: 1. Hoffmannn, E .M.; Breitenbach, A.; Breitkreutz, J.: Exp.Opin. Drug Deliv. 2011, 8(3): 299-316. 2. Preis, M et al.: AAPS Pharm Sci Tech. 2015, 16(2): 234-241. 3. Klug, E.D.: J. Polymer Sci.:Partc C 1971, 36(1): 491-508. 4. Beilke, D.; Preis, M.: TechnoPharm. 2014, 4(6): 334-337. POS.050 Application of numerical simulations in pharmaceutical tableting Hildebrandt, C.1; Gopireddy, S. R.2; Scherließ, R.1; Urbanetz, N. A.2 1 Department of Pharmaceutics and Biopharmaceutics, Kiel University, Grasweg 9a, 24118 Kiel, Germany 2 Daiichi-Sankyo Europe GmbH, Pharmaceutical Development, Luitpoldstrasse 1, 85276 Pfaffenhofen, Germany Even though alternative forms of pharmaceuticals exist, tablets remain the most common and preferred dosage form to date due to its stability and ease of administration. With the huge volume of tablet production, analytical tools are in high demand as they are used to control the final product quality as per the regulatory requirements. Besides process analytical tools (PAT), the pharmaceutical industry looks for alternative solutions such as numerical simulations in formulation development and product quality control. In this study, such a numerical approach is presented with the focus on the investigation of powder flow within a tablet press as well as content uniformity in dies thereby increasing the process understanding of this part of the tableting procedure and to impact product quality in a positive way. The powder flow within the tablet press, i.e. from the force feeder to the dies, depends on material characteristics on the one hand and process conditions on the other hand. The feed frame often contains a number of paddle wheels which convey the powder from the hopper outlet over the die opening (see Figure). In this study, particle micro-dynamics are computed through the three-dimensional discrete element method (DEM). The DEM captures individual particle trajectories through Newton’s equations of motion, which provide dynamic particle scale information. The considered particle characteristics correspond to microcrystalline cellulose which is a well-known pharmaceutical tablet excipient. The numerical model is applied to a lab-scale tablet press force feeder to predict the effect of feeder paddle wheel speed as well as rotating speed of the die table on the inter-tablet variability. PHARMACEUTICAL TECHNOLOGY AND DRUG FORMULATIONS The preliminary simulation results show that increasing the paddle wheel speed at constant die table speed leads to increased inter mixing of powder within the force feeder and simultaneously, the amount of powder mass collected in the dies is decreasing. This is due to the fact that with increased paddle wheel speed the net force acting on particles in the radial direction within the force feeder increases and it is higher than the gravitational force. Furthermore, results indicate that at lower paddle wheel speed the powder tends to segregate in case of poly-disperse particle size, i.e. the fine particles settle down at the bottom whereas the large size particles rise to the top of the feeder. This may cause content variation between the dies when using multicomponent mixtures. This inter-tablet variability can cause wrong dosing for the patients. Numerical results also show that the stresses acting on the feeder walls, which apply shear stress on the powder thereby affecting its flowability and lubrication, are influenced by the paddle wheel speed. A A B POS.052 The effect of material properties and process parameters on roll compacted ribbons Csordás, K.1; Kleinebudde, P.1 Side view Top view high luciferase and EGFP activity, respectively. Moreover biological activity remains high even after storage up to three months at room temperature. We also observed PVA microparticles to be dependent on the manufacturing process. In addition, tested formulations were shown to be nontoxic. Taken together, this indicates that PVA-MP may represent a promising system for long term storage and controlled release of PEI-based polyplexes and lipopolyplexes. B A: Force feeder with paddle wheel and hopper B: Die table Simulation of powder flow in the force feeder of a lab scale tablet prodcution press (RoTab, kg-pharma). Poly-disperse, spherical particles (in total 1,500,000) with a mean diameter of 453 µm were simulated. The material properties correspond to microcrystalline cellulose. The paddle wheel and die table speed are both 30 rpm. The current simulation study helps in visualizing the powder flow from the feeder to the die, which gives rise to better understanding of the die filling process. The results show that the particle micro-dynamics are significantly influenced by the material and process conditions. To fulfil the requirements of the authorities and to ensure product quality, process simulations can help in identifying the critical attributes of the tableting unit operation whereas experimental and analytical approaches could not consider features such as quantification of the shear stress within the force feeder. In conclusion such numerical studies will support the product quality control concurrently with established means of pharmaceutical development. POS.051 Encapsulation of PEI-based nanoparticles into a poly(vinyl alcohol) matrix – a new application for an old excipient Schulze, J.1; Aigner, A.1 1 Rudolf Boehm Institute for Pharmacology and Toxicology, Clinical Pharmacology, University of Leipzig, Leipzig, Germany The nucleic acid based treatment of diseases offers great opportunities, even in the case of targets considered as otherwise undruggable. Due to its potential applications, the interest in gene therapy, and consequently in gene delivery, has grown continuously over the past years. The development of non-viral delivery systems is under intense investigation and has led to various lipid and polymeric excipients. Among those, polyethylenimine (PEI) is a promising candidate due to its high biological activity. The combination of this cationic polymer with a lipid delivery system may further improve properties of formed nanoparticles by combining the beneficial features of a lipid system (low cytotoxicity, high stability) and the advantages of PEI. The establishment of systems for their controlled delivery and controlled release remains another formidable challenge. Here we describe the fabrication and extensive characterization of a controlled release system for PEI-based nanoparticles. The purpose of this study was to evaluate if the combination of nanoscale polyplexes or lipopolyplexes with a hydrophilic polymer will yield an appropriate carrier system. To this end, we incorporated PEI-based nanoparticles into poly(vinyl alcohol) microparticles (PVA-MP). This feasibility study aimed at investigating whether high transfection efficacy, zeta-potential and particle size of embedded nanoparticles remain unchanged during the manufacturing process and storage and upon release. We show that polyplexes as well as lipopolyplexes released from PVA microparticles do not change in size compared to unformulated nanoparticles, while a profound alteration of the zeta potential was observed. Furthermore we demonstrate the released nanoparticles to be taken up by cells and their DNA payload to be transcribed, yielding in Institute of Pharmaceutics and Biopharmaceutics, Heinrich Heine University, 40225 Duesseldorf, Germany 1 Roll compaction/dry granulation is a widely used agglomeration process in the pharmaceutical industry. The powder is compacted by applying a defined force resulting in long shaped or debris ribbons. The ribbons are milled into granules to achieve desired granule size. Ribbon porosity is one of the critical quality attributes of roll compaction process, because it is closely related to the mechanical properties of granules, thus affects the downstream processes e.g.: tableting [1]. The goal of this work was to investigate the impact of material properties and process parameters on roll compacted ribbons. The average ribbon porosity and universal hardness were determined. Microcrystalline cellulose (MCC) (Avicel PH 101 FMC BioPolymer, USA), spray-dried mannitol (Pearlitol 200 SD, Roquette, France) and the 1:1 mixture of them were roll compacted using AlexanderWerk BT 120 (AlexanderWerk AG, Germany) roll compactor. True density of the starting materials was determined by AccuPyc 1330 Helium Pycnometer (Micromeritics, USA). Ribbons were manufactured using side-sealing system and a pair of smooth rolls. The hydraulic pressure (HP) was set at 18 bar, 24 bar, 36 bar, 48 bar and 60 bar; the gap width (GW) was adjusted at 1.5 mm and 2.3 mm. The roll speed was kept constant at 3 rpm. For each material 13 batches were prepared in total according to a full factorial design of experiment (Modde 9.0, Umetrics, Sweden). The intermediate products, called ribbons were collected and their average porosity was measured by GeoPyc 1360 (Micromeritics, USA) in a 25.4 mm diameter chamber using a consolidation force of 51.0 N. DryFlo was used as medium to obtain the measurements (n=3). The universal hardness was determined using Fischerscope HM 2000 (Helmut Fischer, Sindelfingen, Germany) excerting 1000 mN indentation force in 20 s in the middle along the length of the ribbon (n=15). Good models are observed for ribbon microhardness (R2= 0.88) and porosity (R2= 0.96). Hydraulic pressure and gap width are found to be significant quantitative factors for both responses. Ribbon microhardness increases with increased hydraulic pressure. The contrary is observed in case of porosity: the higher HP is set, the lower porosity is measured. Mannitol ribbons represent a higher increase of the microhardness than MCC ribbons using the same parameter settings. It can be explained by the breakage of the brittle mannitol particles during feeding into the nip area and the compaction between the two counter-rotating rolls, thus more particles could be densified [2]. The microhardness of the 1.5 mm thin mixture ribbons are between the microhardness values of MCC and mannitol ribbons, as expected. 2.3 mm thick mixture ribbons are produced solely using 18 and 36 bar HP, because of the low surface friction between the powder blend and roll surface. Compacting mannitol using 48 bar HP and 1.5 mm GW causes the minimum porosity value (9%), whereas the same parameter setting results in 15% porosity manufacturing with MCC. This difference is observed due to the plasticelastic recovery of MCC after roll compaction. The most porous ribbons (34%) are produced using MCC and setting 18 bar and 24 bar HP regardless of GW. However, compacting with 36 bar and 48 bar HP 5% increase of porosity is achieved, when the GW is increased from 1.5 mm to 2.3 mm. Acknowledgments: ”Roll compaction using AlexanderWerk BT 120 and the envelope density measurements were supported by Astra Zeneca Ltd. Macclesfield, UK.; This work was supported by the IPROCOM Marie Curie initial training network, funded through the People Programme (Marie Curie Actions) of the European Union’s Seventh Framework Programme FP7/2007-2013/ under REA grant agreement No. 316555.” References: 1. N. Souihi et al.: Intern. J. Pharm. 2015, 484: 192-206. 2. Y. Teng, Z. Qui, H. Wen: Eur. J. of Pharm. and Biopharm. 2009, 73: 219-229. DPhG Annual Meeting 2015 Conference Book • 133 POSTERS POS.053 Excipient effect on colloidal structure and BCS-II drug solubility in bicarbonate based intestinal model medium FaSSIFmod6.5 and cholesterol containing biorelevant intestinal medium (FaSSIF-7C) Khoshakhlagh, P.1; Johnson, R.1; Nawroth, T.1; Langguth, P.1; Schmueser, L.2; Hellmann, N.2; Decker, H.2; Szekely, N. K.3 1 Pharmacy and Biochemistry Institute, Division of Pharmaceutical Technology, Johannes Gutenberg University, Staudingerweg 5, D-55099 Mainz, Germany 2 Molecular Biophysics Institute, Johannes Gutenberg University, Jakob Welder Weg 26, D55128 Mainz, Germany 3 JCNS-FRM-II Outstation at the MLZ, Lichtenbergstraße 1, D-85747 Garching Surfactants are commonly used in drug formulations for increasing drug solubility in case of hydrophobic APIs. They can have interaction with transient colloidal structures of the bile and food in the intestine, which are develop from micelles (5-10 nm) to liposomes (30-200 nm) and large cholesteric particles (up to 50 µm). This study investigated the effect of some model surfactants (Tween 80, Tween 20, decyl sulfate and dodecyl sulfate) on the solubility of Fenofibrate in bicarbonate model medium FaSSIFmod6.5 [1] and a novel cholesterol containing biorelevant medium, FaSSIF-7C [2]. In parallel the structural development was investigated with dynamic light scattering DLS and neutron small angle scattering SANS (static and time resolved). The results indicated that the solubility of Fenofibrate increased by a factor of four with the highest concentration of Tween 80 to a two-fold reduction with the maximum concentration of Tween 20. DLS data showed that increasing surfactant concentration caused destruction of the native liposome structure but only at high detergent concentration increased the formation of surfactant rich micelles in the medium, which can resolve the API. In the intermediate surfactant regime a drug solubility gap was observed. With cholesterol containing media, the existence range of the liposomes was increased (stabilized), while the solubility gap was observed at increased surfactant concentrations (shift). The neutron study indicated a co-existence of micelles and liposomes, and a developing of the liposome entity during the development of FaSSIF-C in the first hour after dilution of FeSSIF with the transfer medium representing the fluid input from gastric emptying. According to the results only a few surfactants (based on the type and concentration) have a stabilizing effect on the colloidal structure and solubility of hydrophobic drugs in biorelevant media. Lipophilc drug (BCS-II, Feno) incorporates in different nano-structures in the presence of surfactant excipients. As small scale feeder a MiniTwin Feeder (DDW-MD1-MT-1HD, Brabender Pharma, Germany) was used and to compare it to bigger scales, a KT 20 (K-Tron, Switzerland) and a Flexwall (Brabender Pharma, Germany) were used. The actual feed-rates, were taken from the feeder controllers. To evaluate the feeder performance offline, different feed-rates were set and powder was fed onto an external scale. Change in weight was recorded with a frequency of 5 Hz and the actual feed-rates were calculated. Feed-rates from 0.5 to 10 g/min were investigated. Mean feed-rate and coefficient of variation (CV) were employed as quality criteria. For all of the used feeders, feed-rates and materials, the internal controllers of the powder feeders reported accurate feeding, as they usually displayed actual feed-rate values, which were close to the set values. Thus, the results of the feed-rate data, given by the balance was analysed more in detail. At feed-rates of 6 and 10 g/min and the use of maize starch the mini-twin was superior to the bigger scaled feeders regarding precision. At 6 g/min it featured a CV of only 34% compared to the KT-20 and the Flexwall with 114% and 134%. In the case of accuracy the KT20 feeder showed the best results. At 6 g/min and 10 g/min it almost exactly met the set value, independent from the applied powder, compared to 6.18 g/min (Mini twin, maize starch) and 5.63 g/min (Flexwall, maize starch). At low feed-rates of 0.5 g/min to 4 g/min the Mini-twin was capable of feeding the free flowing mannitol very precise with CV of around 40% compared to CV for the cohesive maize starch from 243% (0.5 g/min) to 94% (4 g/min). However, despite those good precision values, the feeder was not able to achieve an accurate feeding result for the mannitol, as it always resulted in values of excessive fed powder. The reason for this could be seen in the good flowing properties of the mannitol. Additionally to the feeding, caused by the screw revolutions the powder freely flushed out of the feeder, resulting in feed-rates higher than the set values. This problem may be solved by the utilization of a screen, as it was successfully done by Engisch and Muzzio [1]. The feeding of maize starch always resulted in accuracy values, which were closer to the set value, compared to the feeding of mannitol. The results of this study underline the need of evaluating powder-feeder performances offline and separated from the data, given by the feeder, because it is not advisable to trust into these values. Furthermore, it is important to get control over the feeding systems, by choosing the appropriate equipment for the set powders and feed-rates. References: 1. Engisch, W. E.; Muzzio, F. J.: J. Pharm. Innov. 2015, 10: 56-75. Acknowledgments: We are grateful for the funding by the OrBiTo Eu-project 2013-2018, and JCNS. References: POS.055 1. Buch P. et al.: J. Pharm. Sci. 2010, 99(10): 4427–4436 2. Khoshakhlagh P. et al.: J. Pharm. Sci. 2015, 104(7): 2213-2224. Elucidating the release mechanism of a poorly soluble drug from amorphous solid dispersions by in situ Raman imaging POS.054 Vukosavljevic, B.1,2; Punčochová, K.3; Štěpánek, F.3; Windbergs, M.1,2,4 Institute of Pharmaceutics and Biopharmaceutics, Heinrich-Heine-University, Universitaetsstr. 1, 40225 Duesseldorf, Germany of Biopharmaceutics and Pharmaceutical Technology, Saarland University, Campus A 4.1, 66123 Saarbruecken, Germany 2 Helmholtz Centre for Infection Research (HZI) and Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), Department of Drug Delivery, Campus A 4.1, 66123 Saarbruecken, Germany 3 Department of Chemical Engineering, University of Chemistry and Technology Prague, Technická 3, 166 28 Prague 6, Czech Republic 4 PharmBioTec GmbH, Science Park 1, 66123 Saarbruecken, Germany Several continuous manufacturing processes, such as twin screw extrusion or -granulation, coating and continuous milling require accurate and precise feeding systems. In early pharmaceutical development such as preformulation studies, only minor amounts of drugs are available, which results in the need of small scale equipment. Especially from the handling of powders the need arises to use gravimetrically controlled feeders. Thus, inhomogeneities caused by consolidation of the powder through normal forces, bridge building and other processes that may cause differences in bulk density can be minimized. These problems get even worse in small scale processes. The aim of this study was to compare different powder feeders regarding precision and accuracy, while feeding a cohesive and a free flowing powder. Mannitol (Parteck M 100, Merck, Germany) was used as a free flowing powder and maize starch (CPharm 03406, Cerestar, Germany) as a poorly flowing powder. Solid dispersions based on hydrophilic polymers are valuable carrier systems for the effective delivery of poorly soluble drugs. For rational development of such systems, the drug release mechanism has to be elucidated. In this study, we analysed the release behaviour of aprepitant, a poorly water soluble drug embedded within two different solid dispersions. Solid dispersions were fabricated based on either Soluplus® or on polyvinylpyrrolidone K30 (PVP) by spray drying (Mini Spray Dryer B-290, Büchi, Switzerland, drug:polymer 1:3 w/w, on a dry basis). In a next step, the spray-dried particles were compressed to tablets. We performed modulated temperature differential scanning calorimetry to determine the glass transitions temperature. One single glass transition temperature for both formulations confirmed that the drug was molecularly dispersed, and hence a solid solution was formed. The glass transition temperatures of pure Soluplus, Soluplus:Aprepitant mixture, Performance evaluation of powder feeders – small scale feeding as a challenge Meier, R.1; Kleinebudde, P.1 1 134 • DPhG Annual Meeting 2015 Conference Book 1 Department PHARMACEUTICAL TECHNOLOGY AND DRUG FORMULATIONS PVP and Aprepitant:PVP mixture were 68.2, 57.8, 159.5 and 142.9 °C, respectively. Raman spectra of the amorphous solid dispersions show distinctive peaks indicating the presence of the drug and polymer, without any evidence of crystalline aprepitant. Further, CRM allows for distribution analysis of aprepitant and polymer within the tablets. In addition, we visualized distribution changes within the amorphous solid dispersions during dissolution by CRM. Upon dissolution, swelling and gel layer formation influence diffusion and release of the drug. In order to preserve intactness of the gel-layer during the analysis, we applied virtual cross section imaging (x/z scans) with around 100 µm depth through the gellayer. The Soluplus:Aprepitant formulation exhibits a continuous release with no phase separation and drug precipitation observed. Based on these results, Soluplus could be identified as a crucial factor for stabilization of the amorphous drug. The PVP:Aprepitant formulation shows a different release mechanism. Firstly, due to the good affinity of PVP to water, the polymer dissolves very fast from the matrix, thus a highly hydrated gel layer is formed on the surface. However, after approx. 30 minutes, under a viscous PVP layer, the drug recrystallizes (depicted in pink) on the surface of the tablet as depicted in Figure. New approaches for in situ characterization and elucidation of drug release from solid dispersions are needed. In this context, CRM is an upcoming technique for non-invasive and chemically selective elucidation of release mechanisms of poorly soluble drugs from solid solutions. compressive strengths of up to 33±2 MPa and with a pore structure that allows cells to grow deep into the scaffolds and form mineral deposits. Compressive moduli between 27±7 MPa and 568±98 MPa were obtained depending on the hybrid composition and problems associated with the inherent brittleness of sol-gel glass materials could be overcome. SaOS-2 cells showed strong mineral accumulation on hybrid glass scaffolds as early as day 7. On day 14, we also found mineral accumulation on control scaffolds without cells indicating a positive effect of the hybrid glass on mineral accumulation. We fabricated macroporous hybrid glass scaffolds using rapid prototype templates. By using this templating technique, we were able to produce hybrids of a wide variety of compositions. In addition, the templating technique allows for modification of the scaffolds’ pore size and geometry as well as for an individualization of size and shape of the implant, with a high reproducibility. Acknowledgments: The authors thank Prof. Rieger (HTWK Leipzig, Germany) for access to the compression testing equipment, Dr. Anderegg (University hospital of Leipzig) for support in the preparation of histosections and Jörg Lenzner (University of Leipzig) for access to the electron microscope. The authors would also like to thank the AiF Projekt GmbH (Grant no: KF2734502MU1), the Saxon ministry for science and arts (Grand no: 4-7531.60/64/18) and the German Research Council (DFG SFB/Transregio 67) for financial support. References: 1. Jones, J.; Ehrenfried, L.; Hench, L.: Biomaterials 2006, 27(7): 964-973. POS.057 Towards a new bacterial-epithelial cell co-culture model to test novel drug delivery systems against Pseudomonas aeruginosa biofilms Juntke, J.1; Türeli, N. G.2; de Souza Carvalho-Wodarz, C.1; SchneiderDaum, N.1; Lehr, C. M.1 Drug Delivery, Helmholtz Institute for Pharmaceutical Research Saarland, 66123 Saarbrücken, Germany 2 MJR PharmJet GmbH, 66424 Homburg, Germany 1 Figure: Virtual cross section (x/z scan) of the PVP:Aprepitant tablet formulation during dissolution after 30 min. Gel layer is depicted in light blue, crystalline drug in pink and inner tablet in blue. POS.056 Hybrid glass scaffolds for bone regeneration by indirect rapid prototyping Hendrikx, S.1; Kascholke, C.1; Flath, T.2; Schumann, D.3; Gressenbuch, M.4; Schulze, P.2; Hacker, M. C.1; Schulz-Siegmund, M.1 Pharmaceutical Technology, Institute of Pharmacy, Universität Leipzig, Leipzig, 04317, Germany 2 Faculty of Mechanical and Energy Engineering, Leipzig University of Applied Sciences, Leipzig, 04277, Germany 3 Bubbles and Beyond GmbH, Leipzig, 04229, Germany 4 DMG Chemie GmbH, Leipzig, 04347, Germany 1 Implants for bone regeneration have to withstand mechanical stress on the one hand and, on the other hand, provide high macroporosity to allow for tissue ingrowth, remodelling and vascularization, making the fabrication a complex task. Moreover, materials need to be biocompatible and bioactive with regards to the formation of a mechanically stable bone-implant interface. Last but not least the production costs of these implants should be reasonable and the technology should allow for easy up-scaling at reproducible product quality [1]. We present a series of hybrid sol-gel derived bioactive glasses with oligovalent organic cross-linkers functionalized with 3Isocyanatopropyltriethoxysilane, which were susceptible to heat sterilization. The hybrids were processed into pore-interconnected scaffolds by an indirect rapid prototyping method. A large panel of 2- to 4-armed cross-linkers of different molecular weight were incorporated and their effect on scaffold mechanical properties was investigated. By multiple linear regression, ‘organic content’ and the ‘content of ethylene oxide units in the hybrid’ were identified as the main factors that determined compressive strength and modulus, respectively. In general, 3- and 4-armed cross-linkers performed better than linear molecules. Compression tests and cell culture experiments with osteoblast-like SaOS-2 cells showed that macroporous scaffolds can be produced with In the lungs of cystic fibrosis (CF) patients, the thick and sticky mucus offers good growth conditions for bacteria. Especially Pseudomonas aeruginosa, known as the major pathogen in CF, is almost impossible to eradicate once a biofilm is formed. An intriguing approach to overcome drug delivery problems, arising from the thick mucus layer and the biofilm, is the development of nano-scaled drug delivery systems (DDS). For this reason, ciprofloxacin-complex-loaded PLGA nanoparticles (NP) are prepared under controlled conditions by nanoprecipitation using the novel MicroJet Reactor technology. The NP should be able to penetrate the mucus and biofilm thus releasing the drug in a controlled manner. To mimic disease relevant conditions in vitro, our aim is to establish a coculture model of human bronchial epithelial cells with P. aeruginosa on top, allowing the formation of a biofilm. The bronchial epithelial cell lines Calu-3 and CFBE41o- are used, representing the healthy and CFdiseased airways, respectively. Once the cells have formed an epithelial barrier they are infected with different amounts of P. aeruginosa to identify a critical seeding density of the bacteria which enables biofilm formation and still allows the human cells to survive. This model holds potential for evaluating novel DDS regarding their safety and their ability to disrupt the bacterial biofilm and to reach P. aeruginosa. Acknowledgments: Thanks to the BMBF for funding this KMU-innovativ project : „FiDel“ FKZ: 13N12530 POS.058 A unified in vitro test system for tight junction modulators Saaber, D.1; Reichl, S.1 Institut für Pharmazeutische Technologie, Technische Universität Carolo-Wilhelmina zu Braunschweig, Mendelssohnstr. 1, 38106 Braunschweig, Germany 1 Tight junctions (TJ) consist of different transmembrane proteins and are located in the paracellular space. In addition to effects on cell proliferation and differentiation they contribute to absorption barrier of epithelial and endothelial cells for many drug substances. Therefore modulation of TJ DPhG Annual Meeting 2015 Conference Book • 135 POSTERS barrier function is considered to be a promising approach for drug permeation enhancement and to improve bioavailability of BCS class III/IV drugs. Some substances influencing TJ barrier, so-called TJ modulators of first generation, have been reported over the last two decades, but based on better knowledge and comprehension of TJ regulation several mechanism-based (MB) TJ modulators have been developed in the past few years [1]. In contrast to first generation modulators they should show less cytotoxic effects and allow regeneration of barrier properties, determined as transepithelial electrical resistance (TEER), when application is finished and cells are incubated with modulator free medium. However, comparison of described TJ modulators as well as evaluation of usefulness and toxicity is difficult because most studies include different cell lines and examination methods. The purpose of this study was to standardize the characterization of TJ modulators and to develop a unified test system to assess TJ modulators for pharmaceutical applications. In our study MDCK I cells were used, exhibiting high and constant TEER level of 3,000-3,500 Ω·cm² for several days, 5 days after seeding. Evaluation of the TJ modulating effect was performed by continuous impedance measurement under standardized conditions using cellZscope®. Cell viability was tested by MTT assay. For determination of membrane interference fluorescent micrographs were taken after 3-fold staining with DAPI, Hoechst 33342 and calcein-AM. Overall eight different TJ modulators, five of first generation like EDTA, sodium caprate, sodium nitroprusside, benzalkonium chloride, sodium fluoride, and three MB modulators like AT-1002, PN159 and labradimil were tested. In principle, first generation as well as MB modulators, were able to cause significant TEER reduction up to 100% within the first 30 min of incubation. Only for labradimil smaller TEER reductions of 17% for 1 µM and 27% for 10 µM were detected in our experimental setup. However, effective doses of first generation modulators induced lower cell viability in comparison to MB modulators. AT-1002 and PN159 caused fast TEER reduction within the first 15 min after exposure. Furthermore, the TEER regeneration after AT-1002 and PN159 incubation was higher and faster than observed for first generation modulators. In the case of AT-1002 TEER regenerated after 60 min and reached a maximum of 70% in comparison to the control 15 h after substance depletion. For PN159 regeneration started after 90 min and reaches 100% after 11 h. Interestingly, MDCK I cells exhibited spontaneous regeneration of TEER approx. 60 min after incubation with 100 µM sodium caprate and 10 mM sodium fluoride. For sodium caprate and AT-1002 similar mechanisms of action have been reported, resulting in an activation of proteinkinase C [2,3]. A unified test system has been developed to compare efficacy and toxicity of various TJ modulators. This study showed that MB TJ modulators can cause significant TEER reduction and exhibit less toxicity compared to first generation TJ modulators. The test system will be extended by permeation data and membrane integrity assays. The final system will offer the opportunity to characterize and evaluate the potential of new developed TJ modulators. powder discharge from these containers also depends on the material characteristics, so the characteristics of discharge may vary between formulations possibly leading to powder segregation. To evaluate the variation in the composition of the discharged powder, most commonly NIR techniques are used, which not only require modification of the instrument but also a lot of calibration effort. Alternatively, numerical simulations offer in-depth understanding of the powder dynamics within these containers. In this study, such simulations are implemented to investigate the rate of discharge and segregation during discharge from a production size container. The simulations are carried out using the three-dimensional discrete element method, which computes the trajectories of each and every particle. Usually, the production size container can hold few hundred kilograms of material, which means several billions of few hundred micron sized particles to be accounted for in these simulations. Handling such amounts of material is nearly impossible with the current computational capacity. To reduce the number of particles for the simulations, the container is downsized to a scale of 1:10. The effect of downsizing is computed and validated on the one hand side by simulating the downsized scale of 1:7.5 and on the other hand side with an experiment. The materials considered in this study include VIVAPUR MCC spheres (JRS Pharma GmbH) and Ludipress (BASF Ludwigshafen), which have poly-disperse particle sizes. The material properties such as particle size distribution, density and flowability are measured and entered as input parameters in simulations. After having established and validated the desired downsized geometry i.e. 1:10 scale, the rate of powder discharge is computed. One example of the simulation results is displayed in the figure below. The green line shows the number of particles which have already exited the container. The red line is the mean diameter of the remaining particles. The diagram shows that at the beginning the mean particle size does not change significantly, but after discharge of about 80% of the initially loaded material the particle size starts to decrease indicating segregation. This variation in the particle size with time is of special interest as it is usually the drug particles having the smallest size in a mixture, meaning that the discharge of a mixture would lead to a segregation according to the particle size of the components. The study also investigates the influence of material properties such as cohesion on the discharge rate, dynamic flow angles, flow modes (funnel flow or mass flow). In conclusion, the present study helps in better understanding of the powder dynamics within a container during discharge, can help in identifying sources of poor content uniformity and may be used to identify critical material attributes. References: 1. Saaber et al.: Expert. Opin. Drug Discov. 2014, 9(4): 367-81. 2. Tomita, M.; Hayashi, M.; Awazu, S.: J. Pharm. Sci. 1996, 85(6): 608-11. 3. Goldblum et al.: FASEB J. 2011, 25(1): 144-58. POS.059 Numerical investigation of uniformity of powder discharged from a production container Rötzer, K. M.1,2; Gopireddy, S. R.2; Schlosser, E.1; Urbanetz, N. A.2 1 Lehrstuhl fuer Verfahrenstechnik disperser Systeme, Technische Universitaet Muenchen, Maximus-von-Imhof-Forum 2, 85354 Freising, Germany 2 Daiichi-Sankyo Europe GmbH, Pharmaceutical Development, Luitpoldstrasse 1, 85276 Pfaffenhofen, Germany In solid dosage form production, similar containers or bins are used for handling materials in different unit operations as well as in intermediate steps, e.g., the bin used in mixing the drug and excipients is also used in tabletting unit operation or vice versa. This interchangeability allows to have less types of containers, and enables to handle similar scales of material in every unit operation. When such containers are used in tableting, they are designed to discharge the bulk solid homogenously and uninterruptedly. However, besides the container geometry the 136 • DPhG Annual Meeting 2015 Conference Book POS.060 Biorelevant in vitro dissolution testing of oily suspensions for intramuscular administration Probst, M.1; Schmidt, M.1; Seidlitz, A.1; Weitschies, W.1 1 University Germany of Greifswald, Institute of Pharmacy, Felix-Hausdorffstr. 3, 17489 Greifswald, It was the aim of the presented study to test and develop biorelevant in vitro dissolution test setups for intramuscularly applied oily suspensions. For the generation of in vivo data an oily suspension of prednisolone (2.5 mg suspended in 100 µL medium chain triglycerides) was injected into the muscle tissue of the thigh of rats. The volume and surface area of the depots were determined and pharmacokinetic data were obtained. The same formulation was investigated in vitro by adapting the results from the volume and surface area measurements to the test setups as possible. PHARMACEUTICAL TECHNOLOGY AND DRUG FORMULATIONS In one test setup, adapted from Bhardwaj and co-workers [1], the compendial flow through cell (FTC) was combined with a dialysis method. For this purpose a dialysis tube (MWCO 50 kDa, regenerated cellulose) was mounted on an adapter which was placed into the FTC. The suspension was injected into the dialysis tube and the FTC was perfused with PBS pH 7.4 (Figure A). The dimensions of the adapter and the FTC were modulated to simulate the obtained in vivo data of the shape of the depot. This whole setup was either placed in upright or in horizontal position. Additionally, a new in vitro dissolution test method was performed. Here, a wire rack was placed in a compendial FTC which was filled with PBS. The oily suspension was injected into the release medium. The wire rack prevented the removal of the oily droplet (Figure B). Subsequently, the cell was perfused with the medium while the oily droplet was kept in position. The results of these test methods were compared to the compendial paddle apparatus and the reciprocating holder. In the latter, the dialysis tube was fixed in an adapter and reciprocated within the release medium. The results (Figure C) showed that the testing method had a major influence on the release of prednisolone. The release of the oily suspension of prednisolone in the paddle apparatus was by far the fastest. The dissolution of prednisolone from the oily droplet was faster as in the test setups using the dialysis bag in the FTC but considerably slower than in the paddle apparatus. The positioning of the FTC with the dialysis bag had a major influence on the release behaviour. The reciprocating cylinder and the dialysis bag in upright position showed a similar release profile being the slowest compared to the other tested methods. Compared to the pharmacokinetic profiles observed in the rats where cmax occurred after 6 h in median, the release from the oily droplet in the FTC corresponds best to the in vivo observation. System (DynaMiTES) within the scope of the Center of Pharmaceutical Engineering (PVZ) of the TU Braunschweig. This system should allow dynamic donor and acceptor control, continuous impedance measurement and the application of widely used cell culture inserts. These advantages would enable the application of the novel dynamic in vitro test system with various existing cell culture models and facilitate its transfer to other laboratories. To approach this aim, at first an established and prevalidated human hemicornea model (classic HC; [2], [3]) was modified. The classic HC had to be altered into an inverse HC model for the use in the newly designed DynaMiTES. In the present study various cell characteristics were compared in both models to prove their equivalence. For this comparison the in vitro models were cultivated under serum-free conditions on permeable polycarbonate filters (Transwells®) using SV40 immortalized human keratocytes (HCK-Ca) and human corneal epithelial cells (HCE-T). The classic HC was cultivated as described before [2]. In contrast to this, for the inverse HC the HCE-Ts were seeded on the external side of the insert’s membrane. After seven days of submerse cultivation, both models were lifted to air-liquid interface until day ten. Barrier properties of the HCs were estimated by transepithelial electrical resistance (TEER) measurements and static permeability studies with sodium fluorescein. Cell viability was evaluated via MTT testing. Cell morphology was investigated by cross-sectional slices (Technovit® 7100) stained with haematoxylin and eosin. Although the compared models showed different TEER developments, neither the TEER values nor the apparent permeation coefficients (Papp) were different at the end of cultivation. The obtained Papp was similar to the values presented in the prevalidation study [3]. Moreover, cell viability after cultivation was not statistically different between both groups. The histologic observations could support this equivalence by morphologic similarity. The inverse HC developed a multi-layered epithelium similar to the classic HC and human cornea. These findings underline that the modification of existing in vitro models for the DynaMiTES is possible and has been performed successfully for the prevalidated HC. Thus, it can be assumed that other established and well characterized in vitro models can also be adapted to the DynaMiTES. In subsequent investigations the inverse HC shall be used within the DynaMiTES to simulate tear dynamics and to investigate their effects. This ocular DynaMiTES will then be one important improvement of current in vitro models for a better prediction of safety and efficacy data. References: 1. Marx, U. et al.: ATLA Altern. Lab. Anim., 2012, 40: 235–257. 2. Hahne, M.; Reichl, S.:Int. J. Pharm. , 2011: 416, 268–279. 3. Hahne, M. et al.: J. Pharm. Sci., 2012, 101: 2976–2988. References: 1. Bhardwaj, U.; Burgess, D.: Int. J. Pharm. 2010, 388: 287-294 POS.061 Modification of an organotypic human hemicornea model for the use in a novel Dynamic Micro Tissue Engineering System (DynaMiTES) Beißner, N.1,3; Mattern, K.2,3; Dietzel, A.2,3; Reichl, S.1,3 1 Institut für Pharmazeutische Technologie, Technische Universität Carolo-Wilhelmina zu Braunschweig, Mendelssohnstraße 1, 38106 Braunschweig, Germany 2 Institut für Mikrotechnik, Technische Universität Carolo-Wilhelmina zu Braunschweig, Alte Salzdahlumer Str. 203, 38124 Braunschweig, Germany 3 Zentrum für Pharmaverfahrenstechnik, Technische Universität Carolo-Wilhelmina zu Braunschweig, Franz-Liszt-Straße 35 A, 38106 Braunschweig, Germany Preclinical drug absorption studies are frequently performed using ex vivo tissue or in vitro cell culture models. The bioavailability of topically applied drugs is often low since at various physiological barriers like the eye or the nose drugs are drained away within minutes. However, current in vitro tests for the assessment of pharmacokinetic and toxicological parameters are mostly performed in static cell culture systems, which cannot emulate the physiological conditions of drug administration. This essential disparity may result in misleading interpretation of the outcomes and wrong prediction of the drug’s safety and efficacy [1]. For this reason, the Institute of Microtechnology (IMT) and the Institute of Pharmaceutical Technology (IPhT) developed a Dynamic Micro Tissue Engineering DPhG Annual Meeting 2015 Conference Book • 137 POSTERS 4.5 Analytics POS.062 Method development for determination of neurosteroids in cerebrospinal fluid as potential biomarkers of postoperative cognitive dysfunction Teubel, J.1; Wüst, B.2; Schipke, C.3; Peters, O.4; Parr, M. K.1 1 2 Institute of Pharmacy, Berlin, Germany, Freie Universität Berlin, Berlin, Germany Agilent Technologies, R&D and Marketing, Santa Clara, California, USA 3 Charité-Universitätsmedizin Berlin, Experimental and Clinical Research Center, Berlin, Germany 4 Charité Berlin, Departement of Psychatry and Psychotherapy, Berlin, Germany Post-operative cognitive dysfunction (POCD) is a condition affecting mainly elderly patients (> 60 yrs) that undergo surgery and anesthesia. It may last for days up to weeks and may even persist over time, leading to lasting health problems. Since the detailed etiology of POCD and other post-interventional cognitive deficits is unknown, the project aims to investigate (neuro-)biological correlates and predictors. One group of potential correlates are neurosteroids, i.e. steroids that are synthesized in the central and peripheral nervous system and appear to have numerous physiological effects on cognition and brain function. A comprehensive method to quantify specific neurosteroids in cerebrospinal fluid (CSF), including pregnenolone sulfate that is emerging as a key substance for cognitive ageing, is developed. As concentrations in CSF are at the ultra-trace level (generally at a pg/mL-range) highly sensitive methods have to be used. Further, high structural similarities of chosen steroids challenges achievement of chromatographic selectivity. Therefore, different chromatographic techniques were evaluated. Detection and separation of 40 selected steroids, including androstanes, corticoids, estranes and pregnanes, was achieved utilizing mass spectrometry following chromatographic separation. POS.063 Determination of phytosterols in herbal medicinal products for the treatment of lower urinary tract symptoms and food products marketed in Europe Müller, C.1; Bracher, F.1 1 Department für Pharmazie – Zentrum für Pharmaforschung, Ludwig-Maximilians-Universität München, Butenandtstraße 5-13, 81377 München, Germany For more than 50 years herbal medicinal products and food products containing phytosterols have been used for treatment of lower urinary tract symptoms such as benign prostatic hyperplasia and overactive bladder in Europe. Mainly products with preparations out of pumpkin seeds, saw palmetto fruit, and nettle root are used, as well as β-sitosterol and other poorly defined phytosterol preparations [1]. The Δ7phytosterols, typical constituents of pumpkin seeds, are assumed to be effective in therapy of lower urinary tract symptoms due to an inhibition of 5α-dihydrotestosterone binding at cellular androgen receptors in the prostate [2]. Urinary obstructive symptoms are improved [2], and a clinical reduction of the International Prostate Symptom Score [3], or at least a better quality of life are achieved [4]. Due to the heterogeneity of the phytosterol preparations on the market an analytical tool is needed for the exact analysis of the sterol compositions as a basis for quality control and estimation of phytoequivalence and comparability of clinical data. We have worked out a convenient extraction and isolation method which, in combination with sophisticated gas chromatography ion trap mass spectrometry, is suitable for analysing phytosterols in complex matrices like unprocessed material (pumpkins seeds, pumpkin seed oil) and phytosterol-containing preparations (hard and soft gelatine capsules, tablets) from various medicinal plants [5]. The phytosterols of each sample were separated after alkaline hydrolysis by liquid/liquid extraction, and analyzed as their corresponding trimethyl silyl ethers. We were able to detect ten different phytosterols, the ubiquitary phytosterol campesterol was detected in 27 samples (n = 38). ΒSitosterol was the quantitatively predominating phytosterol among the non Δ7-phytosterols (n = 5), and was detected in significant amounts in 138 • DPhG Annual Meeting 2015 Conference Book most, but not all preparations for which this sterol or “phytosterol” was declared as a main component. It was detected in 23 samples in a range from 0.0-53.4 mg/dosage. The content of total non Δ7-phytosterols ranged from 0.0-58.4 mg/dosage. As expected, Δ7-phytosterols (n = 5) were only found in pumpkin seed preparations. Surprisingly, a few pumpkin seed preparations contained only non Δ7-phytosterols. Δ7,25Stigmastadienol was the main Δ7-phytosterol in all samples and ranged from 0.0-6.1 mg/dosage, and the content of total Δ7-phytosterols per dosage ranged from 0.0-15.4 mg. References: 1. Bracher, F.: Urologe 1997, A 36: 10-17. 2. Gossell-Williams, M.; Davis, A.; O'Connor, N.: J. Med. Food 2006, 9: 284-286. 3. Vahlensieck, W. et al.: Urol. Int. 2015, 94: 1-10. 4. Shirvan, M. K et al.: JPMA 2014, 64: 683-685. 5. Müller, C.; Bracher, F.: Planta. Med. in press 2015. POS.064 LC-ESI-MS method development and validation for determination of five hypoglycemic agents in mixture using monolithic silica column Asmari, M.1; Alhazmi, H. A.1; Wölker, J.1; El Deeb, S.1 Institute of Medicinal and Pharmaceutical Chemistry, TU Braunschweig, D-38106 Braunschweig, Germany. 1 In this work a LC-ESI-MS method has been developed and validated for the simultaneous determination of metformin, alogliptin, sitagliptin, vildagliptin and linagliptin in mixture. The method is applicable for the quality control of a number of pharmaceutical products since metformin is usually combined in one dosage form with either of the above dipeptidyl peptidase-4 inhibitors for the treatment of type II diabetes mellitus. The LC analyses were performed using an Agilent 1100 LC system with second-generation monolithic silica column Chromolith® HighResolution RP-18e column (100×4.6 mm, Merck). A mobile phase consisting of acetonitrile/ ammonium formate buffer (20:80, v/v) at a pH 3 was used at a flow rate of 0.4 mL/min. A single-quadrupole mass spectrometer (Agilent 1620) equipped with an electrospray ionization source was used. After optimization, the best conditions were set with a nebulizer pressure of 30 psi, drying gas temperature of 250 °C, capillary voltage of 3 kv and a fragmentor voltage of 70 v under the positive ion mode. After identification of the individual molecular ions using the scan mode, which gave excellent mass accuracy, the MS was set on selected-ion monitoring (SIM) mode using target ions at m/z [M+H+] 130.1 for metformin, m/z [M+H+] 304.2 for vildagliptin, m/z [M+H+] 340.2 for alogliptin, m/z [M+H+] 408.1 for sitagliptin and m/z [M+H+] 473.2 for linagliptin for quantitation. The method was linear over the concentration range of 0.039-20 µg/ml for metformin and 0.19-100 µg/ml for the others. The correlation square of the linear regressions were better than 0.99 for the five quantitated compounds. RSDs% of peak areas and retention times were less than 0.027% and 0.00079%, respectively. This precise method would be further tested for stability and bioanalysis investigations. POS.065 Child-appropriate high throughput HPLC-MS/MS for enalapril and enalaprilat in small sample volumes of serum and urine within a GCLP-compliant environment Schaefer, J.1; Burckhardt, B. B.1; Tins, J.1; Läer, S.1 Institute of Clinical Pharmacy and Pharmacotherapy, Heinrich-Heine-University Duesseldorf, Universitaetsstr. 1, 40225 Duesseldorf, Germany 1 Background: Heart failure is a life-threatening disease in neonates up to adolescents. The angiotensin-converting enzyme inhibitor enalapril is a recommended therapy in paediatric heart failure, although it is not labelled for patients <20 kg in European countries. The LENA (Labeling of Enalapril from Neonates up to Adolescents) project aims to generate data for devising a paediatric-use marketing authorization for an ageappropriate enalapril formulation. For enalapril, pharmacokinetic data drug assays for serum and urine specifically tailored to paediatric ANALYTICS populations were developed and used in the GCLP-compliant setting of the LENA phase I study. Objective: Proof of concept of the paediatric tailored assays by verification of the reliable conduct of sample analysis according to international bioanalytical guidelines and determination of enalapril and its active metabolite enalaprilat in the LENA phase I study. Methods: All LENA phase I samples were analysed applying the developed high throughput analysis for enalapril and enalaprilat via HPLC-MS/MS. According to the established LENA GCLP-compliant quality system, the evaluation of all study samples was conducted by using freshly prepared quality standards to obtain calibration curves of enalapril and enalaprilat in serum and urine. Results: In total, 22 calibration curves in serum and 7 in urine were required to investigate all samples of the phase I study of LENA. All 29 calibration curves complied with the limits of FDA and EMA bioanalytical guidelines [1,2] and the applicability of the established high throughput method in the GCLP environment was proven. About 2100 study samples were successfully determined within 26 days. Conclusion: The developed paediatric tailored high throughput HPLCMS/MS analysis proved its applicability in a GCLP-compliant environment and is suitable for the upcoming phase II and phase III studies of the LENA project focussing on paediatric patients. Perspectives: Method development is still ongoing. The LC-separation achieved for ten substances will be further improved. Parameter settings for the simultaneous quantification of the drugs from study samples have to be optimised. The final method will be validated according to the particular EMA Guideline [1] and may help to improve the medication safety of the neonates. Acknowledgments: The research leading to these results has received funding from the European Union Seventh Framework Programme (FP7/2007-2013) under grant agreement n°602295 (LENA). 1 Institute References: 1. EMA. Guideline on Bioanalytical Method Validation. EMA, Committee for Medicinal Products for Human Use, London, UK 2011. 2. US FDA. Guidance for Industry: Bioanalytical Method Validation. US Department of Health and Human Services, US FDA, Center for Drug Evaluation and Research, Rockville, MD, USA 2001. POS.066 Development of an LC-MS/MS method to investigate the pharmacokinetics of antibiotics and concomitant drugs given to neonates suffering from sepsis Fürtig, M.-A.1.; Burau, D.1; Kloft, C.1 1 Dept. of Clinical Pharmacy and Biochemistry, Institute of Pharmacy, Freie Universitaet Berlin, Kelchstr. 31, 12169 Berlin, Germany Background: Safety of drug therapies is usually proven by clinical trials before administration to patients in clinical routine, but for some special patient populations e.g. neonates, no clinical data is normally available. If severe diseases occur in these patients, often the high risk of administering non-approved drugs needs to be accepted. In a recent clinical trial, drug concentrations in newborns (n=8) suffering from sepsis are monitored using microdialysis as sampling technique in the interstitial fluid of subcutaneous tissue. Microdialysis enables investigations to unbound drug concentrations in a particular tissue without removal of blood or any other body fluid which is the main limitation of plasma sampling in neonates. In addition to vancomycine (VAN), which was administered as initial antibiotic treatment, 17 concomitant drugs were given to the patients covering mainly antibiotics, antifungals, diuretics and sedatives. This trial will give the opportunity to gain an insight into the pharmacokinetics of several drugs given to neonates. To gain as much information from the patient samples as possible, we are developing an LC-MS/MS method for simultaneous determination of VAN and the concomitant drugs. Methods: An Agilent 1290 HPLC system coupled with an Agilent 6490 triple quadrupole LC-MS/MS detector with electron spray ionisation (ESI) and iFunnel technology, which combines the Agilent Jetstream technology, Hexabore sampling capillary and a dual-stage ion funnel, is used for method development. VAN and the concomitant drugs are both, separately and simultaneously injected to find optimal setting for the simultaneous quantification of all drugs. Different gradients of Milli-Q water containing formic acid (FA) and acetonitrile with FA are tested to improve drug separation prior to detection. Ion source parameters, collision energy, etc. will be optimised to achieve high accuracy and precision for the substances on the one hand and a very low quantification limit on the other hand. Results: Current experiments showed good separation of VAN and nine concomitant drugs. To further improve the method performance concerning accuracy, precision, selectivity and sensitivity; detection needs to be optimised. References: 1. European Medicines Agency (EMA): Guideline on bioanalytical method validation 2012 (http://www.ema.europa.eu/docs/en_GB/document_library/Scientific_guideline/2011/08/WC500 109686.pdf; 29.06.15) POS.067 Affinity capillary electrophoresis as an appropriate technique to investigate the interactions of Pentosan Polysulfate sodium and related substances with various proteins Mozafari, M.1; El Deeb, S.1; Wätzig, H.1 of Medicinal and Pharmaceutical Chemistry, TU Braunschweig, Beethovenstrasse 55, 38106 Brunswick, Germany Affinity capillary electrophoresis has become a powerful method for separation of peptides and proteins. Moreover it is a qualified technique for analysing pharmaceuticals and biopharmaceuticals [1,2,3]. The affinity capillary electrophoresis methods are based on identification of changes in the electrophoretic mobility of analytes due to changes in charge through interacting with ligands [1,2,3]. Pentosan polysulfate (PPS) is highly sulfated polysaccharide derived from beechwood hemicelluloses by sulfate esterification. Beside the well known antithrombotic activity, other properties such as anti-inflammatory, anti-angiogenesis and inhibition of cell adhesion are still under investigation for this substance as well as for heparins [4,5]. Accordingly, it is important to know how strong PPS interacts with some important proteins compared to heparin and other heparinoids. For this purpose affinity capillary electrophoresis methods have been developed due to its numerous advantages such as short analysis duration and minute consumption of samples. In this study we used some model proteins namely HSA, BSA, myoglobin, ovalbumin, beta-lactoglobulin and two other important proteins namely vitronectin and P-selectin, which are involved in the coagulation cascade. The interactions were calculated using mobility ratios (R) of the EOFmarker, acetanilide and the proteins to avoid effects from the migration time shifts, which are not related to interactions [1,2,3]. Among the investigated proteins myoglobin, ovalbumin and beta lactoglobulin, showed no significant interactions with the heparinoids as expected. HSA and BSA showed stronger interactions with PPS than heparin and other heparinoids. In case of HSA, the shape of the peak obtained by electrophoresis was changed and splitted into two peaks after interaction with PPS. The higher the concentration of PPS, the more distinct this effect was. P-selectin showed very strong interaction with the capillary wall so that the use of coated capillaries and a special rinsing procedure were necessary. As well, p-selectin showed strong interactions with PPS and heparinoids, which was probably dependant on the presence of Ca2+ ions. With appropriate ACE methods we could demonstrate that heparinoids exhibit strong affinity to some of these important proteins. Acknowledgments: bene pharmaChem for financial support of this project, Polymicro for the donation of capillary material References: 1. Redweik, S. et al.: Electrophoresis 2012, 33: 3316-3322. 2. Alhazmi, H. A. et al.: J. Pharm. Biomed. Analysis 2015, 107: 311-317. 3. El Deeb, S. et al.: Electrophoresis 2015, 35: 170-189 4. Abdel-Haq, H. et al.: J. Chrom. Analysis 2012, 1257: 125-130 5. Degenhardt, M. et al.: Arch. Pharm. Pharm. Med. Chem. 2001, 334: 27-29. DPhG Annual Meeting 2015 Conference Book • 139 POSTERS POS.068 A novel fluorogenic probe for the investigation of free thiols: Application to kinetic measurements of acetylcholinesterase activity Mertens, M. D.1; Bierwisch, A.2; Li, T.1; Gütschow, M.1; Wille, T.2; Elsinghorst, P. W.1,3 1 Pharmaceutical Institute, Pharmaceutical Chemistry I, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany 2 Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstraße 11, 80937 München, Germany 3 Supervisory Agency for Public Law Tasks of the Bundeswehr Medical Service South, Dachauer Straße 128, 80637 München, Germany In vitro measurements of acetylcholinesterase (AChE) activity are usually carried out following the method of Ellman [1], where a chromogenic reagent (DTNB) is applied to detect the thiocholine liberated from acetylthiocholine by action of AChE. However, spectrophotometric detection of the reaction product can be impaired in highly coloured solutions or in samples of low AChE activity. These drawbacks can to a reasonable extent be overcome by use of a fluorogenic substrate providing improved signal selectivity and sensitivity. Several fluorescent probes for in vitro and in vivo detection of free thiols have been developed and in rare cases have also been applied to detect AChE activity [2]. However, when used for determination of AChE activity only end-point measurements were carried out without further kinetic analysis [3,4]. variety of N-hydroxylated compounds including amidines, guanidines, amidinohydrazones, aromatic amines as well as N-hydroxylated nucleosides and nucleobases. In addition, the three component Nreductive enzyme system is able to reduce N-oxides [2-4]. According to these findings, TMAO may also serve as a substrate for mARC. So far, the physiological role of mARC is unknown though it contributes both to the activation of prodrugs and to the metabolism of xenobiotics [5]. The mARC mediated reduction of TMAO would be a hint to a participation of the molybdo enzyme in prevention of CVD. To investigate whether TMAO is reduced through mARC it is necessary to develop a specific assay and analytical method to quantify the metabolite TMA whereupon the volatile character as well as the lack of a chromophore of TMA poses particular challenge for this task. The derivatization of TMA to a non-volatile compound displays a promising opportunity to quantify the metabolite via LC/MS. References: 1. Wang, Z. et al.: Nature 2011, 472 (7341): 57-63. 2. Havemeyer, A. et al.: J. Biol. Chem. 2006, 281(46): 34796-34802. 3. Gruenewald, S. et al.: J. Med. Chem. 2008, 51(24): 8173-8177. 4. Jakobs, H. et al.: ChemMedChem 2014, 9: 2381-2387 POS.070 The GAPAGPLIVPY - Ca2+-Complex – An example for comparing differences in observations of different techniques used for peptide-metal ion binding studies Nachbar, M.1; Mozafari, M. 1; Alhazmi, H. A. 1; Preu, L.1; Redweik, S.1; El Deeb, S.1; Lehmann, W. D.2; Wätzig, H.1 1 Institute Figure: A nosyl-protected coumarin (top) was developed to allow for fluorescence detection of free thiols, e.g., in kinetic measurements of acetylcholinesterase activity using acetylthiocholine (right). Here, we describe the preparation of a nosyl-protected/quenched coumarin (Figure, left) designed as a probe for free thiols based on the well-known susceptibility of the nosyl-group towards thiol-mediated cleavage. We subsequently used this probe to develop a fluorimetric assay of AChE activity that allows for kinetic measurements rather than end-point observations (Figure, right). Details of this assay and associated kinetic analyses are presented. Acknowledgments:This study was funded in part by the German Ministry of Defence. References: 1. Ellman, G. L. et al.: Biochem. Pharmacol. 1961, 7: 88–95. 2. Peng, H. et al.: Sensors 2012, 12(11): 15907–15946. 3. Maeda, H. et al.: Angew. Chem. Int. Ed. 2005, 44(19): 2922–2925. 4. Gainullina, E. T. et al.: Bull. Exp. Biol. Med. 2006, 142(6): 751–752. POS.069 mARC and its potential role in prevention of cardiovascular disease Schneider, J.1; Girreser, U.1; Clement, B.1 Department of Pharmaceutical Chemistry, Christian-Albrechts-University Kiel, Gutenbergstraße 76, 24118 Kiel, Germany 1 Cardiovascular disease (CVD) is the leading cause of morbidity worldwide. It is therefore of great interest to reveal pathogenic as well as diagnostic factors to establish valid diagnostic tests and therapeutic approaches [1]. It was discovered that the metabolic profile of trimethylamine N-oxide (TMAO) in plasma predicts the risk for CVD, potentially through TMAOrelated promotion of atherosclerosis. TMAO is a physiological metabolite that derives from dietary choline. It is well accepted that gut microbiota converts dietary choline and its precursors to trimethylamine (TMA). Hepatic flavin monooxygenase metabolizes the TMA to TMAO which is subject to renal excretion [1]. The recently in our lab discovered mitochondrial amidoxime reducing component mARC forms along with the heme-containing cytochrome b5 and its flavin-containing cytochrome b5 reductase an N-reductive enzyme system which is able to reduce a 140 • DPhG Annual Meeting 2015 Conference Book of Medicinal and Pharmaceutical Chemistry, TU Braunschweig, Beethovenstrasse 55, 38106 Brunswick, Germany 2 Core Facility Molecular Structure Analysis, German Cancer Research Center (DKFZ), Heidelberg, Germany Quantitative structure-activity relationships (QSARs) are an important tool in generating new bioactive compounds. However, the quality of these models is strongly depending on the used empirical data [1]. The choice of the used technique and method has a big impact on the results of binding studies. Direct electrospray ionization mass spectrometry (ESI-MS) as technique for binding essays, for example, has some pitfalls described in literature e.g. in-source dissociation, ESI-induced changes in solution pH and temperature, non-uniform response factors and nonspecific binding [2]. However, there are also other obstacles related to this technique. Lehmann et al. reported that the peptide GAPAGPLIVPY, derived from galectin-3 by a combined chymotrypsin/AspN digest, showed a strong, sequence specific affinity towards alkaline earth metal ions in nanoESI-MS experiments [3]. The relevance of these GAPAGPLIVPY-complexes for aqueous solutions was not quite certain, since Dunbar et al. described differences in the binding mode between the gas and aqueous phase [4]. For this purpose, the binding behaviour of the peptide GAPAGPLIVPY towards various metal ions was also investigated in aqueous phase using mobility-shift affinity capillary electrophoresis (ACE) and compared to the reported results. The outcome of the ACE experiments showed no significant interactions in aqueous phase, except for Cu2+- and Mn2+-ions. To explain these contradictions, computational methods were used to make a GAPAGPLIVPY-Ca2+-complex for the gas phase. Afterwards, its behaviour in vacuum phase and aqueous solution was investigated by molecular dynamics simulations. These investigations showed that the complex was only stable in the gas phase due to differences in the gas phase binding mode compared to the aqueous solution. In vacuum, the complex is stabilized by the Ca2+-ion, since the complex then exhibits a hydrophobic surface and solvates highly charged species like ions inside. Typical interactions of polar groups like hydrogen bonds, dipole-dipole interactions and ligand exchanges are more favourable in aqueous solution and therefore destabilize the GAPAGPLIVPY-Ca2+-complex, which obviously results in a fast disassembly of the complex. Acknowledgments: The authors thank Polymicro Technologies for providing the capillaries used in this work. References: 1. Wätzig, H. et al.: J. Comput. Aided Mol. Des. 2015, DOI: 10.1007/s10822-015-9851-6 2. Kitova, E. N. et al.: J. Am. Soc. Mass Spectr. 2012, 23(3): 431–441. 3. Lehmann, W.D.; et al.: Rapid Commun Mass Sp. 2006, 20(16): 2404–2410. 4. Dunbar, R.C.; et al.: Int. J. Mass Spectrom. 2013, 354-355: 356-364. ANALYTICS POS.071 Identification of photodegradation products of ketoprofen utilizing online photoreactor-HPLC, GC/MS and isotopic labeling Assaf, J.1; Zulkiewicz Gomes, D.1,2; Schulze, T.1; Wuest, B.3; Parr, M. K.1 1 Freie Universität, Institute of Pharmacy, Königin-Luise-Straße 2+4,14195 Berlin. IPT – Institute for Technological Research LAQ - Chemical analysis Laboratory, Av. Prof. Almeida Prado, 532 Cidade Universitária, 05508-901 São Paulo, SP, Brazil. 3 Agilent Technologies, 5301 Stevens Creek Blvd, Santa Clara, CA 95051, United States. 2 Photochemical properties of drug substances and their associated phototoxic properties are becoming more important in the drug characterisation as well in drug developments.Therefore pharmaceutical industries are also requested by the regulation agencies to provide data for new or renewed licenses of medicines. The aim of this work is to study the photostability of Ketoprofen by using a modified HPLC system, which consists of a tailored online photoreactor with back-flush and two column system. Identification was performed using a mass selective detector and confirmed by isotopically labelled (deuterated) ketoprofen. Irradiation of the nonsteriodal anti-inflamatory agent ketoprofen with UVA resulted in decreasing amounts of the target compound. First order kinetics was found as best model. Several photoproducts were detected after irradiation in aqueous solution (pH 7.0), when maintaining the temperature in the reactor at 25-30 °C. Mass spectrometric characterization supported the structures reported from literature. Ketoprofen and one of its proposed photoproducts are shown in Figure. These data showed the potential for the use of the new device for fast and easy photostability studies that may help to reduce time consuming in vitro experiments and animal trials. Targeted MS/MS-based methods may be generated using the results obtained by these online-irradiations for use in vitro and in vivo. Scale-up may also be realized for the generation of reference material for quantification as well as for toxicity testing. Figure: Ketoprofen (left) and one of its proposed photoproducts 2-ethylbenzophenon (right). Acknowledgments: Katholischer Akademischer Ausländer-Dienst (KAAD) is thankfully acknowledged for financial support. References: Methods: The investigations were performed consecutively with CMA71 probes (100 kDa cut-off) and a mixture of Ringer`s solution (RS) and human serum albumin solution (HSA) (0.5%, v/v) as perfusate and probe-surrounding medium. The AFG concentration in the probesurrounding medium was 1 µg/mL for (i) uncoated probes and (ii) CFG (50 µg/mL in perfusate) coated probes. Microdialysate (µDialysate) was collected (nprobes=3) in 40 min intervals over 8 h (flow rate of 1 µL/min). Probes in (ii) were coated 16 h prior to and during the actual recovery investigation with the CFG containing perfusate. Quantification of AFG was performed with a previously developed and validated HPLC assay (LiChrospher 100 RP-18 column (125 x 4 mm, 5 µm), isocratic method with eluent: methanol and ammonium dihydrogen phosphate 85:15 (v/v), concentration range: 0.1–20 µg/mL, accuracy and precision of the LLOQ (0.1 µg/mL) were RE ≤ ±19.6% and CV ≤ 10.6%, respectively). The influence of coating the probe with CFG containing perfusate was investigated with regard to the relative recovery (RR) values of AFG. Recovery investigation with (i) was compared with (ii) over time. All results of RR were volume-corrected by their volume of µDialysate. RR, %= cdialysate *100 cmedium Results: One µDialysate sample after the first sampling interval of (i) and another of (ii) had concentrations of AFG < LLOQ and were not included in the calculations. All µDialysate and medium sample concentrations were quantified by the HPLC assay and afterwards RR was calculated from these concentrations. RR showed a steep increase in the first 3 h for both approaches. Afterwards RR was stable for the following 5 h with max. RR ranging from 23.2%-26.9% and 27.0%-30.1% for (i) and (ii), respectively. The comparison of approach (i) and (ii) led to very similar results with regard to the time necessary to reach stable RR values. Conclusions: AFG displayed substantial adsorption to µD probe material for both approaches. CFG was considered to block the AFG binding sites on the probe material to prevent AFG from adsorption. It was however observed that AFG was still adsorbed on the probe material which was displayed by the increase of RR within the first 3 h of investigation. Further research is required to reduce the initial increase in RR due to adsorption and obtain constant recovery of AFG in µD as this is the prerequisite for reproducible and reliable in vivo µD to gain profound knowledge of target-site pharmacokinetics and ultimately evaluate dosing regimens. References: 1. Weiser et al.: 25th ECCMID, Copenhagen, Denmark. 2015, P-0222. 2. Traunmüller et al.: J. Chromatogr. 2006, B 843: 142-146. 1. Salgadoa, R. et al.: J.Hazard. Mater. 2013, 244-245: 516– 527. 2. Schulze, T.: Entwicklung einer In-Vitro-Methoden zur Beurteilung des photosensibilisierenden Potentials von Arzneistoffen, Freie Universität Berlin, Universitätsbibliothek 2013. POS.073 POS.072 A new approach for in vitro microdialysis of anidulafungin Weiser, C.1; Zeitlinger, M.2; Kloft, C.1 1 Dept. of Clinical Pharmacy and Biochemistry, Institute of Pharmacy, Freie Universitaet Berlin, Kelchstr.31, 12169 Berlin, Germany 2 Dept. of Clinical Pharmacology, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria Objectives: Anidulafungin (AFG), approved in Germany in 2007, is a lipophilic echinocandin antifungal drug and a treatment option for invasive candidiasis in non-neutropenic adult patients. Microdialysis (µD), a minimally invasive technique, can serve in clinical settings to determine the drug concentrations directly at the target-site e.g. in the interstitial fluid of organ and muscle tissue. Since only unbound drug molecules are able to pass the semipermeable membrane of the µD catheter solely the pharmacologically active fraction of the drug is collected and subsequently quantified. Because AFG shows high adsorption on probe material (membrane, tubing) [1] caspofungin (CFG), also known to be highly adsorbing on µD probes, was chosen to overcome AFG adsorption by coating the probes with CFG prior to the actual investigation [2]. Therefore CFG was selected to adsorb on and thus block the potential AFG binding sites on the probe material, so that AFG would be prevented from adsorption. Handling small sample volume for capillary electrophoresis investigations Mozafari, M.1; Nachbar, M.1; El Deeb, S.1 1 Institute of Medicinal and Pharmaceutical Chemistry, TU Braunschweig, Beethovenstrasse 55, 38106 Brunswick, Germany Capillary electrophoresis is an appropriate technique for analysing pharmaceuticals and biopharmaceuticals [1,2] for which using small volume samples is advantageous. There are numbers of benefits involved in handling small sample volume in capillary electrophoresis, among which using less material, generating less waste, and applying high concentrations of valuable samples which are all very important from an economic perspective stand out [3,4]. Most of the conventional CE instruments need approximately 50 µL of the sample in the injection vial to perform the analysis, even though the injected sample volume in capillary electrophoresis is only in the nanoliter range. Hence, in order to fully profit from the low injection volumes, smaller vial volumes are required. Furthermore the immersion depth of the capillary of most CE instruments is not properly adjustable. Even if it is, one cannot ensure that the capillary does not break at the end or at the detection window if it comes against the bottom of the vial in lower settings. Therefore it is important to fill the dead volume of the sample vials, which is inaccessible for the capillary with a chemically inert liquid. DPhG Annual Meeting 2015 Conference Book • 141 POSTERS Thus experiments were performed using silicone oil as a filler of the vial dead volume and the results were compared to those performed without this filling. As study example five standard proteins namely beta-lactoglobulin, BSA, HSA, Myoglobin and Ovalbumin, and one of the coagulation cascade involved proteins called vitonectin were investigated using capillary electrophoresis. The experiments were performed once without the addition of silicone oil in the sample vial and once in the presence of silicone oil in the sample vial. The equation of migration times of EOFmarker and proteins (mobility ratios) as well as peak areas were compared. However no significant changes were observed (RSDs% for mobility ratios and peak areas were better than 0.9% and 5.8% respectively). Afterwards an affinity capillary electrophoresis method was used to investigate the interactions of two proteins, namely HSA and vitronectin, with three ligands namely enoxaparin sodium, unfractionated heparin and pentosan polysulfate sodium (PPS). Mobility shift precision results over 12 hours analysis showed that the employment of the filling has no noticeable effect on any of the protein-ligand interactions. Accordingly, the employed silicone oil is suitable as a water immiscible and inert liquid for filling the dead volume of sample vials. Using a commercial instrument and an autosampler the required sample volume is reduced down to 10 µL, and almost this complete volume can be subsequently injected during repeated experiments. Acknowledgments: bene pharmaChem for financial support of this project, Polymicro for the donation of capillary material References: 1. Redweik, S. et al.: Electrophoresis 2012, 33: 3316-3322. 2. Alhazmi, H.A. et al.: J. Pharm. Biomed. Analysis 2015, 107: 311-317. 3. Walter, R. et al.: Anal. Chem. 2002, 74: 3575-3578. 4. Beutner, A. et al.: Microchim Acta 2015, 182: 351-359. POS.074 Towards selective CK2α and CK2α’ inhibitors: A novel screening assay by Autodisplay of heterotetrameric human protein kinase CK2 evaluation of the relative inhibition of a set of known ATP-competitive CK2 inhibitors. Additionally the system was used to screen a small library of CK2 inhibitors with a indeno[1,2-b]indole scaffold [3] in order to identify a selective inhibitor of CK2α or CK2α’. Our results demonstrate that this assay allows the discovery of CK2 inhibitors with a distinct effect on the two different catalytically active CK2 isoforms, thus enabling the identification of inhibitors with a selective affinity towards either CK2α or CK2α’. References: 1. Cozza, G.; Bortolato, A.; Moro, S.: Med. Res. Rev. 2010, 30: 419-462. 2. Gratz, A. et al.: Microb. Cell Fact. 2015, 14: 74. 3. Gozzi, G. J. et al.: J. Med. Chem. 2015, 58: 265-277. POS.075 Site specific labelling of human protein kinase CK2 for drug discovery applications Nienberg, C.1; Becher, K.2; Mootz, H. D.2; Jose, J.1 Institut für Pharmazeutische und Medizinische Chemie, PharmaCampus, Westfälische Wilhelms-Universität Münster, Corrensstraße 48, D-48149 Münster, Germany. 2 Institut für Biochemie, Westfälische Wilhelms-Universität Münster, Wilhelm-Klemm-Straße 2, D-48149 Münster, Germany. 1 Human CK2 is a heterotetrameric constitutively active serine / threonine protein kinase, phosphorylating and regulating a variety of cellular processes. In tumor and rapidly proliferating cells the activity of CK2 is increased. Downregulating the activity of the protein kinase in the affected tumor cells initiates apoptosis [1]. Thus, CK2 represents a promising target in current cancer research. The kinase is composed of two catalytic CK2α subunits and two regulatory CK2β subunits. Most protein-protein interaction (PPI) studies or screening assays are based on fluorescence detection and require the labelling of the target enzyme by a fluorophore. The catalytic subunit CK2α loses activity after labelling by commercial applications. Furthermore, the labelling ratio of the protein sample differs and is not exactly reproducible. Bollacke, A.1; Le Borgne, M.2; Jose, J.1 Institut für Pharmazeutische und Medizinische Chemie, PharmaCampus, Westfälische Wilhelms-Universität Münster, Corrensstraße 48, 48149 Münster, Germany. 2 Université de Lyon, Université Lyon 1, Faculté der Pharmacie-ISPB, EA 4446 Biomolécules Cancer et Chimiorésistances, SFR Santé Lyon-Est CNRS UMS3453-INSERM US7, Lyon Cedex 8, 69373, France. 1 Novel inhibitors that target the human protein kinase CK2 are of great interest in the development of new therapeutics for the treatment of cancer. The human protein kinase CK2 commonly occurs as a heterotetrameric enzyme, the so-called CK2 holoenzyme, and is composed of two catalytically active α- and/or α’-subunits attached to a dimer of non-catalytically active β-subunits [1]. We recently reported the successful co-expression of CK2 subunits α and β on the cell surface of Escherichia coli via Autodisplay and demonstrated the interaction of the surface presented CK2 subunits, as well as the possible application for the purpose of inhibitor screening by a CE-based assay [2]. The solution for this problem was an incorporation of an unnatural amino acid into the CK2α subunit followed by a Strain-Promoted Alkyne-Azide Cycloaddition (SPAAC) [2]. Therefore, a suitable position in the sequence of CK2α was selected and mutated to the amber nonsense DNA codon, TAG. By suppression of the mutation with an amber suppressor tRNA, the unnatural amino acid para-acidophenylalanine (pAzF) could be incorporated at this position [3]. Performing the SPAAC click reaction by the use of dibenzylcyclooctyne-fluor 545 (DBCO 545) led to a specifically labelled CK2α and CK2 holoenzyme. This specific kind of labelling does not impair the phosphorylation activity of the CK2α subunit alone nor the holoenzyme, which was evaluated by capillary electrophoresis. The innovatively labelled kinase in combination with the Autodisplay technology could be a significant advancement for inhibitor screening assays by flow cytometry and for CK2α/CK2β interaction studies [4]. References: 1. Wang, G. et al.: Mol. Cell. Biochem. 2005, 274(1-2): 77-84. 2. Mbua, N. E. et al.: ChemBioChem. 2011, 12(12): 1912-21. 3. Chin, J. W. et al.: J. Am. Chem. Soc. 2002, 124(31): 9026-7. 4. Jose, J.; Meyer, T. F.: Microbiol. Mol. Biol. Rev. 2007, 71(4): 600-19. POS.076 We herein describe the successful Autodisplay of a paralogous isoform of the catalytically active CK2 subunit, the CK2α’-subunit. Analysis of the kinase activities at different NaCl concentrations for the individually expressed α’-subunit and for the α’-subunit and the non-catalytically active β-subunit co-expressed, confirms the interaction of the subunits on the cell surface. We further illustrate the potential application of the two surface presented CK2 holoenzymes as a novel inhibition assay by the 142 • DPhG Annual Meeting 2015 Conference Book Dried Blood Spot Analysis for Therapeutic Drug Monitoring of Antipsychotics Weber, J.1; Hempel, G.1 1 Department of Pharmaceutical and Medical Chemistry – Clinical Pharmacy, University of Münster, Corrensstrasse 48, 48149 Münster, Germany Background: Dried blood spot (DBS) analysis is an innovative sample technique to collect and analyse very small volumes of whole blood. The blood obtained from the finger pulp is spotted on a filter paper. After ANALYTICS drying, the analyte can be extracted and analysed. The main benefit of DBS analysis is the possibility of long storage, simple transfer and noninvasive blood sampling [1]. Increasingly sensitive analytics enable quantitative measurements of very small volumes of blood and thereby facilitate DBS analysis for Therapeutic Drug Monitoring (TDM) [2]. Various antipsychotics and antidepressants show a correlation between drug effect and blood concentration, such as risperidone, paliperidone, mirtazapine and citalopram [3]. These substances are commonly prescribed in nursing homes where physicians are mostly absent. Collecting DBS by nurses or even by the patient himself at any time offers an excellent possibility to perform TDM. Thus, the aim is to develop a DBS method for several antipsychotics and antidepressants for TDM. Methods: Blank whole blood was spiked with the analytes risperidone, paliperidone, mirtazapine and citalopram. By using a heparinized capillary a defined volume of spiked blood was spotted on treated and non-treated DBS cards (FTATM DMPK-C Cards and Whatman 903 paper, GE Healthcare, Freiburg, Germany). After a minimum drying period of two hours the spots were cut out and transferred into an Eppendorf cap. The internal standard (IS) clozapine was added and the analytes were extracted by several organic solvents using an ultrasonic bath. After drying under nitrogen gas the filter paper were removed. The samples were subsequently purified by liquid-liquid extraction (LLE) in order to prevent matrix effects and to achieve a high selectivity. After evaporating and dissolving with methanol the extracted samples were analysed on a Shimadzu LCMS-2020 system (Shimadzu, Duisburg, Germany). Chromatographic separation was achieved using a Kinetex C18 reversed phase column (3 x 100 mm, particle size 2.6 µm, Phenomenex, Torrance, CA, USA) and a gradient elution with a flow rate of 0.5 ml/min. Results: The best selectivity especially with regard to matrix effects was achieved by non-treated Whatman 903 paper cards. Extracting the analytes from the DBS in an ultrasonic bath, the best results were achieved by using methanol. In order to improve the purity level of the samples a LLE is well suited because the hydrophilic substances were separated from the analytes. The best recovery for all four analytes was achieved by using hexane/butyl acetate and a sodium hydroxide solution as liquid phases. The calibration curve showed an excellent fit over a concentration range of 2.5-300 µg/l for risperidone and paliperidone and a concentration range of 10 – 300 µg/l for mirtazapine and citalopram. Conclusion: This investigation shows a successful method to combine DBS analysis with LLE for preventing matrix effects and to ensure selectivity. It sets the basis to develop a quantifying DBS method for risperidone, paliperidone, mirtazapine and citalopram which can be used for TDM. Method: For separation, Chromabond® C18 50 mg solid phases (Machery-Nagel, Germany) and a SPE 12G glass column processor (J.T. Baker®, Avantor Performance Materials, USA) were used. Conditioning of the columns was done with 300 µL methanol, 750 µL phosphate buffer pH 7.4 and 100 µL of a mixture of blank plasma and 5% glucose solution. 50 µL of spiked plasma were mixed with 50 µL internal standard and 100 µL 5% glucose solution and applied to the solid phase. To remove plasma components and liposomes, 100 µL of a mixture of blank plasma and 5% glucose solution, 500 µL phosphate buffer pH 7.4 and 100 µL demineralized water was used for rinsing. An additional washing step with 200 µL 80% methanol was implemented in order to purify the eluate. Afterwards, the columns were dried under vacuum for 5 minutes and 400 µL elution solvent was applied. All samples were evaporated under nitrogen stream and dissolved in mobile phase for HPLC analysis. Samples were analysed using a Shimadzu Prominence HLPC System (Shimadzu, Germany) with a RF-20Axs fluorescence detector (excitation: 495 nm, emission: 580 nm) and a Kinetex® 2.6 µm C18 100 Å, 100 x 3 mm (Phenomenex, USA) LC column for chromatographic separation. Results: Three different attempts to improve elution were carried out and the best result of each attempt was implemented in the next one. Elution with additional 2% formic acid compared to pure methanol increased peak area of DAUN by 54% (±10%) and DAUNol by 24% (±9%). In comparison to Chromabond® C18 50 mg, Bond Elut C18 50 mg and Bond Elut Plexa 30 mg solid phases increased peak area of DAUN by 105% (±3%) and 121% (±1%) and DAUNol by 98% (±3%) and 115% (±2%), respectively, and also lowered the variance between different samples. Separating the elution solvent into two portions of 200 µL showed an increase of DAUN and DAUNol peak areas by 3% (±1%) each. Conclusion: Best results for improving the amount of free DAUN and DAUNol being eluted were achieved with Bond Elut Plexa 30 mg solid phases and 400 µL methanol containing 2% formic acid. Double elution did not show any difference, but was more time-consuming and thus was not implemented in the advanced method. References: 1. Bellot, R.; Pouna, P.; Robert, J.: J. Chromatogr. B 2001, 757(2): 257-67. 2. Griese, N. et al.: J. Chromatogr. A 2002, 979(1-2): 379-88. References: 1. Wagner, M. et al.: Mass Spectrom. Rev. 2014, doi: 10.1002/mas.21441 2. Edelbroek, P.M.; van der Heijden, J.; Stolk L.M.: Ther Drug Monit. 2009, 31: 327–336 3. Hiemke, C. et al.: Pharmacopsychiatry 2011, 44: 195–235 POS.077 Improved separation of free daunorubicin and its metabolite daunorubicinol from liposomal daunorubicin in plasma using solid phase extraction Liebich, M.1; Hempel, G.1 1 Department of Pharmaceutical and Medical Chemistry - Clinical Pharmacy -, University of Münster, Corrensstraße 48, 48149 Münster, Germany Background: Liposomal daunorubicin (DaunoXome®) is used in induction therapy for the treatment of acute myeloid leukemia in pediatric patients. The liposomal form is designed to reduce side effects, especially cardiotoxicity, and to increase therapeutic response. After administration, daunorubicin (DAUN) is slowly released from the liposomes and free DAUN is metabolized to daunorubicinol (DAUNol). Separating the liposomal encapsulated from the free drug in plasma is challenging. The method should avoid high mechanical stress, organic solvents, strong acids or bases to prevent destruction of the liposomes. Solid phase extraction is a method to separate the liposomal and the free drug, because the liposomes show no adsorption to the solid phase and are eluted by buffer solutions while free DAUN and DAUNol adsorb to the solid surface and can be eluted by an organic solvent. The developed method is based on two publications by Bellot et al. [1] and Griese et al. [2], however, aiming at increasing the amount of free DAUN and DAUNol being eluted from the solid phase and reducing the required sample volume. DPhG Annual Meeting 2015 Conference Book • 143 POSTERS 4.6 GPCR/Ion channels POS.078 GluN2B selective NMDA receptor antagonists with 3benzazepine scaffold: The deconstruction reconstruction approach Dey, S. 1,2; Wünsch, B.1 1 Institut für Pharmazeutische und Medizinische Chemie, Corrensstraße-48, D-48149, Münster, Germany. Graduate School of Chemistry, University of Münster, Wilhelm-klemm-str., D-48149, Münster, Germany 2 A functional NMDA receptor is built from at least one GluN1 and one GluN2 subunit. The simultaneous binding of two agonists, (S)-glutamate at the GluN2 subunit and glycine at GluN1 or GluN3 subunit activates the NMDA receptor. The NMDA receptor has received significant attention due to its unique properties and therapeutic potential. It is considered as an important therapeutic target for a variety of conditions like epilepsy [1], ischemia [2], depression [3] and chronic neurodegenerative diseases like Alzheimer’s disease and Parkinson’s disease [4]. Without affecting the main pharmacophore, the deconstruction approach of known ligands can provide a clear rationale of essential structural elements. According to this deconstruction-reconstruction approach a lead compound is deconstructed by removal of structural elements and functional groups and subsequently reconstructed to produce a diverse set of compounds with different functional groups [5]. (Figure 1) essential pharmacophore could be derived from its phloroglucinol moiety. The first mono- and diacylated phloroglucinol compounds were tested for their hyperforin-like activity profile and it turned out that the active ones were also specific for TRPC6 but did not induce CYP3A4 [1,2]. Because of this more derivatives were synthesized and investigated for their structure-activity-relationships. It was shown that for the activation of TRPC6 two acyl side chains are necessary but their symmetry is of no importance. However, it seems that the activity increases only slightly along with the lipophilicity of the compounds. The molecules were tested in a calcium sensitive fluorescence assay in different concentrations using PC12 cells as a model for neuronal cells. Additionally the assay was performed with calcium-free extracellular buffer to differentiate between intracellular and extracellular calcium release and barium influx was measured to investigate TRPC6-like properties. We could also demonstrate that the known CYP3A4-inducing antibiotics doxycycline, erythromycin, griseofulvin, nafcillin and rifampicin have no activity towards TRPC6. Acknowledgments: We thank ERA-Net NEURON for funding the HypZiTRP consortium. References: 1. Leuner, K. et al.: Mol. Pharmacol. 2010, 77: 368–77. 2. Kandel, B.A. et al.: J. Pharmacol. Exp. Ther. 2014, 348: 393–400. POS.080 Selective activation of different pathways by dualsteric compounds in muscarinic M1 acetylcholine receptors Bödefeld, T.1; Messerer, R.2; Dallanoce, C.3; De Amici, M.3; Holzgrabe, U.2; Mohr, K.1; Schrage, R.1 1 Pharmacology The 3-benzazepine 1 is a conformationally restricted analogue of the flexible ligand ifenprodil with high GluN2B affinity [6]. In this project deconstruction of the lead compound 1 is envisaged, resulting in simplified compounds 3, which will be decorated with different functional group at the phenyl ring to obtain the compounds 4. This concept will allow the identification of essential structural elements essential for high GluN2B Affinity. Acknowledgments: A special thanks to Graduate School of Chemistry for the funding of S.D. during his PhD. References: 1. Löscher, W.; Rogawski, M. A.: Selected Works of Micheal Rogawski; 2002. 2. Di, X. et al.: Stroke 1997, 28: 2244-2251. 3. Mony, L. et al.: Br. J. Pharmacol. 2009, 157: 1301-1317. 4. Wessell, R. H. et al.: Neuropharmacol. 2004, 47: 184-194. 5. Chen, H. et al.: Drug Discov. Today 2015, 20: 105-117. 6. Wünsch, B. et al.: ChemMedChem 2010, 5: 687-695. POS.079 2, 4-Diacylphloroglucinols as classic transient receptor potential-6 activators Fritz, N.1; Eberle, J.1; Bouron, A.2,3,4; Nowak, G.5; Heinrich, M.6; Friedland, K.1 1 Department of Chemistry and Pharmacy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Cauerstraße 4, 91058 Erlangen, Germany 2 Université Grenoble Alpes, LCBM, F-38054 Grenoble, France 3 CNRS, UMR 5249, F-38054 Grenoble, France 4 CEA, LCBM, 17 rue des Martyrs, F-38054 Grenoble, France 5 Department of Neurobiology, Institute of Pharmacology, Polish Academy of Sciences, Smetna 12, 31-343 Krakow, Poland 6 Department of Chemistry and Pharmacy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Schuhstraße 19, 91052 Erlangen, Germany Hyperforin, the major antidepressant component of St. John´s wort, is a specific activator of canonical transient receptor potential-6 (TRPC6) and its antidepressive properties are mediated by activation of this nonselective cation channel. But there are a few disadvantages like its minor stability and its ability to induce Cytochrome P450 3A4 (CYP3A4), therefore leading to pharmacokinetic interactions with other drugs. As it is an acylated phloroglucinol derivative we hypothesized that the 144 • DPhG Annual Meeting 2015 Conference Book and Toxicology Section, Institute of Pharmacy, University of Bonn, GerhardDomagk-Straße 3, 53115 Bonn, Germany; of Pharmaceutical Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany; 3 Department of Pharmaceutical Sciences, University of Milan, Via Mangiagalli 25, 20133 Milan, Italy 2 Institut G protein-coupled receptors (GPCRs) respond to a broad range of different extracellular stimuli, thereby evoking intracellular signaling. Dualsteric compounds bind simultaneously to the receptor’s orthosteric transmitter binding site and its allosteric vestibule [1]. In the muscarinic acetylcholine M2 receptor (M2 mAChR) these dualsteric compounds were shown to impair the activation-related conformational change of the receptor protein by spatial restriction. This can eventually control the signaling pattern of a GPCR that promiscuously activates a variety of different signaling pathways [2]. In the present work, we checked whether this concept could be transferred to the M1-subtype of muscarinic receptors (M1 mAChR) which preferentially signals into Gq/11-dependent pathways, but can also activate Gs and Gi proteins [3]. Therefore, M1 receptor-mediated signaling induced by the orthosteric full agonist iperoxo and several dualsteric compounds were investigated. The latter consist of iperoxo as the orthosteric building block linked to an allosteric phthalimide (phth) or naphthalimide (naph) moiety via alkyl chains of different length. To distinguish between Gq/11- and Gs-dependent signaling pathways, CHO cells stably transfected with the human M1 muscarinic receptor were applied in IP1 and cAMP accumulation assays, respectively. Our findings show that the bulky allosteric naph residue impaired both signaling pathways to a greater extent than the smaller substituent phth. In particular, the hybrid iper-6-naph completely lost intrinsic activity at the M1 mAChR, although M2 mAChR activation by this compound had been demonstrated in a previous study [2]. In contrast to iper-6-naph, the less spacious congener iper-6-phth only lost intrinsic activity for Gs-activation, while it was still a partial agonist for Gq/11-dependent signaling. All other hybrids tested were able to activate both the Gq/11 protein and the Gs protein. Remarkably, iper-7-phth had a significantly higher efficacy for Gs protein activation than all the other compounds under investigation. However, there was no significant difference between iper-7-phth and iper-8-phth for Gq/11-dependent signaling. Furthermore, iper-7-naph and iper-8-naph showed no significant differences in both Gq/11- and Gsassays. Our data indicate that it might be possible to gain subtype selectivity by exploiting the allosteric moiety or the length of the linker chain. In particular, the bitopic derivative iper-6-naph was shown to be a partial GPCR/ION CHANNELS agonist at the M2 mAChR, but was not able to activate the M1 mAChR [2]. Taken together, these data demonstrate that, in comparison to Gq/11mediated signaling, activation of the Gs protein in M1 mAChR is more sensitive to spatial restriction in the allosteric vestibule. Thus, it is possible to control signaling of the M1 mAChR by allosteric constraint of the conformational flexibility. References: 1. Antony, J. et al.: FASEB J. 2009, 23:442-450. 2. Bock, A. et al.: Nat. Commun. 2012, 3:1044 doi: 10.1038/ncomms2028. 3. Gregory, K.J.; Sexton P.M.; Christopoulos A.: Curr. Neuropharmacol. 2007, 5:157-167. POS.081 mAChR - G protein dissociation kinetics reflect coupling efficiencies and allow quantification of G protein subtype selectivity. receptors (ADOR). The ADORA3 is a potential drug target [1,2], and several antagonists have been developed and pharmacologically evaluated [3,4]. Apparently this receptor is involved in the progression of inflammatory diseases, and shows ameliorating effects on plasma extravasation [4], asthma bronchiale, chronic obstructive pulmonary disease and idiopathic pulmonary fibrosis [5]. Moreover, ADORA3 antagonists may be useful for the treatment of cancer, either as monotherapy are in combination with chemotherapy [6]. The A3 receptor is coupled to Gi proteins mediating inhibition of adenylate cyclase [7]. In the present study quinazolines were modified by nucleophilic substitution at position 4 of the quinazoline scaffold. The potency increased considerable by the introduction of 3-aminopyrazole residues. Elongation with alkyl groups led to a dramatic increase in subtypeselectivity for the ADORA3. Additional substitution at position 6 and 7 of the quinazoline core led to extraordinarily potent and selective derivatives. Compound 1 was found the most potent derivative of the present series at human ADORA3 displaying affinity in the subnanomolar range (Ki = 0.248±0.017 nM) combined with high selectivity. Prokopets, O. S.1; Buenemann, M.1 1 Department of pharmacology and clinical pharmacy, Philipps-University of Marburg, Karl-vonFrisch Str.1, 35043 Marburg, Germany Although recent structural research gave an idea about the GPCR structure, the degree of selectivity of receptor-G protein coupling and its underlying mechanisms are still unclear. Here we establishing a quantitative method based on Fluorescence resonance energy transfer (FRET) for measuring the affinity of Gα-subunit towards muscarinic receptors and characterize dynamics of Go/i - and Gq/11-proteins binding to activated M1-, M2- and M3-AChRs and their subsequent dissociation in single permeabilized HEK293T cells under conditions of GTPdepletion. In accordance with currently accepted models of ternary complex formation of agonist, receptor Gα and Gβγ subunits we tested for Gαsubtype dependent stabilization of these complexes. As a measure of affinity we measured receptor G protein dissociation after agonist withdrawal and found fast Go and slow Gq proteins dissociation kinetics (>10-fold slower) from M3- and M1-AChRs during agonist withdrawal under the GTP depletion conditions. Furthermore, we observed a significant shift (> 15 fold) of concentration-response curves of Go proteins binding to M3-AChR in comparison to Gq. In order to relate these findings to the coupling efficiency of these receptors towards these G proteins we determined concentration response curves for G protein activity in intact cells by means of FRET. Our results show a > 10 fold higher sensitivity of Gq towards M3-AChR receptors compared to Go. To test for subtype selectivity of M2-AChR towards Gi/o proteins, we have determined the kinetics of Gi1-, Gi2-, Gi3-, and Go-proteins dissociation from M2-AChR. We found two-fold faster kinetics for Gi1 and Gi3 in comparison to Gi2 and Go proteins, indicating the existence of a subtype selectivity of M2-AChR for Gαi2 and Gαo containing G proteins. Taken together our study illustrates a suitable method for quantification of the affinity of receptor-G-protein interactions and shows that the affinity of this interaction reflects coupling efficiency and G protein selectivity for a given receptor. Acknowledgments: Heinrich-Heine University, Department of Lasermedicine, Universitätsstr. 1, 40225 Duesseldorf, Germany, Prof. Dr. Horst Lemoine POS.082 Synthesis of quinazoline derivatives as adenosine A3 receptor antagonists Acknowledgments: Thanks are due to J. Ortwein for HPLC analysis, Dr. L. Hennig for recording and analysis of NMR data and C. Vielmuth for the biological testing. References: 1. Fredholm, B. B. et al.: Pharmacol. Rev. 2011, 63(1): 1–34. 2. Gessi, S et al.: Pharmacol. Ther. 2008, 117: 123–140. 3. Lee, J. et al.: Am. J. Pathol. 2013, 183: 1488–97. 4. Mikus, E.G. et al.: Eur. J. Pharmacol. 2012, 699(1–3): 62–66. 5. Della Latta, V. et al.: Pharm. Res. 2013, 76: 182–189. 6. Merighi, S. et al.: Pharmacol. Ther. 2003, 100: 31–48. 7. Birnbaumer, L.: Biochim. Biophys. Acta 2007, 1768(4): 772–93. POS.083 CRTC1-deficient mice show cardiac hypertrophy and reduced cardiac function Morhenn, K.1,2; Geertz, B.3; Eschenhagen, T.2,3; Cardinaux, J.-R.4; Lutz, S. 5,6; Oetjen, E.1,2,7 1 Department of Clinical Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg Eppendorf, Martinistraße 52, 20246 Hamburg, Germany (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck 3 Department of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg Eppendorf, Martinistraße 52, 20246 Hamburg, Germany 4 Center for Psychiatric Neuroscience, Site Cery, 1008 Prilly-Lausanne, Switzerland 5 Department of Pharmacology, University Medical Center Göttingen, Robert Koch Straße 40, 37075 Göttingen, Germany 6 DZHK (German Center for Cardiovascular Research), Partner Site Göttingen 7 Institute of Pharmacy, University of Hamburg, Bundesstraße 45, 20146 Hamburg, Germany 2 DZHK Cardiac hypertrophy leads to heart failure, one of the common causes for hospitalization. Chronic β-adrenergic signaling contributes to the pathogenesis of cardiac hypertrophy, as evidenced by the therapeutic success of β-adrenoceptor antagonists. The cAMP Regulated Transcriptional Coactivator 1 (CRTC1) is regulated by increases in cAMP and calcium/calcineurin, as elicited by β-adrenergic signaling, both known to participate in the development of cardiac hypertrophy [1]. Our previous data showed that the protein content of CRTC1 is elevated in hearts of mice and humans under conditions of maladaptive hypertrophy. Mice globally deficient in Crtc1 show signs of hypertrophy indicated by a higher ratio of heart weight to tibia length as well as increased myocyte size. Lang, M.1; Hinz, S.2; Schäke, F.1; Kubicova, L.3; Müller, C. E.2; Briel, D.1 1 University of Leipzig, Institute of Pharmacy, Bruederstraße 34, 04103 Leipzig, Germany 2 Rheinische Friedrich-Wilhelms-Universität Bonn, Pharmazeutisches Institut, An der Immenburg 4, 53121 Bonn, Germany 3 University of Vienna, Division of Molecular Systems Biology, Faculty of Life Sciences, Althanstrasse 14, Vienna, A-1090, Austria Quinazolines that were modified at position 2, 4, 6 and 7 were synthesized to evaluate their structure-activity relationships at the four subtypes (A1, A2A, A2B and A3 [1]) of G-protein-coupled adenosine DPhG Annual Meeting 2015 Conference Book • 145 POSTERS To study the role of CRTC1 in the pathogenesis of cardiac hypertrophy, in the present study Crtc1-deficient mice were further investigated. Echocardiographically assessed, the ejection fraction, the fractional area shortening and the cardiac output were reduced by 47±8%, 49±12% and 42±9%, respectively, in Crtc1-deficient mice compared to their wild-type littermates, indicating a systolic dysfunction of the heart. Meanwhile, Serca and Phospholamban protein contents remained unchanged, as measured by immunoblot. As measured by RT-qPCR, no differences in mRNA levels of the markers for cardiac hypertrophy Nppa, Nppb, Acta1 and Myh7, as well as the pro-fibrotic Ctgf were observed. It is known that the Regulator of G-Protein Signaling 2 (RGS2) reduces hypertrophy via reduction of Gαq-protein induced signaling and that Rgs2 gene transcription is induced by CREB [2,3]. We previously showed a stimulation of the transcriptional activity of the Rgs2 promoter by CRTC1 in HEK cells and decreased Rgs2 mRNA and protein levels in Crtc1deficient mice. By chromatin immunoprecipitation, we now showed the recruitment of endogenous CRTC1 to the Rgs2 promoter in cardiac tissue. mRNA levels of Rgs3, Rgs4, Rgs5 and Rgs6 did not differ in Crtc1-deficient mice. In conclusion, our data indicate that increased CRTC1 protein content in maladaptive cardiac hypertrophy is a compensatory mechanism to delay disease progression, in part by upregulation of RGS2. Thus, CRTC1 might represent a novel player in cardiac hypertrophy. References: 1. Bittinger, M. et al.: Curr. Biol. 2004, 14(23): 2156-61. 2. Xie, Z. et al.: J. Biol. Chem. 2011, 286(52): 44646-58. 3. Zhang, P.; Mende, U.: Trends Cardiovasc. Med. 2014, 24(2): 85-93. POS.084 A homogenous A2B adenosine receptor fluorescence binding assay based on flow cytometry POS.085 GPR17: Understanding its signaling and physiological function Simon, K.1; Hennen, S.1; Merten, N.1; Blättermann, S.1; Gomeza, J.1; Kostenis, E.1 1 Institute for Pharmaceutical Biology, University of Bonn, Nussallee 6, 53115 Bonn, Germany Oligodendrocyte differentiation and maturation, ultimately leading to formation of myelin sheaths, is regulated temporally during postnatal development but also during repair of demyelinating lesions [1]. Recent studies suggest G protein-coupled receptors (GPCRs) as key players in the development of proliferating oligodendrocyte precursor cells to myelinating oligodendrocytes [1,2,3]. Activation of the orphan GPCR GPR17, which is predominantly expressed in the oligodendrocytelineage cells of the central nervous system (CNS), has been linked to arrest of primary mouse oligodendrocytes in an immature, less myelinating stage [1,3]. Accordingly, GPR17 inhibition emerges as therapeutic strategy to enhance repair of lost myelin sheaths in demyelinating diseases, such as multiple sclerosis (MS). However, the intracellular signaling pathways linking activated GPR17 to oligodendrocyte differentiation block are poorly understood at present. To further delineate these downstream components, we took advantage of the recently identified first surrogate GPR17-agonist MDL29,951 [3] and applied this molecule in Oli-neu cells, an immortalized cell line derived from primary murine oligodendrocytes [4], and primary rat oligodendrocyte cultures isolated from brain tissue. We anticipate that detailed understanding of cellular mechanisms governing GPR17mediated inhibition of oligodendrocyte differentiation will aid to develop novel therapies for demyelinating pathologies. References: 1. Chen, Y. et al.: Nat. Neurosci. 2009, 12: 1398–1406. 2. Ackerman, S. D. et al.: Nat Commun 2015, 6: 6122. 3. Hennen, S. et al.: Sci. Signal 2013, 6: ra93. 4. Jung, M. et al.: Eur. J. Neurosci. 1995, 7: 1245–1265. Köse, M.1; Karcz, T.2; Gollos, S.1; Fiene, A.1; Heisig, F.1; Spanier, C.1, Kieć-Kononowicz, K.2; Müller, C. E. E.1 1 PharmaCenter Bonn, Pharmaceutical Chemistry I, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany 2 Department of Technology and Biotechnology of Drugs, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland Homogenous binding assays based on fluorescence measurement represent attractive alternatives to the traditional radioligand binding assays as they are faster, cheaper and less hazardous. Recently the first fluorescence binding assay for the A3 adenosine receptor (AR) was described [1,2]. So far, no fluorescence binding assay is available for the human A2B AR. In the present study, we developed a new flow cytometry-based fluorescence binding assay for the human A2B AR using a newly synthesized fluorescent antagonist PSB-12105. In radioligand binding assays PSB-12105 was shown to display high affinity at the human A2B receptor with a Ki value of 15.0 nM. It was also found to be selective versus the A1, A2A and A3 receptor subtypes. CHO cells stably transfected with A2B ARs were preincubated with adenosine deaminase (ADA) for 2 h at 37 °C before adding the fluorescent ligand PSB 12105 (1 nM). Nonspecific binding was determined in the presence of the potent and selective A2B antagonist PSB-603 (100 nM). The cell suspension was incubated for 1 h at 37 °C. The fluorescence was subsequently measured using flow cytometry. We determined an IC50 value of 0.543 nM for the standard A2B antagonist PSB 603 which is in good agreement with the Ki value determined in radioligand binding assays (0.553 nM) [3]. The results indicate that our assay is robust and reproducible and provides a simple, convenient alternative method to radioligand binding assays adaptable to highthroughput screening. To our knowledge, this is the first fluorescence binding assay for the human A2B AR reported to date. This new method may serve as an important pharmacological tool for the development and investigation of new A2B AR ligands. Acknowledgments: Partly supported by Polish National Science Center founding, project DEC2012/04/M/NZ4/00219 and GLISTEN COST Action CM1207 References: 1. Kozma, E. et al.: Biochem. Pharmacol. 2013, 85: 1171-1181. 2. Kozma, E. et al.: Biochem. Pharmacol. 2012, 83: 1552-1561. 3. Borrmann, T. et al.: J. Med. Chem. 2009, 52: 3994-4006. POS.086 Novel 6-benzamidochromen-4-one-2-carboxylic acids as pharmacological tools for investigating the orphan receptor GPR35 Meyer, A.1; Funke, M.1; Thimm, D.1; Müller, C. E.1 1 Pharmaceutical Institute, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany G protein-coupled receptors (GPCRs) are the largest protein family in the human genome with about 800 different sequences. about one third of all marketed drugs and also those in (pre)clinical development are targeting GPCRs. GPR35 is an orphan GPCR whose endogenous ligand is still unknown. It is expressed in the gastrointestinal tract and in liver, in the immune, cardiovascular and the central nervous system. GPR35 agonists have potential for the treatment of inflammation and pain whereas antagonists might be useful for treating cardiovascular diseases. We identified and developed a novel class of potent GPR35 agonists based on the chromen-4-one scaffold [1]. One of our most potent agonists, PSB 13253, was obtained in tritium-labeled form with a specific activity of 36 Ci (1.33 TBq) / mmol. [3H]PSB-13253 displayed a KD value of 5.27 nM, and its binding was saturable and reversible. [3H]PSB 13253 is the first radioligand for GPR35 and represents a useful tool for investigating this poorly studied orphan receptor. Based on radioligand binding studies we further optimized the series developing agonists with subnanomolar affinity, e.g. 6 bromo-8-(2,6-difluoro-4methoxybenzamido)-4-oxo-4H-chromene-2-carboxylic acid (PSB13007). Its Ki value of 0.589 nM makes it the most potent GPR35 agonist known to date [2]. With a calculated clogP of 2.6 and a molecular weight of 454 g/mol it meets the criteria for drug-likeness. In summary, a new series of 8 benzamidochromen-4-one-2-carboxylic acid derivatives was developed resulting in valuable tools for the characterization of the so far poorly studied GPR35, which may represent a new drug target. References: 1. Funke, M. et al.: J. Med. Chem. 2013, 56: 5182-5197. 2. Thimm, D. et al.: J. Med. Chem. 2013, 56: 7084-7099. 146 • DPhG Annual Meeting 2015 Conference Book GPCR/ION CHANNELS POS.087 Binding mode prediction and validation of bile acid and neurosteroid agonists of the G-Protein coupled receptor TGR5 Gertzen, C. G. W.1; Spomer, L.2; Häussinger, D.2; Keitel, V.2; Gohlke, H.1 1 Institute for Pharmaceutical and Medicinal Chemistry, Universitätsstr. 1, 40225 Düsseldorf, Germany 2 Clinic for Gastroenterology, Hepatology, and Infectious Diseases, Heinrich-Heine-University, Moorenstr. 5, 40225 Düsseldorf, Germany The structurally unknown G-protein coupled bile acid receptor (GPCR) TGR5 is the first bile acid sensing GPCR and directly interacts with several G-protein subtypes [1]. Tissues with high expression levels of TGR5 include the brain, the liver, and the gastrointestinal tract. TGR5 is an emerging target for the treatment of metabolic diseases [2-4]. Therefore, developing selective and potent agonists of TGR5 is of high importance [5]. However, without an experimentally determined binding mode, the rational design of compounds is difficult. Recently, Macchiarulo et al. [6] proposed a binding mode of natural and synthetic bile acids in TGR5 based on single template homology modeling, molecular docking, and mutational analysis. However, this binding mode does not interact with transmembrane helices (TM) 5 and 6, which are crucial for GPCR activation [7, 8]. Additionally, the binding mode of Macchiarulo et al. does not address E169 in TM 5, which the authors found to be important for receptor activation and is a conserved residue among the TGR5 family [6]. Here, we present an experimentally validated binding mode of 68 TGR5 agonists, including natural and synthetic bile acids and neurosteroids. Our strategy consisted of multi-template homology modeling, molecular docking, and structure-based 3D-QSAR with subsequent mutational analysis and molecular dynamics simulations. After two cycles of this strategy, the binding mode model of the TGR5 agonists results in a good 3D-QSAR model (q² = 0.50), thus indicating that differences in the agonist structures correlate with differences in experimentally determined pEC50 values in the predicted binding mode. Based on this binding mode, nine mutants of binding site residues were suggested that should either influence agonist binding or TGR5 activation. Activity analysis using cAMP reporter gene assays and FACS analysis for membrane localization confirmed these predictions in all cases. In particular, through the Y240F mutant we could demonstrate the importance of hydrogenbonding interactions between the tyrosine and TGR5 agonists. Additionally, we identified the stereoselectivity-determining residue Y89 for hydroxyl-groups in position seven on the cholane scaffold. This provides strong support to the validity of the binding mode. Our binding mode differs from the binding mode by Macchiarulo et al. in three important aspects: I) The cholane moiety is rotated by 180°; II) the sidechains of bile acids bind to R79, which is 12 Å away from the respective interaction partner postulated in [6]; III) agonists address residues in TM 5 and 6, which are essential for receptor activation. Our binding mode could facilitate the structure-based design of new TGR5 agonists. Acknowledgments: We are grateful to the ‘‘Zentrum für Informations und Medientechnologie’’ (ZIM) at the Heinrich Heine University for computational support, Stefanie Lindner und Waltraud Kuß for technical assistance, and to Dr. Nadine Homeyer, Yasemin Bilgic, and Alina Völz for help with the molecular modeling. This work was supported by the Deutsche Forschungsgemeinschaft through the Collaborative Research Center SFB 974 (‘‘Communication and Systems Relevance during Liver Damage and Regeneration’’, Düsseldorf) and the Clinical Research Group KFO 217 (‘‘Hepatobiliary Transport in Health and Disease’’, Düsseldorf). References: 1. Hannah, M.: International Journal of Interferon, Cytokine and Mediator Research 2014, 6: 27-38. 2. Pols, T. W. H. et al.: Cell Metab. 2011, 14: 747-757. 3. Pols, T. W. H. et al.: J. Hepatol. 2011, 54: 1263-1272. 4. Perino, A. et al.: The Journal of Clinical Investigation 2014, 124: 5424-5436. 5. Sato, H. et al.: J. Med. Chem. 2008, 51: 1831-1841. 6. Macchiarulo, A. et al.: ACS Med. Chem. Lett. 2013, 4: 1158-1162. 7. Zimmerman, B. et al.: Sci. Signal. 2012, 5: ra33. 8. Xu, F. et al.: Science 2011, 332: 322-327. POS.088 Differential modulation of Gβγ effectors by ligands targeting GPCRs Büllesbach, K.1; Bautista, O.2; Schröder, R.1; Gütschow, M.2; Kostenis, E.1 1 Molecular-, Cellular- and Pharmacobiology Section, Institute of Pharmaceutical Biology, University of Bonn, Bonn, Germany 2 Pharmaceutical Institute, Pharmaceutical Chemistry I, University of Bonn, Bonn, Germany G protein-coupled receptors constitute the largest family of membrane signaling proteins. They respond to a wide array of stimuli and serve to translate extracellular information to the inside of the cell via coupling to heterotrimeric αβγ G proteins [1]. G proteins are central to signal transduction because they connect GPCRs to diverse intracellular effectors whose activities they regulate via both Gα and Gβγ subunit complexes. A common dogma in G protein signaling stated that G protein heterotrimers are regulated uniformly by activated GPCRs, whereby receptors discriminate between individual G protein isoforms depending on the inherent bias of each ligand used to stimulate the receptor. We recently challenged this dogma demonstrating that the small molecule Gue1654 acts as a non-competitive inhibitor of OXE-R, a GPCR responding to the endogenous lipid mediator 5-oxo-eicosatetraenoic acid [2]. Specifically, Gue1654 exclusively inhibited Gβγ but not Gαi signaling triggered upon activation of OXE-R in both recombinant and human primary cells. The question therefore arose whether biased perturbation of G protein heterotrimers can extend to effectors further downstream of Gβγ. To this end we investigated the ability of Gue1654 to discern between effectors of Gβγ such as extracellular regulated kinases 1 and 2 (ERK1/2), phospholipase Cβ (PLCβ), and G protein inward-rectifying potassium (GIRK) channels [3] in human embryonic kidney (HEK293) cells forcibly expressing OXE-R. We compared the resulting profile to that obtained with the small molecule Gβγ inhibitor Gallein [4], which targets a hotspot on the Gβ protein thereby preventing Gβγ from interacting with its downstream effector molecules in a receptorindependent fashion. We find that both Gue1654 and Gallein completely blunt IP1 production, yet differ in their inhibition profile concerning activation of ERK1/2 and GIRK channels. This pattern of inhibition is distinct from that observed with canonical competitive OXE-R inhibitors [5] which blunt Gβγ downstream signaling equally. Our data therefore suggest capacity to selectively engage or disrupt individual Gβγ downstream effects in a receptor-specific manner demonstrating another layer of regulation within the GPCR-G protein signaling axis. Acknowledgments: K.B. is a member of the DFG-funded Research Training Group RTG 1873 References: 1. Rosenbaum, D. M. et al.: Nature. 2009, 459(7245):356-63. 2. Blättermann, S. et al.: Nat. Chem. Biol. 2012, 8(7): 631-8. 3. Clapham, D. E.; Neer E. J.: Annu. Rev. Pharmacol. Toxicol. 1997, 37: 167-203. 4. Lehmann, D. M. et al.: Mol. Pharmacol. 2008, 73(2): 410-8. 5. Gore, V. et al.: J. Med. Chem. 2014, 57(2): 364-77. POS.089 A mechanistic view on GPCR modulation Bermudez, M.1; Wolber, G.1 Computer-Aided Drug Design, Institute of Pharmacy, Department Pharmaceutical Chemistry, Freie Universität Berlin, 14195 Berlin 1 G-protein coupled receptors (GPCRs) enable the transmission of signals into cells, which makes them highly interesting drug targets. Recent achievements in GPCR crystallography provide us with new structural data on GPCRs in distinct activation states [1]. However, these structures represent only a single static view on highly flexible proteins that display their functionality by recognizing extracellular stimuli, subsequent adaption of their conformation and hence allows for an intracellular response [2]. The combination of crystallographic data with state-of-theart computer-driven simulations allows for a mechanistic view on the first two steps of GPCR function: ligand binding and the resulting conformational change. Taking muscarinic acetylcholine receptors (MAChRs) as representative examples we explained how GPCR can be modulated in a predictable fashion. DPhG Annual Meeting 2015 Conference Book • 147 POSTERS Our mechanistic models comprise both inactive and active-like receptor states (Figure). After a characterization of the orthosteric binding side, we focused on dualsteric ligand binding [3,4]. These kinds of ligands simultaneously bind to the orthosteric and the allosteric binding site and combine the high affinity of orthosteric ligands with the high specificity of the allosteric binding site. Here we report the structural basis for a specific modulation of MAChRs focusing on dualsteric ligand binding. This includes subtype selectivity, biased signaling and a novel concept for partial agonism. Our dynamic models illustrate how distinct conformational states can be stabilized in a ligand-dependent manner. Finally, our structural and functional models proved their high explanatory power in biological experiments and offer the possibility to rationally design specific modulators for MAChRs but also for other GPCRs. comparing the binding modes of known antagonists for MCH1R and H3R will allow us to identify potent dual antagonists of H3R and MCH1R. Acknowledgments: Financial support for David Schaller by the Elsa-Neumann-Scholarship is gratefully acknowledged. References: 1. Kopelman, P. G.: Nature 2000, 404: 635-43. 2. Kim, G. W. et al.: Clin. Pharmacol. Ther. 2014, 95(1): 53-66. 3. Singh, M.; Jadhav, H. R.: Mini Rev. Med. Chem. 2013, 13(1): 47-57. 4. Szalai, K. K. et al.: Recent. Pat. CNS Drug Discov. 2014, 9(2): 122-40. 5. Parks, G. S. et al.: J. Physiol. 2014, 592(10): 2183-2196. 6. Thorsen, T. S. et al.: Structure 2014, 22(11): 1657-1664. 7. Chien, E. Y. et al.: Science 2010, 330(6007): 1091-5. 8. Shimamura, T. et al.: Nature 2011, 475(7354): 65-70. POS.091 Histamine H4 receptor affinity of 1,3,5-triazine derivatives Kamińska, K.1; Więcek, M.1; Adami, M.2; Stark, H.3; Kieć-Kononowicz, K.1 1 Department of Technology and Biotechnology of Drugs, Jagiellonian University Medical College, Medyczna 9, 30-688 Cracow, Poland. 2 Department Neuroscience, University of Parma, Via Volturno 39, 43125 Parma , Italy. 3 Institute of Pharmaceutical and Medicinal Chemistry, Heinrich-Heine-University, Universitaetsstr. 1, 40225 Duesseldorf, Germany. Figure: Superimposition of homology models for all subtypes of muscarinic receptors in different activation states. References: 1. Venkatakrishnan, A. J. et al.: Nature 2013, 494(7436): 185–194. 2. Bermudez, M.; Wolber, G.: Bioorg. Med. Chem. 2015, 23(14): 3907-3912. 3. Schmitz, J. et al.: J. Med. Chem. 2014, 57(15):6739-50. 4. Bermudez, M. et al.: Mol. Inf. e-pub ahead of print, 2015. POS.090 Rational multi-target drug design by ligand-guided homology modeling Schaller, D.1; Wolber, G.1 1 Institut für Pharmazie, Freie Universität Berlin, Königin-Luise-Str. 2+4, 14195 Berlin Obesity has become a major human health risk and is associated with many serious diseases including type 2 diabetes, stroke and cancer [1]. Although being studied for decades, anti-obesity drugs still lack efficacy and show serious or unpleasant side effects [2]. Using a chemogenomics approach we identified histamine H3 receptor (H3R) and melaninconcentrating hormone receptor 1 (MCH1R) as promising target pair for the development of multi-target directed ligands. Targeting these single receptors alone did not yet result in an effective obesity treatment [3,4]. Recent studies suggest that antagonizing H3R and MCH1R at the same time may induce a synergistic effect [5]. This study aims at generating homology models that are able to explain the structure activity relationship of known antagonists of H3R. To achieve our goal we implemented a ligand-guided homology modeling workflow that is able to sample the conformational space of side chains in the orthosteric binding pocket of H3R. The increasing number of crystal structures of G protein-coupled receptors (GPCRs) allows the identification of comparable interaction patterns between bound ligands and receptors. In particular an ionic interaction between the tertiary or quaternary amine of ligands and the conserved residue ASP3.32 of GPCRs can be observed in many crystal structures including muscarinic receptors (4U15), dopamine receptors (3PBL) and histamine H1 receptor (3RZE) [6,7,8]. The crystal structure 3RZE of H1R was used as template to create 1000 homology models of H3R. A specific set of tertiary aminecontaining H3R antagonists was docked against these models. Subsequently, the homology models were ranked by calculating the distance between the tertiary amine of the docked ligands and the key amino acid ASP3.32 of H3R. Analyzing the resulting models revealed distinct side chain conformations that favor the interaction of the tertiary amines with ASP3.32 of H3R. This knowledge was employed to create homology models that explain the structure activity relationship of known ligands in a highly consistent way. Several steps of minimization and redocking were performed to ensure an optimized geometry. This methodology is adaptable to other GPCRs like MCH1R that show the described ionic interaction with bound ligands. Elucidating and 148 • DPhG Annual Meeting 2015 Conference Book Histamine plays its function through binding with four already known histamine receptors, designed as H1-H4. It is assumed, that the youngest member of the family – histamine H4 receptor (H4R), which was discovered and cloned in 2000/2001 by several independent research groups [1], is involved in inflammatory processes and immune responses, because of its mainly expression in various cells of the immune system (monocytes, mast cells, dendritic cells, eosinophils and basophils) [2]. Potential therapeutic effects of H4R antagonists/inverse agonists in animal models of acute inflammations, allergic rhinitis, asthma or pruritus were confirmed [3]. As physiological role of H4R is not clear - new, potent and selective ligands are required to investigate its action. Among H4R ligands already described in the literature and patent data there can be found a large group of triazine derivatives [4,5]. The aim of this study was to evaluate in vivo activity of five 4-(4methylpiperazin-1-yl)-1,3,5-triazine derivatives (KB-4, KB-30, JN-38, TR11 and TR-40), Compounds were tested in croton oil-induced ear edema model and ear pruritus model in vivo in mice. Compounds examined in the presented studies were selected from the library of compounds synthesized in our Department. The obtained results showed that pre-treatment with KB-4, KB-30 or TR11 has strong to moderate influence on ear edema and pruritus. Among the tested compounds, KB-4 and TR-11 seem to have the most favorable profile, combining a good affinity at the human histamine H4 receptor with a high efficacy in the intact animal. The results in detail will be presented and discussed. Acknowledgments: This work was kindly supported by National Science Center DEC2011/02/A/NZ4/00031 and GLISTEN: COST Action CM1207. References: 1. Nguyen, T. et al.: Mol. Pharmacol. 2001, 59(3): 427-433. 2. Walter, M. et al.: Eur. J. Pharmacol. 2011, 668(1-2): 1-5. 3. Tiligada, E. et al.: Expert Opin. Investig. Drugs. 2009, 18(10): 1519-1531. 4. Kiss, R.; Reserű, G.M.: Expert Opin. Ther. Pat. 2012, 22(3): 205-221. 5. Stark, H.: H4 receptor: A Novel Drug Target in Immunomodulation and Inflamation , 2012 (Versita). POS.092 Multiple targeting with histamine H3 receptor antagonists: Epilepsy Schwed, J. S.1,2; Sadek, B.3; Khan, N.3; Subramanian, D.3; Weizel, L.2; Walter, M.2; Stark, H.1,2 1 Institute of Pharmaceutical and Medicinal Chemistry, Heinrich Heine University, Universitaetsstr. 1, 40225 Duesseldorf, Germany 2 Biocenter, Institute of Pharmaceutical Chemistry, Goethe University, Max-von-Laue-Str. 9, 60438 Frankfurt, Germany 3 Department of Pharmacology and Therapeutics, College of Medicine & Health Sciences, P.O. Box 17666, Al Ain 0097, United Arab Emirates University, United Arab Emirates Epilepsy is a chronic disorder of the brain and characterised by an enduring predisposition to generate recurrent epileptic seizures and by the neurobiological, cognitive, psychological and social consequences of GPCR/ION CHANNELS this condition [1]. For medical therapy different antiepileptic drugs (AEDs) are in use, but are hampered in clinical usage by either having an effectiveness within a maximum of 60-80% of all patients and diverse unwanted side effects like headache, nausea, cognitive impairment etc. [2]. Central histamine was found to play an important role in epilepsy [3], especially the third histamine receptor subtype (H3R) has by neurotransmitter modulating abilities raised hopes for novel AEDs [3,4]. Pitolisant (Wakix®), a non-imidazole H3R antagonist/inverse agonist submitted to EMA for market authorisation, was found to be effective in different in vitro and in vivo animal seizure models [3] and also in photosensitivity epilepsy in human patients [5]. Safinamide (Xadago®), a recently marketed anti-parkinson MAO B inhibitor, was primarily designed as an AED [6] and shown to have good affinity at voltage-gated Na+ and Ca++ channels as well as good inhibitory potency on glutamate release. As a multi-targeting or polypharmacological approach we combined the pharmacophore elements of H3R antagonist to that of safinamide or phenytoin with different spacer moieties. All compounds showed moderate to good affinity at hH3R (pKi values between 7.36 and 8.21) with good selectivity profile among the other histamine receptor subtypes tested (hH1R and hH4R, pKi values < 5). The ligands were further characterized in vivo in different convulsion models in rats using chemical-induced (pentylenetetrazole (PTZ)- or strychnine (STR)-induced seizure) or in maximum electroshock-induced seizure models (MES). The safinamide related derivatives with bulky elements showed significantly and dose-dependently reduced seizures or exhibited full protection in MES and PTZ convulsions model with some stereochemical preference for the R-configured isomer. Acknowledgments: This work was partly supported by the DFG (INST 208/664-1) as well as by the European COST Actions CM1103 and CM1207. Grants are from CMHS, UAE University. References: 1. Fisher, R. S. et al.: Epilepsia 2005, 46: 470-472. 2. Sadek, B. et al.: Eur. J. Med. Chem. 2014, 77: 269-279. 3. Bhowmik, M. et al.: Br. J. Pharm. 2012, 167: 1398-1414. 4. Sander, K. et al.: Biol. Pharm. Bull. 2008, 31: 2163-2181. 5. Bialer, M. et al.: Epilepsy Res. 2015, 111: 85-141. 6. Fariello, R. G.: Neurotherapeutics 2007, 4: 110-116. POS.093 Derivates of imidazolylpropylguanidine (SK&F-91486): Synthesis and pharmacological in vitro activities at histamine receptors (hH1,2,3,4R and gpH1,2R) Pockes, S.1; Buschauer, A.1; Elz, S.1 1 Institute of Pharmacy, University of Regensburg, Universitätsstraße 31, 93053 Regensburg, Germany 3-(1H-Imidazol-4-yl)propylguanidine (SK&F-91486 [1]) is the long-known prototypic pharmacophore of highly potent histamine H2-receptor (H2R) agonists of the guanidine class of compounds including, e.g., arpromidine [2] and the recently discovered acylguanidines [3]. In order to gain more insight into the structure-activity relationship of simple analogues of SK&F-91486, we started a project aiming at the synthesis and in vitro characterisation of alkylated imidazolylpropylguanidines and synthesised alkylated, aminoalkylated, ureidoalkylated and guanidinoalkylated derivatives of SK&F-91486 as well as the corresponding dimers, equipped with different spacer lenghts (C3-C12) (1). Furthermore, we picked out the most promising molecules of each group and replaced the imidazol-4-yl group by the bioisosteric moieties imidazol-1-yl, 2-amino-4-methylthiazol-5-yl and 2-aminothiazol-5-yl, respectively, to study their influence on histamine receptor subtype selectivity. In addition, we created a batch of simple alkylated imidazolylpropylcyanoguanidines (2) belonging to the imidazol-4-yl and the imidazol-1-yl series. To finalise the project, we also synthesised a few derivatives by heteroatomic exchange at the guanidine group to give, for example, (acylated) urea or thiourea analogues of SK&F-91486. We analysed the in vitro properties of 78 compounds in the guinea-pig ileum assay (gpH1R), the guinea-pig right atrium assay (gpH2R), in radioligand binding assays (hH1,2,3,4R), and in the [35S]GTPγS assay (hH1,2,3,4R). There were just a small number of compounds with mentionable affinity at gp/hH1R. A longer alkyl spacer length afforded a higher (agonistic) affinity at the gpH2R as well as at the hH2,3,4R. Dimeric compounds turned out to have highest (agonistic) affinity at these receptors (gpH2R: pEC50 ≈ 8.5, hH2,3,4R: pKi ≈ 7.5-8.0). Bioisosteric exchange with imidazol-1-yl decreased the affinity at each receptor. Replacement with amino(methyl)thiazoles gave only decreased affinities at hH3,4R, affording selective ligands for gp/hH2R. Surprisingly, two compounds of the imidazol-1-ylpropylcyanoguanidines proved to be strong agonists at hH3,4R (hH3,4R: pEC50 ≈ 10.0-11.4, pKi ≈ 9.3-9.4), while being nearly inactive vis-à-vis the gp/hH1,2R. References: 1. Parsons, M. E. et al.: Agents Actions 1975, 5: 464. 2. Buschauer, A.: J. Med. Chem. 1989, 32: 1963-1970. 3. Igel, P. et al.: J. Med. Chem. 2009, 52: 2623-2627. POS.094 Multiple targeting with histamine H3 receptor antagonists: Morbus Parkinson Affini, A.1; Walter, M.2; Schwed, J. S.1,2; Esteban, G.3; Unzeta, M.3; Stark, H.1 1 Institute of Pharmaceutical and Medicinal Chemistry, Heinrich Heine University, Universitaetsstr. 1, 40225 Düsseldorf, Germany 2 Biocenter, Institute of Pharmaceutical Chemistry, Goethe University, Max-von-Laue-Str. 9, 60438 Frankfurt, Germany 3 Universitat Autonoma de Barcelona, Institut de Neuroscìencies, Department de Bioquimica i Biologia Molecular, 08193 Barcelona, Spain Parkinson’s disease (PD) is among the most common neurodegenerative diseases and a major health problem in the aging society. Dopaminergic therapy including L-DOPA is highly effective during the early stages of the treatment, but after several years of medications is complicated by motor fluctuations and the onset of dyskinesia [1,2]. Safinamide (Xadago®) has recently been introduced as a reversible monoamine oxidase B (MAO-B) inhibitor (IC50= 9.0 nM) with some additional properties for Parkinson add-on therapy [3]. In the non-dopaminergic therapy development line, pitolisant (Wakix®) is in late stage of clinical premarket development for the treatment of excessive daytime sleepiness with Parkinson patients [4]. This histamine H3 receptor (H3R) antagonist (Ki = 0.3-1.0 nM) possess procognitive and wake-enhancing properties [5]. Since both approaches produce different effects in vivo on different targets for the same disease, the combination of both pharmacophore elements in one molecule may produce additional or synergistic effects with simple interaction and pharmacokinetic profiles. This multi-targeted or polypharmacological [6] approach may be promising on the H3R in combination with MAO-B inhibition for novel therapeutics for Parkinson´s disease. Here, we have combined the basic H3R pharmacophore with the main elements of safinamide and tested this small series of compounds for hH3R affinity as well as for MAO-A and MAO-B inhibitory potencies. All compounds showed affinities in the nanomolar concentration range at hH3R and moderate inhibitory potency at MAO-B isoenzyme in the micromolar concentration range. The compounds showed slight stereochemical discrimination with MAO-A and –B inhibition as also observed with the parent safinamide structure. So far, it is unclear what would be the best balance of H3R affinity and MAO-B inhibitory potency. The number of derivatives prepared is too small to drive any general conclusion, but as a proof-of-concept the combination of the specific G-protein coupled receptor affinity and the enzyme inhibitory property has simultaneously been shown for both targets. The more we understand the molecular mechanism of multifactorial diseases, the better we can design on this polypharmacological approach novel compounds with designed profiles for a potentially improved medical therapy. Acknowledgments: This work was partly supported by the DFG (INST 208/664-1) as well as by the European COST Actions CM1103 and CM1207. References: 1. Meissner, W.-G. et al.: Nat. Rev. Drug Discov. 2011, 10: 337-393. 2. Kassel, S. et al.: Eur. Neuropsychopharmacol. 2015, in press. 3. Borgohain, R. et al.: Mov. Disord. 2014, 29, 1273-1280. 4. Schwartz, J.- C.: Br. J. Pharmacol. 2011, 163: 713-721. 5. Sander, K. et al.: Biol. Pharm. Bull. 2008, 31: 2163-2181. 6. Nikolic, K. et al: J. Taiwan Inst. Chem. Eng. 2015, 46: 15-29. DPhG Annual Meeting 2015 Conference Book • 149 POSTERS POS.095 Studies on biased signaling of novel dopamine D2 and D3 receptor ligands Kassel, S.1; Saur, O.2; Kottke, T.1,2; Stark, H.1 Institute of Pharmaceutical and Medicinal Chemistry, Heinrich-Heine-University, Universitaetsstr. 1, 40225 Duesseldorf, Germany Biocenter, Institute of Pharmaceutical Chemistry, Goethe University, Max-von-Laue-Str. 9, 60438 Frankfurt, Germany 1 2 The knowledge on the structural and functional properties of G-protein coupled receptors as one of the most important targets for drugs has tremendously increased within the last decade. Ligands acting at the same G-protein coupled receptor can stabilize multiple and distinct receptor conformations linked to different signaling pathways and functional outcomes, termed as biased signaling [1]. The investigation of biased signaling pathways for dopamine D2 and D3 receptor ligands provides the opportunity to separate wanted therapeutic effects from unwanted side effects in the treatment of dopamine-related diseases [2], e.g. biased signaling behavior was described for the antipsychotic agent aripiprazole approved for the treatment of schizophrenia [1,3]. Aripiprazole was assumed as a partial agonist at the dopamine D2 receptor explored for cAMP inhibition or β arrestin recruitment, but appears as an antagonist of Gβγ signaling and in GTPγS binding assay at dopamine D2 receptors. Aripiprazole´s unique signaling profile is considered to be responsible for its antipsychotic efficacy producing only minimal extrapyramidal symptoms, leading to its consideration as third generation antipsychotic [4]. In our study a N-propyl etrabamine-like derivatives were combined via an alkyl spacer of different length to an most probably orthosteric 2,3-dichlorophenylpiperazine substructure like the key element of aripiprazole´s pharmacophore [4]. Determinants for affinity, selectivity and functional activities of synthesized compounds were examined. Biased signaling behavior and dependencies on the linker length were investigated evaluating G-protein activation and ERK1/2/CREB phosphorylation. The compounds showed high affinity at dopamine D2 and D3 receptors in the nanomolar concentration ranges with slight preference for the D3 receptor. Biased signaling on D2 receptors could be obtained after introduction of an extended alkyl linker, whereas connection via an ethyl linker showed no biased signaling. Our studies may give new impetus in investigation of novel and high affine D2/D3 receptor agonists [5] with unique signaling patterns, hoping to disclose and discriminate the therapeutically most relevant pathways in dopamine-related disorders like schizophrenia or Parkinson´s disease. Acknowledgments: This work was partly supported by the DFG (INST 208/664-1) as well as by the European COST Actions CM1103 and CM1207. References: 1. Shonberg, J. et al.: Med. Res. Rev. 2014, 4: 1286−1330. 2. Beaulieu, J.-M.; Gainetdinov, P. R.: Pharmacol. Rev. 2011, 63: 182−217. 3. Brust, T.: Biochem. Pharmacol. 2015, 93: 85-91. 4. Stark, H. et al.: International Patent PCT WO 2009 056805 A1. 2009 (07.05.2009). 5. Kassel, S. et al.: Eur. Neuropsychopharmacol. 2015, in press. 150 • DPhG Annual Meeting 2015 Conference Book CANCER/EPIGENETICS 4.7 Cancer/Epigenetics POS.096 Inhibition of HSP90 as a tool to increase chemosensitivity towards cisplatin in the human ovarian cancer cell line A2780 Rodrigues Moita, A. J.1; Hamacher, A.1; Spanier, L.1; Ciglia, E.1; Hansen, F. K.1; Gohlke, H.1; Kurz, T.1; Kassack, M. U.1 Institute of Pharmaceutical and Medicinal Chemistry, Heinrich-Heine-University Düsseldorf, Germany 1 Heat shock protein 90 (HSP90) is an abundantly found molecular chaperone of 90 kDa, induced by stress conditions such as infection, inflammation, starvation, hypoxia and cancer. Folding and maturation of client proteins such as steroid hormone receptors, protein kinases, and transcription factors are the main functions of HSP90. To fulfill that task, HSP90 forms a homo-dimer and assembles with several co-chaperones to form the so called HSP90 machinery. The C-terminus contains the dimerization domain whereas the N-terminus harbors the classical ATP binding domain. Given that HSP90 has over 280 identified client proteins, many of which being present in pathways leading to cancer, HSP90 inhibitors are promising anti-cancer drugs. Geldanamycin and radicicol were the first HSP90 inhibitors both binding to the N-terminal ATP pocket. Both compounds show antiproliferative and antitumor effects. NVPAUY922 is another small molecule inhibitor of the N-terminal ATPbinding site of HSP90 and has entered phase I clinical trials. Novobiocin is an HSP90 inhibitor binding to the C-terminal domain of HSP90 and thus inhibiting the dimerization of the monomers. This is a different mode of action leading to inhibition of dimerization and disrupted interactions of HSP70 with HSP90 [1]. The aim of this study was to test a recently designed inhibitor (compound A) of the C-terminal domain for its inhibitory effect on HSP90 function and anticancer effects with a focus on reverting chemoresistance. Compound A was able to increase chemosensitivity of the ovarian cancer cell line A2780 towards cisplatin. This effect was also observed at the resistant cell line A2780CisR but to a smaller extent. Furthermore, compound A inhibited HSP90-dependent refolding of luciferase. Cytotoxicity measured by MTT assay was similar in the sensitive A2780 (IC50 of compound A: 5.84 µM) and its resistant subclone A2780CisR (IC50 of compound A: 6.54 µM). In addition, effects of compound A on apoptosis and cell cycle were investigated. To prove activity of compound A via HSP90 inhibition, the occurrence of oligomers of HSP90 was investigated via EGS-mediated crosslinking and Western blot analysis. Compound A reduced formation of oligomers compared to blank and a negative reference control. In conclusion, compound A is a novel inhibitor of HSP90, increases chemosensitivity towards cisplatin similar to NVP-AUY922, and may thus constitute a new therapeutic option against chemoresistant cancers. Recently, in the parasite Schistosma mansoni we have identified a class I zinc-dependent HDAC subtype, HDAC8 (smHDAC8), as a standalone target to control infection [5]. These small flat worm parasites are one of the causative agents of a neglected human parasitic disease called bilharzia or schistosomiasis; infect around 200 million people worldwide and cause at least 300 000 deaths yearly, with about 800 million people further at risk of infection [6,7]. Increased risk of resistance and reduced efficacy of the only drug of treatment, praziquantel [8], has evoked the search for potential novel drug candidates against schistosomes for which vaccination is still not available. Here, we discuss several assays employed as a screening tool to identify novel inhibitors of smHDAC8 and to determine their selectivity against human HDACs. These assays are non-isotopic, include the use of fluorescence-labeled substrates enabling convenient and reproducible measurement of enzyme activity [9]. Our in-house synthetic small molecule substrates ZMTFAL (Z-Lys(F 3 Ac)- AMC) [10] and ZMAL (ZLys(Ac)-AMC) [11] are easy to synthesize, cheap and can be used in a trypsin-based set-up. In addition to ZMTFAL, a commercial substrate based on the p53 tetra-peptide {RHK(Ac)K(Ac)} has also been used to determine inhibition of schistosomal or human HDAC8 [12]. We have shown that in many cases IC50 values are very similar but may differ with different substrates up to 20 fold in potency for certain inhibitors [13]. For selectivity testing with human counterparts HDAC1 and 6, ZMAL is our substrate of choice [11]. Using a combination of these assays we have identified novel inhibitors of smHDAC8 with in vivo efficacy [5,14]. Currently, such assay-based approaches to screen Trypanosoma cruzi HDAC inhibitors are also in progress in our lab. Acknowledgments: The authors have been supported by funding from the European Union's Seventh Framework Programme for research, technological development and demonstration under grant agreements nos. 241865 (SEtTReND) and 602080 (A-ParaDDisE). References: 1. Seto, E.; Yoshida, M.: Cold Spring. Harb. Perspect. Biol. 2014, 6(4): a018713. 2. de Ruijter, A. J. et al.: Biochem. J. 2003, 370 (Pt 3): 737-749. 3. Haberland, M. et al.: Nat. Rev. Genet. 2009, 10(1): 32-42. 4. Chakrabarti, A. et al.: Trends. Pharmacol. Sci. 2015, doi: 10.1016/j.tips.2015.04.013. 5. Marek, M. et al.: PLoS. Pathog. 2013, 9(9): e1003645. 6. Gray, D.J. et al.: BMJ. 2011, 342: d2651. 7. Dömling, A.; Khoury, K.: ChemMedChem. 2010, 5(9): 1420−1434. 8. Doenhoff, M.J.; Cioli, D.; Utzinger, J.: Curr. Opin. Infect. Dis. 2008, 21(6): 659−667. 9. Hauser, A. T.; Gajer Née Wagner, J. M. , Jung, M.: Methods. Mol. Biol. 2013, 981: 211-227. 10. Heltweg, B. et al.: J. Med. Chem. 2004, 47(21): 5235–5243. 11. Heltweg, B. et al.: Anal. Biochem. 2003, 319(1): 42-48. 12. http://www.enzolifesciences.com/BML-KI178/fluor-de-lys-hdac8-deacetylase-substrate/ 13. Kannan, S. et al.: J. Chem. Inf. Model. 2014, 54(10): 3005-3019. 14. Stolfa, D.A. et al.: J. Mol. Biol. 2014, 426(20): 3442-3453. References: 1. Eckl, J. M.; Richter, K.: Int. J. Biochem. Mol. Biol. 2013, 4(4):157-165. POS.098 POS.097 In vitro screening assays for discovery of novel inhibitors of parasitic histone deacetylases Chakrabarti, A.1; The A-ParaDDisE consortium; Sippl, W.2; Jung, M.1 1 Institute of Pharmaceutical Sciences, University of Freiburg, 79104 Freiburg, Germany of Pharmaceutical Chemistry, University Halle-Wittenberg, 06120 Halle/Saale, 2 Department Germany Histone deacetylases (HDACs), a widely studied group of epigenetic enzymes, cleave acetylated ε-amino groups of lysines in histones (and other proteins) resulting in global chromatin condensation and repression of transcription [1]. Human HDACs comprise of four different classes (I– IV); class I, II, and IV, are zinc-dependent amidohydrolases or “classical” HDACs, whereas class III, called sirtuins requires NAD+ [2]. Aberrant expression of HDACs is associated with development of several diseases such as cancer, making these enzymes interesting therapeutic targets in humans [3] but also for parasitic diseases [4]. Reversing cisplatin resistance in ovarian carcinoma cells by inhibition of protein disulfide isomerase 1 Kalayda, G. V.1; Kullmann, M.1; Hellwig, M.1; Kotz, S.2; Hilger, R. A.3; Metzger, S.2,4; Jaehde, U.1 1 Institute of Pharmacy, Clinical Pharmacy, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany 2 Cologne Biocenter, University of Cologne, Zülpicher Str. 47b, 50674 Cologne, Germany 3 Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Hufelandstraße 55, 45147 Essen, Germany 4 IUF-Leibniz Research Institute for Environmental Medicine, Auf’m Hennekamp 50, 40225 Düsseldorf, Germany For abstract see Short Poster Lecture SPL.004 on page 101. DPhG Annual Meeting 2015 Conference Book • 151 POSTERS POS.099 Simultaneous inhibition of EGFR and PI3K/AKT/mTOR can overcome cisplatin resistance in a triple-negative breast cancer cell line with activated growth factor receptors. Gohr, K.1; Hamacher, A.1; Engelke, L. H.1; Kassack, M. U.1 1 Institut für Pharmazeutische und Medizinische Chemie, Heinrich-Heine-Universität, Universitätsstr. 1, 40225 Düsseldorf, Germany Intrinsic or acquired resistance against chemotherapy is a common problem in tumor treatment often leading to therapeutic failure. Cisplatin, a commonly used cytostatic agent, is under investigation for the treatment of triple negative breast cancer. Previously, in the breast cancer cell line MCF-7 activation of EGFR has been demonstrated to play a crucial role in the development of resistance [1]. In this study, we wanted to investigate the underlying mechanisms of cisplatin resistance in the triple-negative breast cancer cell line HCC38 CisR which was generated by weekly exposure to cisplatin from the sensitive subclone HCC38 over a period of 37 weeks. The examination of both cell lines via phospho-receptor tyrosine kinase array and western blotting showed an increased activation of insulin-like growth factor 1 receptor (IGF-1R) and epidermal growth factor receptor (EGFR, ErbB-1) in the cisplatin resistant subclone. The doubling time was lower in HCC38 CisR than in HCC38 underlining the activation of growth factor receptors. The highly activated IGF1-R in HCC38 CisR is also reflected in the sensitivity of HCC38 CisR for IGF1-R inhibition. The IC50 of NVP-AEW-541 determined in MTT assay after 120h of treatment was 4-fold lower in HCC38 CisR than in HCC38. We showed that simultaneous treatment with NVP-AEW541 and Lapatinib heavily reduces growth in HCC38 CisR whereas this effect was not present when inhibiting only one of the receptors. Dual inhibition could also induce apoptosis in HCC38 CisR. Although this treatment reduced cell viability it had no influence on cisplatin cytotoxicity in HCC38 CisR. Therefore we combined the receptor inhibitors with NVP-BEZ235 as an inhibitor of the downstream acting kinases PI3K and mTOR. The combination of Lapatinib and NVPBEZ235 led to a complete resensitization in the cisplatin resistant cell line. We showed that the effect of dual inhibition of IGF1-R and EGFR is superior to the use of single inhibitors in HCC38 CisR containing activated growth factor receptors. Furthermore combining Lapatinib with an inhibitor of downstream acting kinases can overcome cisplatin resistance in this cell line. In conclusion our studies showed that combining targeted therapies may be a useful tool in cancer therapy. References: 1. Eckstein, N. et al.: J. Bio.Chem. 2008, 283(2): 739-750. site, termed PIF-pocket, for mediating the regulation on the enzymatic activity. Along the process of regulation, N- or C-terminal regions are modified by different signaling and these act directly on the residues forming the PIF-pocket which in turn affect the intrinsic activities of the kinases, allosterically. Current state of the art in the drug development to protein kinases involves the targeting of the ATP-binding site (type I inhibitors), and sites directly extending from the ATP-binding site (type II inhibitors). However, the development of drugs targeting sites which are distant from the ATP-binding site (type III inhibitors) are still unconventional. We previously described biochemically and structurally how small compounds binding to the PIF-pocket could allosterically inhibit the activity of aPKCs. Here we describe the development and characterization of a follow-up series represented by PS432, an allosteric inhibitor that specifically targets the PIF-pocket of aPKCs, inhibits proliferation of lung cancer cells in culture and tumour progression of lung cancer in tumour xenograft model. POS.101 Target-sensitive, vascular directed liposomes with entrapped chemokine receptor antagonists for a local interference with tumor cell metastasis Schlesinger, M. 1; Roblek, M. 2; Calin, M. 3; Stan, D. 3; Zeisig, R. 4; Simionescu, M. 3; Bendas, G. 1; Borsig, L. 2 1 Department of Pharmacy, Rheinische Friedrich-Wilhelms-University Bonn, 53121 Bonn, Germany 2 Institute of Physiology, University of Zürich and Zürich Center for Integrative Human Physiology, CH-8057 Zurich, Switzerland 3 Institute of Cellular Biology and Pathology ‘‘N. Simionescu’’ of the Romanian Academy, Bucharest, Romania 4 Experimental Pharmacology & Oncology Berlin Buch GmbH, 13125 Berlin, Germany For abstract see Short Poster Lecture SPL.005 on page 102. POS.100 Allosteric inhibition of atypical PKC for the treatment of cancer D.1; Odadzic, Arencibia, J. Fröhner, W. ; Schulze, Neimanis, S.2; Proschak, E.4; Engel, M.3; Zeuzem, S.2; Stark, H.5; Biondi, R. M.2 M.2; 3 J. 2; 1 German Cancer Research Center(DKFZ), Im Neuenheimer Feld 280, 69121 Heidelberg, Germany 2 Research Group PhosphoSites, Medizinische Klinik 1, Universitätsklinikum Frankfurt, TheodorStern-Kai 7, 60590 Frankfurt, Germany. 3 Pharmaceutical and Medicinal Chemistry, University of Saarland, 66041 Saarbrücken, Germany. 4 Institute for Pharmaceutical Chemistry, J.W. Goethe University,60438 Frankfurt, Germany. 5 Pharmaceutical and Medicinal Chemistry, Heinrich-Heine-University, 40225 Düsseldorf, Germany Protein kinases play key regulatory roles in cells and organisms by responding to specific stimuli and selectively phosphorylating protein substrates. To achieve their specific functions, protein kinases have evolved very sophisticated mechanisms of regulation. Protein kinases possess a conserved catalytic domain, consisting of two lobes, a small lobe and a large lobe, with the ATP-binding site located in the cleft between the lobes. To achieve the required exquisite selective regulation, protein kinases frequently possess additional N- and Cterminal domains. The AGC group of protein kinases, named after its representatives Protein kinase A, Protein kinase G and Protein kinase C, are regulated by very different stimuli but often converge in a conserved 152 • DPhG Annual Meeting 2015 Conference Book POS.102 The class IIa preferential HDAC inhibitor LMK235 enhances the efficacy of cisplatin in HNSCC cell lines. Hamacher, A.1; Schulte, M.1; Hansen, F. K.1, Kurz, T.1, Kassack, M. U.1 Institut für Pharmazeutische und Medizinische Chemie, Heinrich Heine Universität Düsseldorf, Universitätsstr. 1, 40225 Düsseldorf, Germany 1 Cisplatin-based chemotherapy is part of the standard treatment of head and neck squamous cell carcinoma (HNSCC). However, many patients develop cisplatin resistance during treatment. The mechanism of cisplatin resistance is multifactorial and includes genetic and epigenetic alterations such as DNA methylation and histone acetylation. Furthermore, aberrant HDAC expression was found in many tumors to be associated with poor response to chemotherapy. Epigenetic therapies including pan-HDAC inhibitors (HDACi) such as vorinostat have shown suppressive effects against various tumor cells by inhibiting cell proliferation and inducing cell cycle arrest as well as apoptosis. Up to now, it is unknown which HDAC subtype may be the most promising for anticancer effects. Further, pan-HDACi show numerous side effects. Subtype selective HDACi may thus be a promising class of epigenetic anticancer drugs. CANCER/EPIGENETICS A set of 30 hydroxamate-based HDACi was previously evaluated at the HNSCC cell lines Cal27 and Kyse510 and their cisplatin resistant sublines. LMK235 was identified as the most potent compound concerning HDAC inhibition and cytotoxicity [1]. In this study, LMK235 was selected for further characterization of its anticancer effects and reversal of cisplatin chemoresistance. LMK235 showed a concentration-dependent inhibition of cellular proliferation in all 4 HNSCC cell lines. HDAC isoform profiling of LMK235 revealed a preference for HDAC4 and HDAC5 with IC50 values of 11.9 nM and 4.22 nM in contrast to vorinostat. Incubation with LMK235 48 h prior to cisplatin resulted in a 46-fold (Cal27) and 60-fold (Kyse510) enhancement of cisplatin-induced cytotoxicity. Furthermore, complete reversal of cisplatin resistance was observed for both cisplatin resistant sublines Cal27CisR and Kyse510CisR. In addition, preincubation with LMK235 increased the sensitivity of the HNSCC cell lines to cisplatinmediated apoptosis. In summary, our study has identified a potent HDACi with a new class IIa selectivity profile. LMK235 is a promising tool to investigate the function of HDAC4 and HDAC5 concerning cancer development and to improve the treatment of HNSCC. References: 1. Marek, L. et al.: J. Med. Chem. 2013, 56(2): 427-436. POS.103 Interference with chemokines as potential antitumor strategy: Sulfated glycans inhibit the CXCL12-induced activation of both CXCR4 and CXCR7. Alban, S.1; Ehrig, K.; Liewert, I.; Schneider, T.1 1 Pharmaceutical Germany Institute, Christian-Albrechts-University, Gutenbergstr. 76, 24146 Kiel, Introduction: Chemokines mediate leukocyte trafficking during homeostasis and inflammation and are essential for linking innate and adaptive immunity. The chemokine CXCL12 is the main factor to home hematopoietic progenitor cells to the bone marrow but it also plays an important role in cancer cell proliferation, invasion and metastasis. Therefore, blocking of this signaling axis is a promising strategy for tumor therapy [1-3]. Heparins are known to interact with CXCL12 and various other chemokines and they also exert antimetastatic effects in vivo. Objectives: The aim of this study was to elucidate whether algae-derived glycans interact with CXCL12. Furthermore, the potencies of algaederived glycans and heparins concerning the inhibition of CXCL12induced activation of CXCR4 and CXCR7 should be evaluated. Materials and Methods: Sulfated glycans of the brown algae Saccharina latissima and Fucus vesiculosus as well as unfractionated heparin and tinzaparin were selected as test compounds. The approved CXCR4anatgonist plerixafor (AMD3100, Mozobil™) served as reference compound. CXCR4 and CXCR7 activation in a human Burkitt’s lymphoma cell line was monitored by flow cytometry via receptor internalization upon ligand binding. The binding of sulfated glycans to CXCL12 was determined with a competitive sulfated polysaccharide coating-ELISA. As exemplary downstream effects cell migration was quantified and secretion of matrix metalloproteinases was investigated by zymography. Results: In contrast to plerixafor, algae-derived glycans bound CXCL12 and thereby blocked the CXCL12-induced activation of CXCR4 as well as CXCR7. Furthermore, treatment with these glycans or heparins impaired downstream effects in human Burkitt’s lymphoma cells like migration and secretion of matrix metalloproteinase 9. Concerning the inhibition of CXCL12-signaling algae-derived glycans proved to be more active than heparins. Their strong inhibition of CXCL12-signaling and low anticoagulant properties, which reduce the risk of bleeding, make them promising candidates for tumor therapy. Conclusion: Sulfated glycans bind CXCL12 and thereby inhibit the activation of both CXCR4 and CXCR7 in human Burkitt’s lymphoma cells. The inhibitory potency of algae-derived glycans on this axis is significantly superior to that of heparins. Acknowledgments: This work was supported by the Federal Ministry of Research and Education (BMBF) within the national research project “Algae Against Cancer” (0315812F). References: 1. Cojoc M. et al.: Onco. Targets Ther. 2013 6: 1347–1361. 2. Domanska, U. M. et al.: Eur. J. Cancer. 2013 49(1): 219-230. 3. Chatterjee, S.; Behnam Azad, B.; Nimmagadda, S.: Adv. Cancer Res. 2014 124: 31–82. POS.104 Development of 3-amido-benzhydroxamic acids as small molecule inhibitors against smHDAC8 for the treatment of schistosomiasis Heimburg, T.1; Melesina, J.1; Chakrabarti, I.2; Walter, A.2; Hauser, A.-T.2; Schmidtkunz, K.2; Marek, M.3; Lancelot, J.4; Romier, C.3; Pierce, R.4; Schmidt, M.1; Jung, M.2; Sippl, W.1 Institut für Pharmazie, Martin-Luther-Universität Halle-Wittenberg, Wolfgang-Langenbeck – Straße 4, 06120 Halle (Saale), Germany 2 Institut für Pharmazeutische Wissenschaften, Albert-Ludwigs-Universität Freiburg, Albertstr. 25, 79104 Freiburg, Germany 3 IGBMC, Universite de Strasbourg, 1 rue Laurent Fries, 67404 Illkirch Cedex, France 4 Center for Infection and Immunity of Lille (CIIL), Université Lille Nord de France, Institut Pasteur de Lille, 1 rue Professeur Calmette, 59019Lille Cedex, France 1 Schistosomiasis is one of the major human neglected parasitic diseases [1], which is caused by small flat worm parasites from the genus Schistosoma. At the moment, there are no licensed vaccines available against Schistosomiasis and for Praziquantel, the one drug which is effective against all schistosome species, reduced efficiency and drug resistance are reported [2]. Currently inhibitors of human epigenetic enzymes are investigated as novel anti-cancer drugs and have the potential to be used as new anti-parasitic agents [3]. Here, we report that Schistosoma mansoni histone deacetylase 8 (smHDAC8), the most expressed class I HDAC isotype in this organism, is a functional acetylL-lysine deacetylase that plays an important role in parasite infectivity. A combination of virtual screening and biological testing resulted in linkerless aromatic-hydroxamic acidsas novel smHDAC8 inhibitors which induce apoptosis and mortality in schistosomes. Crystal structures of smHDAC8 with the linkerless inhibitors could be solved which showed the accuracy of the applied modeling approach [4,5]. Consequently several 3-amido-benzhydroxamic acids were synthesized and tested against smHDAC8 and optimized in potency and selectivity. Acknowledgments: This work and the authors received funding from the European Union's Seventh Framework Programme for research, technological development and demonstration under grant agreements nos. 241865 (SEtTReND) and 602080 (A-ParaDDisE). References: 1. Hotez, P. J.; Pecoul, B.: PLoSNegl Trop. 2010, Dis 4: e718. 2. Doenhoff, M. J.; Cioli, D.; Utzinger, J.: CurrOpin Infect. 2008, Dis 21: 659-667. 3. Andrews, K. T.; Haque, A.; Jones, M. K.: Immunol Cell Biol. 2012, 90: 66-77. 4. Marek, M. et al.: PLOS Pathogens. 2013. 5. Kannan, S.; Melesina, J.; Hauser, A. et al.: J. Chem. Inf. and Mod. 2014. POS.105 Synthesis and in vitro characterization of hydroxamic acids as small molecule inhibitors for protozoal targets Bayer, T.1; Melesina, J.1; Chakrabarti, A.2; Walter, A.2; Marek, M.3; Romier, C.3; Schmidt, M.1; Jung, M.2; Sippl, W.1 1 Institut für Pharmazie, Martin-Luther-Universität Halle-Wittenberg, Wolfgang-Langenbeck – Straße 4, 06120 Halle (Saale), Germany 2 Institut für Pharmazeutische Wissenschaften, Albert-Ludwigs-Universität Freiburg, Albertstr. 25, 79104 Freiburg, Germany 3 IGBMC, Université de Strasbourg, 1 rue Laurent Fries, 67404 Illkirch Cedex, France Schistosomiasis, also known as bilharzia is caused by a blood-dwelling fluke of the genus Schistosoma. It uses a fresh water snail as an intermediate host and is transmitted through contaminated water. Within the human host the parasite passes through several life-cycle stages and causes various severe symptoms [1]. Taking into account that an estimate of 200 million people are infected worldwide among who 280,000 die annually and another 800 million are at risk of infection schistosomiasis is one of the most important parasitic diseases [2]. Praziquantel is an anthelminthic which is effective against all human forms of schistosomiasis. Without any effective vaccine available and the excessive use of praziquantel for the treatment of infected individuals as well as for preventive treatment the problem of resistant schistosome strains is arising [1]. DPhG Annual Meeting 2015 Conference Book • 153 POSTERS Histone deacetylases (HDACs) take an important part in epigenetics since the state of acetylation of the histones correlates with transcriptional control [3]. The schistosome histone deacetylase 8 (smHDAC8) was recently identified as a potential target for antiparasitic therapy [4]. The screening hit J1075 (a 3-chloro-benzothiophen derivative) has proven effective in terms of smHDAC8 inhibition, mortality and unpairing of adult schistosomes (and therefore the disability of reproduction) and consequently is an interesting lead structure for further derivatisation and optimization [5]. Here we present different classes of compounds derived from the starting point J1075. nanomolar range and a comprehensive DSF screening showing remarkable selectivity over other bromodomains. References: 1. Filippakopoulos, P. et al.: Nat. Rev. Drug Discov. 2014, 13: 337-356. 2. Filippakopoulos, P. et al.: Nature 2010, 468: 1067-1073. 3. Rooney, T. P. C. et al.: Angew. Chem. 2014, 126: 1–6. 4. http://www.thesgc.org/chemical-probes/ICBP112 5. Hay, D. A. et al.: J. Am. Chem. Soc. 2014, 136: 9308-9319. Acknowledgments: SEtTReND, A-ParaDDisE POS.108 References: 1. Deribew, K. et al.: Int. J. Med. Med. Sci. 2013, 5(3): 131-139. 2. Steinmann, P. et al.: Lancet Infect. Dis. 2006, 6(7): 411-425. 3. KrennHrubec, K. et al.: Bioorg Med Chem Lett. 2007, 17(10): 2874 – 2878. 4. Kannan, S. et al.: J. Chem. Inf. Model. 2014, 54(10): 3005–3019. 5. Marek, M. et al.: PLoS pathogens, 2013, 9(9): e1003645. α-Aminoxy oligopeptides: Solid-phase synthesis, conformational investigation and anticancer activity Diedrich, D.1; Rodrigues Moita, A. J.1; Rüther, A.2; Kurz, T.1; Lüdeke, S.2; Kassack, M. U.1; Hansen, F. K.1 1 Institute POS.106 Rational design and diversity-oriented synthesis of peptoid-based selective HDAC6 inhibitors with potent anticancer activity Diedrich, D.1; Syntschewsk, V.1; Hamacher, A.1; Alves Avelar, L. A.1; Gertzen, C. G .W.1; Reiss, G. J.2; Kurz, T.1; Gohlke, H.1; Kassack, M. U.1; Hansen, F. K.1 Institut für Pharmazeutische und Medizinische Chemie, Heinrich Heine Universität Düsseldorf, Universitätsstr. 1, 40225 Düsseldorf, Germany 2 Institut für Anorganische Chemie und Strukturchemie, Heinrich Heine Universität Düsseldorf, Universitätsstr.1, 40225 Düsseldorf, Germany 1 For abstract see Short Poster Lecture SPL.006 on page 103. POS.107 Design of new, selective benzoxazepine-type inhibitors for the CBP/EP300 bromodomains Popp, A. T.1; Fedorov, O.2; Tallant, C.2; Knapp, S.3; Bracher, F.1 Department Pharmazie, Ludwig-Maximilians-Universität, Butenandtstraße 5-13, 81377 Munich, Germany Structural Genomics Consortium, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Headington, Oxford, OX3 7DQ, United Kingdom 3 Institut für Pharmazeutische Chemie, Goethe Universität, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany 1 2 Reversible acetylation of histones and other proteins is involved in the transcriptional regulation and DNA repair. Bromodomains, the “readers” of N-ε-acetylated lysine, are an interesting target for research and pharmaceutical industry [1,2]. Most of the work was focused on bromodomains of BET sub-family such as BRD4, but recently inhibitors of other proteins also emerged. Several inhibitors are now available for phylogenetically closely related BRDs CBP and EP300 [3-5]. Their utilization as research tools may further accelerate and yield better understanding and potential avenues of intervention for diseases such as Rubinstein-Taybi syndrome, leukemia, ovarian, breast and lung cancers, and systemic lupus erythematosus [1,4]. Here we report novel CBP/EP300 inhibitors based on I-CBP112 design. We synthesized and characterized in biochemical assays approximately 50 compounds with scaffold A. This effort gave further insight into the SAR of benzoxazepine type inhibitors and resulted in a CBP/EP300 inhibitor with an IC50 in the 154 • DPhG Annual Meeting 2015 Conference Book of Pharmaceutical and Medicinal Chemistry, Heinrich-Heine-Universität Düsseldorf, 40225 Düsseldorf, Germany 2 Institute of Pharmaceutical Sciences, Albert-Ludwigs-Universität Freiburg, 79104 Freiburg, Germany Cancer is a leading cause of death in economically developed countries. The global burden of cancer continues to increase significantly in particular due to the aging and growth of the world population. Chemotherapy using cytotoxic agents is one major approach to combat cancer. Unfortunately, cancer cells, which are initially suppressed by established antitumor drugs, may develop a resistance to standard drug therapy. Thus, there is a need for new anticancer agents preferably acting with unique modes of action. Over the years, anticancer peptides (ACPs) have been identified as promising class of cytotoxic agents [1]. However, the high production cost and the intrinsic instability of peptides towards proteases have resulted in extensive research in peptidomimetics and the field of foldamers has received massive interest [2]. Despite their very attractive properties such as increased proteolytic and conformational stability [3,4], α-aminoxy peptides have not received as much attention as other foldamers such as β-peptides or peptoids. One important reason for this is the difficult synthetic access to longer oligomers. We herein present an improved synthetic route to α-aminoxy oligopeptides by the straightforward use of a combination of solutionphase and solid-phase supported methods. Although the field of ACPs is considered as a rapidly emerging research area, no anticancer properties have been reported for α-aminoxy peptides thus far. In this context, we report here the remarkable cytotoxic activity of α-aminoxy oligopeptides against cancer cells. CD spectroscopy was employed to get a more profound understanding of the folding properties of α-aminoxy oligopeptides and to investigate possible bioactive conformations. Preliminary mode of action studies revealed that membranolytic and proapoptotic effects may contribute to the anticancer activity of αaminoxy oligopeptides. Acknowledgments: This work was supported by funds from the Strategischer Forschungsfonds of the Heinrich-Heine-Universität Düsseldorf and Fonds der Chemischen Industrie. References: 1. Gaspar, D.; Veiga, A. S.; Castanho M. A. R. B.: Front. Microbiol. 2013, 4: 294. 2. Martinek, T. A.; Fülöp, F.: Chem. Soc. Rev. 2012, 41(2): 687–702. 3. Li, X.; Wu, Y-D.; Yang, D.: Acc. Chem. Res. 2008, 41(10): 1428–1438. 4. Draghici, B. et al.: RSC Adv. 2011, 1(4): 602–606. CANCER/EPIGENETICS POS.109 6‐Arylamino‐3,4‐dihydroisoquinolin‐1(2H)‐ones as new pharmacophores for linear hinge binders inducing the glycine-flip A convenient synthetic methodology to access the scaffold was developed employing an intramolecular Heck reaction as the key step for building up the tricyclic core. A small library of inhibitors was prepared and characterized using in vitro biochemical as well as cellular assays. Potencies down to picomolar range could be achieved along with enhanced selectivity for JAK3 compared to Tofacitinib [4]. Praefke, B. A.1; Laufer, S. A.1 1 Faculty of Science, Pharmaceutical and Medicinal Chemistry, University of Tuebingen, Auf der Morgenstelle 8, 72076 Tuebingen, Germany Disregulation in kinase activity can be related to various diseases such as cancer, diabetes and inflammatory disorders [1]. We developed dibenzosuberones, dibenzoxepinones and benzosuberones as highly potent and selective p38α MAPK inhibitors with a carbonyl based hinge binding motif. When binding the p38α MAP kinase these scaffolds induce a “glycine-flip”, a rotation of Gly110 in the hinge region, resulting in the formation of two hydrogen bonds from the carbonyl oxygen to the amideNH of Gly110 and Met109 [2,3]. Only 46 of the known 518 protein kinases possess a glycine in the corresponding position, making the induction of a glycine-flip a promising way to increase selectivity [4]. Based on our previous scaffolds we developed a new pharmacophore, replacing the ketone by an amide group. We expected the increased electron density of the amide oxygen to strengthen the bidentate hydrogen bonds to the flipped glycine and the adjacent amino acid. The lipophilicity and electron density of the arylamino moiety was varied by different substituents (R2) to explore the hydrophobic region I of kinases bearing a glycine in the hinge region. References: 1. Cornejo, M. G.; Boggon, T. J.; Mercher, T.: Int. J. Biochem. Cell B 2009, 41(12): 2376-2379. 2. O'Shea, J. J.: Annals of the rheumatic diseases 2004, 63 Suppl 2: ii67-ii71. 3. Gehringer, M. et al.: ChemMedChem 2014, 9: 2516-2527. 4. Gehringer, M. et al.: ChemMedChem 2014, 9(2): 277–281. POS.111 New insights into the pro-apoptotic activity of P8-D6, a potent dual topoisomerase inhibitor for antineoplastic therapy Meier, C.1; Koburg, M.2; Bischoff, F.2; Vollmar, A. M.2; Clement, B.1 Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy, CAU Kiel, Gutenbergstraße 76, 24118 Kiel – Germany 2 Department of Pharmacy – Pharmaceutical Biology, LMU Munich, Butenandtstraße 5-13, 81377 Munich – Germany 1 Topoisomerase inhibitors constantly remain an indispensable instrument in antitumor therapy due to their effectiveness and a considerable clinical experience regarding therapy safety and combined administration with other cytostatics [1,2,3]. Nevertheless, resistance mechanisms like the upregulation of the expression of one topo-isomerase form by selective inhibition of the other as well as a membrane-triggered decrease of intracellular concentrations of approved topo agents are frequently described [4,5,6]. Consequently, the design of dual topoisomerase I/II inhibitors that exhibit a high cytotoxicity and a strong affinity to both enzyme classes remains a highly desirable goal in antitumor drug development to date [1,7]. We recently developed P8-D6, a small molecule with strong equipotent topoisomerase I /IIα/IIβ inhibitory activity and an excellent broad spectrum cytotoxicity in the nanomolar range. Besides its physicochemical advantages, e.g. a solubility in the millimolar region, a high tolerability towards P8-D6 was shown in first maximum tolerated dose studies (athymic nude mice) encouraging its further development in in vivo efficacy models. References: 1. Manning, G. et al.: Science. 2002, 298(5600): 1912-1934. 2. Koeberle, S. C. et al.: J. Med. Chem. 2012, 55(12): 5868-5877. 3. Baur, B. et al.: J. Med. Chem. 2013, 56(21): 8561-8578. 4. Martz, K. E. et al.: J. Med. Chem. 2012, 55(17): 7862-7874. POS.110 A Comprehensive Structure-Activity Relationship Study on Tofacitinib Bioisosteres to improve the Selectivity of JAK3 Inhibitors Pfaffenrot, E.1; Gehringer, M.1; Bauer, S. M.1; Laufer, S. A.1 Department of Pharmaceutical and Medicinal Chemistry, Pharmaceutical Institute, University of Tuebingen, Auf der Morgenstelle 8, 72076 Tuebingen, Germany 1 JAK3 is a cytosolic non-receptor tyrosine kinase belonging to the Janus kinase (JAK) family. The four family members JAK1, JAK2, JAK3 and TYK2 (Tyrosine kinase 2) are involved in various signaling cascades initiated by cytokines and growth factors mediating immune response and proliferation. JAK3 plays a key role in the development of lymphocytes such as T cells, B cells and NK cells and represents an attractive therapeutic target for the treatment of inflammatory diseases. Selective inhibition of JAK3 is suggested to provide immunosuppression with less side effects due to the exclusive expression of JAK3 in hematopoietic tissue [1,2]. In an extensive structure-activity relationship study we synthesized a library of Tofacitinib bioisosteres and tested their ability to inhibit JAK3 [3]. A novel class of tricyclic Janus kinase (JAK) inhibitors was designed by applying a rigidization approach to the FDA-approved drug Tofacitinib. Figure: P8-D6 as promising dual topoisomerase inhibitor. Herein presented studies are dealing with the anti-leukemic activity of P8D6 in Jurkat ALL cells, focussing on pro-apoptotic impact. Furthermore, we investigated the growth-inhibitory effects of P8-D6 in combination with the protein disulfide isomerase (PDI) inhibitor PS89. It was shown that this substance could sensitize various cancer cells towards Etoposide treatment [8]. The results were also compared with effects on healthy human lymphocytes to give first information about the selectivity towards tumor cells. References: 1. van Gijn, R. et al.: J. Oncol. Pharm. Pract. 2000, 6: 92–108. 2. Pommier, Y.: Nat. Rev. Cancer 2006, 6: 789–802. 3. Deweese, J. E.; Osheroff, N.: Nucl. Ac. Res. 2009, 37: 738–748. 4. Felix, C. A.; Kolaris, C. P.; Osheroff, N.: DNA repair 2006, 5: 1093–1108. 5. Lewis, L. J. et al.: Anti-cancer drugs 2007, 18: 139–148. 6. Whitacre, C. M. et al.: Cancer Res. 1997, 57: 1425–1428. 7. Salerno, S. et al.: Curr. Med. Chem. 2010, 17: 4270–4290. 8. Eirich, J.; Braig, S. et al.: Angewandte Chemie Int. Ed. 2014, 53: 12960-12965. DPhG Annual Meeting 2015 Conference Book • 155 POSTERS POS.112 Tetra-substituted pyridinylimidazoles as dual inhibitors of p38α mitogen-activated protein kinase and c-Jun Nterminal kinase 3 for potential treatment of neurodegenerative diseases Muth, F.1 ; Günther, M.1; Bauer, S. M.1; Döring, E.1; Fischer, S.1; Maier, J.2; Drückes, P.3 ; Köppler, J.3; Trappe, J.3; Rothbauer, U.2; Koch, P.1; Laufer, S. A.1 1 Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmaceutical Sciences, Eberhard-Karls-University Tuebingen, Auf der Morgenstelle 8, 72076 Tuebingen, Germany. 2 Natural and Medical Sciences Institute at the University of Tuebingen, Markwiesenstrasse 55, 72770 Reutlingen, Germany. 3 Novartis Pharma AG, CH-4002 Basel, Switzerland. Multi-target drugs inhibiting several key drivers involved in neurodegeneration may exhibit additive or even synergistic effects in vivo. For the treatment of complex diseases like neurodegeneration, therapies that act at multiple targets and providing symptomatic as well as neuroprotective effects may thus be more effective. Recently, Taylor et al. reported enhanced levels of MKP-1 are neuroprotective via dephosphorylation and therefore deactivation of JNK3 and p38α MAP kinase [1]. Tetra-substituted imidazoles were designed as dual inhibitors of c-Jun Nterminal kinase (JNK) 3 and p38α mitogen-activated protein (MAP) kinase. A library of 45 derivatives was prepared and evaluated in a kinase activity assay for their ability to inhibit both kinases, JNK3 and p38α MAP kinase. Dual inhibitors with IC50 values down to the low double-digit nanomolar range at both enzymes were identified. Two inhibitors of series 6 (6l and 6o) show an excellent selectivity profile. The best balanced dual JNK3/p38α MAP kinase inhibitors are 6m (IC50 JNK3: 18 nM, p38α: 30 nM) and 14d (IC50 JNK3: 26 nM, p38α: 34 nM) featuring both, excellent solubility and metabolic stability. They may serve as useful tool compounds for preclinical proof-of-principle studies in order to validate the synergistic role of both kinases in the progression of Huntington’s disease. Acknowledgments: We thank Peter Keck for the help with the processing of the raw data. We are grateful to Matthias Gehringer for contributing Figure 3. Daniela Müller and Katharina Bauer are also acknowledged for skillful technical assistance in compound testing. References: 1. D. M. Taylor, et al.: J. Neurosci. 2013, 33: 2313-2325. POS.113 Multicomponent synthesis and anticancer activity of a novel class of peptoid-based HDAC inhibitors Syntschewsk, V.1; Hamacher, A.1; Kurz, T.1; Kassack, M. U.1; Hansen, F. K.1 Institut für Pharmazeutische und Medizinische Chemie, Heinrich Heine Universität Düsseldorf, Universitätsstr. 1, 40225 Düsseldorf, Germany 1 Proteins that are able to regulate epigenetic processes became interesting targets for the development of drugs. In this context, histone deacetylases (HDACs) play an important role, due to their ability to remove acetyl groups from N-acetyl-lysine residues of histones and nonhistone proteins. These posttranslational modifications of chromatin, caused by HDACs, have an influence on the gene expression [1-4]. Thus far, 18 HDAC isoforms have been identified and divided into different classes based on their homology to their corresponding yeast orthologues. HDAC classes I (HDACs 1-3,8), IIa (HDACs 4,5,7,9), IIb (HDACs 6,10) and IV (HDAC 11) contain zinc-dependent deacetylase domains, whereas class III are NAD+ dependent [2]. There are several pathways which are addressed by HDAC inhibitors (HDACi) to inhibit 156 • DPhG Annual Meeting 2015 Conference Book tumor cell growth such as apoptosis, differentiation and cell cycle arrest [2]. HDACi therefore emerged as interesting targets for the development of anticancer drugs. Currently, four HDACi have been approved by the FDA for the treatment of cutaneous/peripheral T-cell lymphoma or multiple myeloma (vorinostat, romidepsin, belinostat and panobinostat). However, there is still an urgent need for the development of novel HDACi with finetuned pharmacokinetic and -dynamic properties. In this poster, we report on the multicomponent synthesis and biological evaluation of a new class of potent HDACi bearing peptoid-based cap groups. An Ugi four-component reaction (U-4CR) was utilized for the straightforward and systematic variation of the cap moiety. A series of post-Ugi transformations allowed the introduction of different zincbindung groups (ZBGs). All novel peptoid-based HDACi were screened for their biological activity in cellular HDAC and MTT assays using different sensitive and chemoresistant cancer cell lines. Notably, the most potent HDACi exceeded the activity of the reference compounds vorinostat and cisplatin, respectively. Selected HDACi were further tested for their enhancement of cisplatin-induced cytotoxicity. Based on these preliminary data, this series of compounds represents a valuable starting point for further elaboration and the results reported here will help to guide future efforts toward developing novel HDACi with optimized anticancer properties. References: 1. Cai, J. et al.: Bioorg. Med. Chem. 2015, 23(13): 3457-3471. 2. Witt, O. et al.: Cancer Lett. 2009, 277(1): 8–21. 3. Minucci, S.; Pelicci, P. G.: Nat. Rev. Cancer 2006, 6(1): 38-51. 4. Bertrand, P.: Eur. J. Med. Chem. 2010, 45(6): 2095-2116. POS.114 Pt(II) complexes – potential inhibitors of seleniumdependent redox enzymes Lemmerhirt, H.1; Behnisch, S.1; Lillig, C. H.2; Bednarski, P. J.1 1 Ernst-Moritz-Arndt-University of Greifswald, Institute of Pharmacy, Friedrich-Ludwig-JahnStraße 17, DE 17489 Greifswald, Germany 2 Ernst-Moritz-Arndt-University of Greifswald, University Medicine, Institute for Medical Biochemistry and Molecular Biology, DZ7, Ferdinand-Sauerbruchstraße, DE 17475 Greifswald, Germany Platinum complexes (e.g. cisplatin, carboplatin, oxaliplatin) are established drugs in cancer therapy. They induce DNA damage, cell cycle arrest and apoptosis in proliferating cancer cells. Since the discovery of cisplatin, a wide range of platinum complexes has been developed with novel mechanisms of action. Hydrogen peroxide is known as a secondary messenger for apoptosis. Glutathione peroxidase (GPx), an enzyme that detoxifies H2O2, is believed to be involved in the development of resistance of cancer cells treated with anti-cancer drugs [1,2]. Due to the overexpression of this enzyme, it is assumed that it acts to increase the degradation of H2O2. Another selenocysteine-dependent redox enzyme is thioredoxin reductase (TrxR), which is targeted by cisplatin and is also a promising target enzyme for anticancer therapy. We have synthesized a library of Pt(II) complexes derived from cisplatin and the inactive isomer transplatin, coordinated to hydrazone ligands known to weakly inhibit GPx (Figure) [1]. It was envisioned that the compounds would interact irreversibly by way of their Pt(II) atom with the active site selenium atom of the enzymes while the organic ligand would increase both selectivity and binding affinity to the enzyme active site. GPx and TrxR inhibitors might find use in cancer treatment to re-sensitize resistant cancer cells to anti-cancer therapy. Furthermore, selectively inhibiting GPx or TrxR could be useful to investigate and to better understand their distinct roles in cell physiology. B A OH O N H3 C N N H N NH3 Cl N+ Pt NH3 H3C OH O N N H N Figure A) hydrazone inhibitor of GPx; B) corresponding trans-Pt(II) complex Here, we present the results of experiments designed to measure the inhibition of GPx and TrxR by these cis- and trans-Pt(II) complexes, their effects on cellular levels of reactive oxygen species (ROS) and their ability to initiate apoptosis. Cell cytotoxicity and interactions of the novel platinum compounds with DNA will also be presented. References: 1. Schulz, R. et al.: Bioorg. Med. Chem. Lett. 2012, 22(21): 6712–6715. 2. Wilde, F. et al.: Mo.l Divers. 2014, 18(2): 307–322. CANCER/EPIGENETICS POS.115 Gold(I) Complexes with N-Heterocyclic Carbene, Phosphane and Alkyne Ligands show Promising Anticancer Properties – An Overview Prochnicka, A.1,2; Andermark, V.1,2; Kankowski, S.1; Meyer, A.1; Rubbiani, R.1; Göke, K.2,3; Bunjes, H.2,3; Ott, I.1,2 1 Institut für Medizinische und Pharmazeutische Chemie, Technische Universität Braunschweig, Beethovenstraße 55, D-38106, Braunschweig, Germany, 2 Zentrum für Pharmaverfahrenstechnik (PVZ), Franz-Liszt-Straße 35A,Technische Universität Braunschweig, D-38106 Braunschweig, Germany 3 Institut für Pharmazeutische Technologie, Technische Universität Braunschweig, Mendelssohnstraße 1, D-38106 Braunschweig, Germany Organometallic gold(I) complexes are an interesting group of drugs, which show activity against tumor relevant enzymes and have good antiproliferative activities [1,2]. They are part of the attempt to develop new anticancer agents, which show fewer side effects, good tumor cell selectivity and no drug resistance. Especially N-heterocyclic carbenes (NHC), phosphanes and alkynes as ligands of gold(I) offer a huge potential as anticancer agents [3,4]. This work gives an overview of our different projects that engage with these complexes (see Figure 1) with a closer look at complexes with alkynyl ligands. We measured good antiproliferative effects in vitro for all combinations of gold(I) with NHCs, phosphanes and alkynes. Cellular uptake studies indicated that such gold(I) complexes can accumulate in cancer cells. Many of these gold(I) complexes also showed selective activity against thioredoxin reductase (TrxR), which is one of the overexpressed enzymes in cancer cells. A decrease in respiration suggested mitochondria as further possible targets for NHC-gold(I)-phosphanes. The group of alkynyl-gold(I)-phosphanes triggered anti-angiogenic effects, as shown in former work with zebrafish embryos [5]. Other targets like the G-quadruplex DNA, which is involved in oncogenetic regulation, and the DNA repairing enzyme poly(ADP-ribose)polymerase (PARP-1) are under discussion for gold(I) complexes (see Figure 2) [6,7]. Taking together previous and new data, this work makes a good step towards an improved overview of possible targets, biodistribution and structure-activity-relationships for the different gold(I) complexes. New fluorinated NHC gold(I) complexes (Figure) of the phenylimidazole type were synthesized, purified and characterized via 1H-/13C-/19F-NMR, MS-ESI/MS-EI and elemental analysis. They were screened against various tumorigenic cell lines as well as a non-tumorigenic human kidney cell line. Their antiproliferative potential varies depending on the lipophilic moieties. Thioredoxin reductase (TrxR) is a well-known target of gold complexes due to their high affinity to selenocysteine in the flexible N-terminus of the enzyme, which is easily accessible. TrxR is upregulated in several carcinoma cells, is involved in cell protection processes and prevents cells from oxidative stress and apoptosis [2,3]. Tests with the isolated enzyme confirm the inhibition by the NHC gold(I) complexes. Beside the enzyme inhibition, the suppression of mitochondrial respiratory activity is also discussed as a possible mode of action [4]. These results were investigated in more detail through cellular uptake studies in MCF-7 breast carcinoma cells using high resolution continuum source atomic absorption spectroscopy (HR CS-AAS) technique for the quantification of the intracellular amount of the metal. Higher accumulations in the cells are accompanied by lower IC50 values in the proliferation inhibition assay. The affinity to transport proteins, such as albumin, transferrin or fetuin can have a huge influence on the biodistribution of the complexes [5]. Protein binding studies with isolated serum albumin as well as fetal calf serum (FCS) show an interesting correlation between the accumulation rate of intracellular gold and the protein binding behaviour. Further studies include in-vitro permeability tests with a Caco-2 cell layer model to demonstrate the tracking of apical and basal transport through intestinal epithelial cells to investigate possible routes of application. References: 1. Oehninger, L.; Rubbiani, R.; Ott, I.: Dalton Trans. 2013, 42: 3269–3284. 2. Fritz-Wolf, K. et al.: Nat. Commun. 2011, 2(383): 1–8. 3. Pratesi, A. et al.: J. Inorg. Biochem. 2014, 136: 161–169. 4. Holenya, P. et al.: Metallomics 2014, 6: 1591–1601. 5. Zheng, X. et al.: Biotechnol. Prog. 2006, 22(5): 1294–1300. Acknowledgments: Financial support by Deutsche Forschungsgemeinschaft (DFG) and the state of Lower Saxony (graduate program SynFoBiA) is gratefully acknowledged. References: 1. Ott, I.: Coord. Chem. Rev. 2009, 253: 1670-1681. 2. Bertrand, B.; Casini, A.: Dalton Trans. 2014, 43: 4209-4219. 3. Oehninger, L.; Rubbiani, R.; Ott, I.: Dalton Trans. 2013, 42: 3269-3284. 4. Rubbiani, R. et al.: Med. Chem. Commun. 2013, 4: 942-948. 5. Meyer, A. et al.: Angew. Chem. 2012, 51: 8895-8899. 6. Bertrand, B. et al.: Inorg. Chem. 2014, 53: 2296-2303. 7. Rubbiani, R. et al.: ChemMedChem 2014, 9: 1205-1210. POS.117 Studies on the cytotoxicity of a novel family of synthetic pentathiepins Behnisch, S.1; Zubair, M.2; Lemmerhirt, H.1; Schulzke, C.3; Bednarski, P. J.1 1 Institute of Pharmacy, University of Greifswald, F.-L.-Jahn-Straße 17, 17489 Greifswald, Germany 2 School of Chemistry, Trinity College Dublin, College Green, Dublin 2, Ireland 3 Institute of Biochemistry, University of Greifswald, F.-Hausdorff-Straße 4, 17489 Greifswald, Germany POS.116 Fluorinated gold(i) n-heterocyclic carbene complexes with anticancer properties – investigation of cytotoxicity, enzyme inhibition, biodistribution and intestine permeability Schmidt, C.1; Reichl, S.2,3; Ott, I.1,3 1 Institut für Medizinische und Pharmazeutische Chemie, Technische Universität Braunschweig, Beethovenstraße 55, 38106 Braunschweig, Germany 2 Institut für Pharmazeutische Technologie, Technische Universität Braunschweig, Mendelssohnstraße 1, 38106 Braunschweig, Germany 3 Zentrum für Pharmaverfahrenstechnik, Technische Universität Braunschweig, Franz-LisztStraße 35 A, 38106 Braunschweig, Germany Auranofin represents a well investigated lead structure for gold Nheterocyclic carbene (NHC) complexes. It is established in the current antirheumatic therapy, shows antiproliferative effects and a remarkable inhibition of enzymes associated with the thioredoxin system, which has the ability to detoxify cell damaging reactive oxygen species (ROS) [1]. To assess the potential anticancer activities of a novel family of synthetic tetra-cyclic pentathiepins [1], we studied the ability of the compounds to induce apoptosis and initiate the formation of reactive oxygen species (ROS) in human cancer cells in culture. Natural pentathiepin derivates such as varacin (Figure, left) are known for their anticancer, antifungal and antibacterial activity. Varacin is also known to cause DNA cleavage in supercoiled plasmid DNA in vitro through a mechanism of hydroxyl radical formation in presence of thiols [2,3]. We have recently found that synthetic tetracyclic pentathiepins such as ZZ-QME2-Pn (Figure, right) have strong cytotoxic activities on a broad range of human cancer cell lines in vitro, with IC50-values ranging between 2.0-7.5 µM. Cancer cells treated with ZZ-QME2-Pn already show massive changes in morphology after an incubation time of only 6 h. These cells undergo apoptosis, as measured by flow of cytometry by the Annexin-V / propidium iodide method and cleavage of PARP, as measured by western blotting. Following a 30 min incubation of cells with ZZ-QME2-Pn, we observed by flow cytometry a stark increase in intracellular ROS levels, which could be dependent on cellular DPhG Annual Meeting 2015 Conference Book • 157 POSTERS glutathione (GSH) [4]. After preincubation of ZZ-QME2-Pn with GSH before application to cells, we found a significant decrease in ROS formation compared to cells without GSH preincubation. Thus, GSH would appear to protect cells from ROS formation. We also observed by flow cytometry an increase in cellular lipid peroxidation, which may be a direct result of ROS formation. These findings suggest that tetracyclic pentathiepins influence strongly the redox homeostasis of cells. Evidence has also been collected indicating that ZZ-QME2-Pn moderately inhibits isolated glutathione peroxidase (GPX1), but not the thioredoxin reductase (TxR). Further studies will be aimed at understanding the importance of GPX inhibition in cells. Figure: Varacin (left); ZZ-QME2-Pn (right) References: 1. Zubair, M. et al.: Chem. Commun. (Camb) 2013, 49: 4343-5. 2. Chatterji, T. et al.: Bioorg. Med. Chem. Lett. 1998, 8: 535-8. 3. Lee, A. H. et al.: Chem. Commun. (Camb) 2002, 2112-3. 4. Chatterji, T. et al.: Bioorg. Med. Chem. Lett. 2003, 13: 1349-52. POS.118 Inhibition of endothelial Cdk5 reduces tumor growth by promoting non-productive angiogenesis Merk, H.1; Zhang, S.1; Lehr T.2; Bibb, J. A.3; Adams R. H.4,5; Zahler, S.1; Vollmar A. M.1; Liebl, J.1 Department of Pharmacy, Pharmaceutical Biology, Ludwig-Maximilians-University, 81377 Munich, Germany Clinical Pharmacy, Saarland University, 66123 Saarbrücken, Germany 3 Department of Psychiatry and Neurology and Neurotherapeutics, The University of Texas Southwestern Medical Center, Dallas, Texas 75390-9070, USA 4 Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, 48149 Münster, Germany 5 Faculty of Medicine, University of Münster, 48149 Münster, Germany 1 2 For abstract see Short Poster Lecture SPL.007 on page 104. 158 • DPhG Annual Meeting 2015 Conference Book DRUG DESIGN/MEDICINAL CHEMISTRY 4.8 Drug design/Medicinal chemistry POS.119 Does FXR antagonism account for liver toxicity of nonsteroidal anti-rheumatic drugs (NSAIDs)? Schmidt, J.1; Klingler, F.1; Proschak, E.1; Steinhilber, D.1; SchubertZsilavecz, M.1; Merk, D.1 1 Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt a. M., Germany; The nuclear farnesoid X receptor (FXR) is a ligand-activated transcription factor, which acts as cellular sensor for bile acids [1-3]. It takes part in the self-regulation of bile acids with the result that bile acid synthesis is blocked and their metabolism is enhanced when high levels of toxic bile acids occur. Hence, FXR is a highly important liver protector [4] but is also involved in metabolic systems such as glucose or lipid homeostasis and seems to have anti-inflammatory effects as well. The most potent bile acid on FXR is chenodeoxycholic acid (CDCA) [3] whereas a number of synthetic FXR agonists and antagonists were discovered in the past years [5]. The 6α-ethyl derivate of CDCA, obeticholic acid (OCA), shows a 100-fold greater agonistic activity on FXR than its natural prototype. A number of studies showed the capacity of OCA to increase insulin sensitivity and to regulate glucose homeostasis. OCA also modulates lipid metabolism, and exerts anti-inflammatory as well as antifibrotic effects in the FXR-expressing organs, liver, kidney and intestine [6]. The most widely used non-steroidal FXR agonist is the synthetic isoxazole GW4064 which is not suitable as drug for its toxicity and poor bioavailability, however. While FXR agonism is interesting for potential therapeutic applications in the treatment of several liver disorders and metabolic diseases, little is known about the pharmacological effects of FXR antagonism. So far, antagonism on FXR does not seem to have broad therapeutic value. FXR seems to be overexpressed in some cancer cells, especially in pancreatic and colon cancer, Barett’s Esophagus and adenoma. These observations hold a limited therapeutic potential for FXR antagonism and on the other hand it might be a treatment strategy for cholestasis. However, in intestinal and hepatic cancer reduced FXR activity is connected with tumor growth and FXR antagonism might furthermore disturb adipogenesis [7]. A recent report by Lu et al. [8] described non-steroidal anti-rheumatic drugs (NSAIDs) as FXR antagonists and correlated the liver damage caused by chronic intake of these drugs with antagonistic activity on FXR. We were interested in this statement because of the possibility to design safer NSAIDs by generating cyclooxygenase inhibitors which spare FXR. On the other hand FXR antagonists are not common but they are required to evaluate the physiological and pathophysiological roles of FXR. The small and drug-like NSAIDs might therefore serve as lead structures for the development of potent FXR antagonists. However, we were confounded by the low maximum FXR activation in the test systems Lu et al. reported and the fact that NSAIDs with such different structures as diclofenac and indomethacin create a comparable activity on FXR. We therefore planned to further characterize the reported antagonistic activity and to scrutinize diclofenac, ibuprofen and indomethacin due to their high structural variety. We trialed the three NSAIDs in several test systems including a full-length reporter gene assay, a hybrid reporter gene assay, quantitative real-time PCR and thermal shift experiments [9]. Our results contradict the reported antagonistic effects of the NSAIDs on FXR as we found no evidence for FXR activity of NSAIDs alone or in competition with the well-known FXR agonists GW4064, OCA or CDCA. In contrast, we discovered that the activity reported by Lu et al. is due to misinterpreted toxicity. We therefore conclude that reduced FXR signaling in presence of NSAIDs is merely a consequence than a cause of hepatotoxicity POS.120 Identification of aza-stilbenes as inhibitors of the cellular checkpoint kinase Myt1 Platzer, C.1; Decker, C.1; Gershkovich, M.2; Erdmann, F.1; Sippl, W.1; Schmidt, M.1 1 Institute of Pharmacy, Department of Medicinal Chemistry, Martin-Luther-University-HalleWittenberg, Wolfgang-Langenbeck-Str. 4, 06120 Halle (Saale), Germany 2 Institute of Biochemistry and Biotechnology, Department of Enzymology, Martin-LutherUniversity-Halle-Wittenberg, Kurt-Mothes-Str. 3, 06120 Halle (Saale), Germany Human Myt1 kinase is a negative regulator of Cdk1/Cyclin B complex and maintains the G2/M transition in the cell cycle especially in cancer cells. Many cancer cell lines only depend on G2 checkpoint because of G1 defect caused by p53 mutation [1]. Therefore, the inhibition of G2 checkpoint represents a promising concept for preferential damaging of cancer cells without affecting normal cells [2]. Investigations of Chow and Poon revealed that Myt1 seems to be more essential for cancer cells than for healthy cells [3], making Myt1 an interesting target for anticancer therapy. At the moment only few compounds are known to inhibit Myt1. In the current work, we established several in vitro assays and tested a library of 800 known kinase inhibitors available from GlaxoSmithKline (Kinase Inhibitor Set I and II). In our screen we identified several hits including aza-stilbenes as new class of Myt1 inhibitors. We investigated the binding affinity and inhibition of the compounds at Myt1 full length and kinases domain75-362 with a fluorescence anisotropy-based binding assay and a short-peptide based activity assay. The obtained results can be used for establishing structure-activity relationships as well as for the structure-based optimization. Acknowledgments: GlaxoSmithKline LLC References: 1. Levine, A.: Cell 1997, 88(3): 321-331. 2. Kawabe, T.: Mol. Cancer Ther. 2004, 3(4): 513-519. 3. Chow, J.P.; Poon, R.Y.C.: Oncogene 2013, 32(10): 4778-4788. POS.121 Identification of new potential inhibitors of GyrB ATPase Schieback, P.1; Lemcke, T.1 1 University of Hamburg, Institute of Pharmacy, Bundesstr. 45, 20146 Hamburg, Germany Since multi drug resistance is present in pathogenic bacteria due to increasing use of antibiotics worldwide, the design of potential new antibiotics becomes more important to deal with bacterial infections like respiratory and urinary tract infections [1]. One very well-known target for antibiotics, such as fluoroquinolones, is DNA gyrase. It is a target that only exists in bacteria and not in human cells, and it has a high degree of sequence conservation throughout many bacterial species [2]. DNA gyrase is divided into two subunits and only drugs inhibiting subunit A (GyrA) are actually in clinical use. In addition to the development of multiresistant strains in bacteria, subunit A inhibitors exhibit also several side effects. Inhibition of the ATPase binding pocket of Gyrase subunit B (GyrB) is getting more important although Novobiocin, which is an aminocoumarine derivate and a highly potent GyrB inhibitor, was unsuccessful in clinic due to severe side effects. A virtual screening campaign [3] of a subset of the ZINC database resulted in a collection of potential ligands of the GyrB ATP-binding site of E.coli. For the final hit ranking and compound selection, a 3D QSAR model [4], generated using literature known GyrB ATPase inhibitors, was used. Acknowledgments: We thank Dr. Krister Bamberg, AstraZeneca R&D Mölndal, Sweden, for the generous gift of the recombinant FXR-LBD. References: 1. Seol, W.; Choi, H. S.; Moore, D. D.: Mol. Endocrinol. 1995, 9(1): 72–85. 2. Forman, B. M. et al.: Cell 1995, 81(5): 687–693. 3. Parks, D. J. et al.: Science 1999, 284(5418): 1365–1368. 4. Kuipers, F.; Bloks, V. W.; Groen, A. K.: Nat. Rev. Endocrinol. 2014, 10(8): 488–498. 5. Merk, D.; Steinhilber, D.; Schubert-Zsilavecz, M.: Future Med. Chem. 2012, 4(8): 1015–1036. 6. Luciano Adorini, L.; Pruzanski, M.; Shapiro, D.: Drug Discov. Tod. 2012, 17(17): 988-997. 7. Lamers, C.; Schubert-Zsilavecz, M.; Merk, D.:Curr. Top. Med. Chem. 2014, 14(19): 2188-2205. 8. Lu, W. et al.: Sci. Rep. 2015, 5: 8114. 9. Schmidt, J. et al.: Sci. Rep.2015, submitted. Figure: Docking pose of hit A in the GyrB ATPase binding site of E. coli DPhG Annual Meeting 2015 Conference Book • 159 POSTERS 23 structures were purchased and tested in a fluorescence quenching assay for gyrase inhibiting activity. A gyrase supercoiling assay was used to verify the inhibitory activity of all hits. Finally, three structurally divers substances were identified as potential GyrB inhibitors. To expand the hit set and to get a preliminary SAR, five additional derivatives of hit A were docked into the GyrB ATP binding site, evaluated using the validated 3D QSAR model and finally purchased and tested. All substances show inhibitory activity in the micro molar range. The results of the hit expansion and biological testing of these new potential GyrB ATPase inhibitors will be presented on this paper. The relevance of CLKs in the onset of the abovementioned diseases and the lack of selectivity of current inhibitors emphasises the need for novel small molecules as tools for biological experiments or as lead compounds in the drug discovery process. We here present the synthesis and biological evaluation of a structurally new class of CLK inhibitors. Representatives of the 6,7-dihydropyrrolo[3,4-g]indol-8-ones selectively inhibit CLKs and are not active on a panel of related serine threonine kinases including DYRK1A. References: 1. Hagiwara, M.: Biochim. Biophys. Acta. 2005, 1754: 324-331. 2. Liu, Y. et al.: Nucleic Acids Res. 2013, 41: 4949-4962. 3. Glatz, D.C. et al.: J. Neurochem. 2006, 96: 635-644. References: 1. Nakaminami, H. et al.: Int. J. Antimicrob. Ag. 2014, 43(5): 478-479. 2. Tari, L.W.; et al.: PLoS ONE 2013, 8(12): e84409. 3. Münsterberg, M.et al.: Presentation EFMC-ISMC Lisbon. 2014. 4. Schieback, P.; Lemcke, T.: Presentation EFMC-ISMC Lisbon. 2014. POS.124 POS.122 Development and characterization of a compound series based on a hit structure obtained from the Tres Cantos Antimalarial Compound Set Weidner, T.1; Nasereddin, A.2; Preu, L.1; Grünefeld, J.1; Dzikowski, R.2; Kunick, C.1 1 Institut für Medizinische und Pharmazeutische Chemie, Technische Universität Braunschweig, Beethovenstraße 55, 38106 Braunschweig, Germany 2 Department of Microbiology and Molecular Genetics, IMRIC, Hebrew University, Hadassah Medical School, P.O. Box 12272, Jerusalem 91220, Israel Publicly available substance libraries provide various starting points for the development of new drugs against malaria parasites. The Tres Cantos Antimalarial Compound Set (TCAMS), containing 13,533 structures with activity against Plasmodium falciparum [1], includes the compound TCMDC-137332, which was suggested as one of “47 quality starting points for malaria drug discovery” [2]. TCMDC-137332 appears to be one of the most potent structures of this collection and, in addition, it matches with the requirements of the Lipinski rules for orally available drugs [3]. Analogues of TCMDC-137332, which are based on a 2phenoxyanilide scaffold, were synthesized by standard amide coupling reactions. The poster reports the synthesis of overall 19 derivatives, which were completely characterized regarding identity and purity by spectroscopic as well as chromatographic methods and elemental analyses. Furthermore, the results of the biological evaluation of all new congeners against P. falciparum erythrocyte stage parasites will be presented. Acknowledgments: This project was funded by the German Federal Ministry of Education and Research (BMBF BioDisc 7; 13GW0024) References: Computer-based studies of Bisamidines as PRMT1 and PRMT6 inhibitors Robaa, D.1; Sauer, B.1; Wagner, T.2; Jung, M.2; Sippl, W.1 1 2 Department of Pharmaceutical Chemistry, Martin-Luther University Halle-Wittenberg Institute of Pharmaceutical Sciences, Albert-Ludwigs University Freiburg Protein arginine methyltransferases (PRMTs) are a conserved family of proteins which catalyze the posttranslational N-methylation of arginine residues. They constitute an important family of epigenetic enzymes, which can regulate gene expression through histone methylation. Besides histones, PRMTs have numerous non-histone proteins as substrates, including RNA-binding proteins, signal transducers and transcriptional coregulators [1]. PRMTs appear to play a role in the pathogenesis of cancer [2], diabetes [3] and cardiovascular diseases [4] and to be important for viral replication [5], suggesting that these enzymes are interesting targets for drug discovery. Virtual screening approaches of various compound databases were successfully deployed in our research group and led to the identification of several lead structures as PRMT1 inhibitors, including bisamidines [6-8]. To study the binding of bisamidines to PRMT1 and PRMT6, docking and molecular dynamic studies were performed on generated homology models of both enzymes. Additionally, structural optimization of these lead compounds, guided by in silico studies, was carried out. References: 1. Lee, Y.; Stallcup, M.R.: Mo.l Endocrinol. 2009, 23(4): 425–433. 2. Yoshimatsu, M. et al.: Int. J. Cancer. 2011, 128(3): 562–573. 3. El-Osta, A. et al.: J. Exp. Med. 2008, 205(10): 2409–2417. 4. Vallance, P.; Leiper, J.: Arterioscler. Thromb. Vasc. Biol. 2004, 24(6):1023–1030. 5. Xie, B. et al.: J. Virol. 2007, 81(8): 4226–4234. 6. Spannhoff, A. et al.: J. Med. Chem. 2007, 50(10): 2319–2325. 7. Spannhoff, A. et al.: Bioorg. Med. Chem. Lett. 2007, 17(15): 4150–4153. 8. Heinke, R. et al.: ChemMedChem 2009, 4(1): 69–77. 1. Gamo, F.-J. et al.: Nature 2010, 465(7296): 305–310. 2. Calderón, F. et al.: ACS Med. Chem. Lett. 2011, 2(10): 741–746. 3. Lipinski, C. A. et. al.: Adv. Drug Delivery Rev. 1997, 23(1-3): 3–25. POS.125 POS.123 Novel selective inhibitors of the splicing regulatory CDC2like kinases (CLKs) Walter, A.1; Helmer, R.1; Loaëc, N.2; Preu, L.1; Meijer, L.2; Kunick, C.1 1 Institut für Medizinische und Pharmazeutische Chemie, Technische Universität Braunschweig, Beethovenstraße 55, 38106 Braunschweig, Germany 2 ManRos Therapeutics, Hôtel de Recherche, Centre de Perharidy, 29680 Roscoff, France CLK family kinases (CDC2-like kinases) play an important role in the regulation of pre-mRNA splicing by phosphorylating several splicing factors [1]. Alternative splicing, occurring after transcription of DNA into pre-mRNA, is an essential process in eukaryotes and an origin of protein diversity. Strict control of the splicing mechanism is required as a change might potentially have significant effects on the cellular proteome or even on the whole organism. Any dysregulation may alter or abolish the original function of an affected protein thus contributing to the development of diseases. Abnormal splicing influenced by the phosphorylation of splicing factors by CLKs was observed in the pathogenesis of frontotemporal dementia and ovarian cancer [2,3]. 160 • DPhG Annual Meeting 2015 Conference Book Development of new Dengue Virus NS2B-NS3 protease inhibitors and the investigation on their mode of action von Hammerstein, F.1; Gellert, A.1; Wu, H.1; Holloway, S.1; Kiefer, W.1; Bodem, J.2; Kanitz, M.3; Steuber, H.3; Diederich, W.3; Hellmich, U.1; Schirmeister, T.1 Institute of Pharmacy and Biochemistry, University of Mainz, Staudingerweg 5, D-55099 Mainz, Germany 2 Institute of Virology and Immunology, University of Würzburg, Versbacher Strasse 7, D-97078 Würzburg, Germany 3 Institute of Pharmaceutical Chemistry, University of Marburg, Marbacher Weg 6-10, D-35032 Marburg, Germany 1 Dengue fever is a mosquito-borne tropical disease caused by the dengue virus (DENV). It is transmitted to humans by the Aedes aegypti and Aedes albopictus mosquitoes [1]. This viral infection is becoming continually a global threat, as there are nearly 3.6 billion people living in the areas, tropical and subtropical regions of the world (predominantly in Southeast Asia, Africa and the Americas), where the infection is common [2,3]. The DENV infection can result in classic dengue fever, dengue hemorrhagic fever (DHF) or dengue shock syndrome (DSS) [4]. Worldwide there are over 50 million infections reported annually and the infection causes over 20,000 deaths each year [5]. DENV is a single DRUG DESIGN/MEDICINAL CHEMISTRY positive-stranded RNA virus of the family Flaviviridae with four distinct serotypes [6,7]. The dengue virus genome encodes a serine protease with a classical catalytic triad (His51, Asp75 and Ser135) [8,9], which is responsible for the post-translational proteolytic processing of the polyprotein precursor and essential for the viral replication [10,11], making it an important and attractive therapeutic target [12]. Our projects [13] include synthesis, characterization and testing of DENV2/3 NS2B-NS3pro inhibitors based on the structure of variously substituted diaryl (thio)ethers. The synthesized compounds are screened both in vitro in fluorometric enzyme assays using different fluorogenic AMC-derived substrates or by microscale thermophoresis (MST) [14] and in cell culture. The possible binding modes are analyzed by docking studies and are being examined by NMR spectroscopy. Acknowledgments: University of Mainz for financial support. References: 1. Yildiz, M. et al.: ACS Chem. Biol. 2013, 8: 2744-2752. 2. Murray, N. E. A. et al.: Clinical Epidemiology 2013, 5: 299-309. 3. Guzman, M. G. et al.: Nat. Rev. Microbiol. 2010, 8: S7-S16. 4. Martina, B. E. et al.: Clin. Microbiol. Rev. 2009, 22: 564-581. 5. Wilder-Smith, A. et al.: Arch. Med. Res. 2002, 33(4): 330-342. 6. Li, H. et al.: J. Virol. 1998, 73(4): 3108-3116. 7. Chambers, T. J. et al.: Annu. Rev. Microbiol. 1990, 44: 649-688. 8. Bazan, J. F.; Fletterick, R. J.: Virilogy 1989, 171: 637-639. 9. Nitsche, C. et al.: Chem. Rev. 2014, 114(22): 11348–11381. 10. Falgout, B. et al.: J. Virol. 1991, 65: 2467-2475. 11. Zhang, L.; Mohan, P. M.; Padmanabhan, R.: J. Virol. 1992, 66: 7549-7554. 12. Zheng, Y. et al.: Bioorg. Med. Chem. Lett. 2006, 16: 36-39. 13. Wu, H. et al.: Antimicrob. Agents Chemother. February 2015, 59(2): 1100-1109. 14. Jerabek-Willemsen, M. et al.: Assay Drug Dev. Technol. 2011, 9(4): 342-353. POS.126 Novel purine-based inhibitors of anaplastic lymphoma kinase (ALK) Schlütke, L.1; Preu, L.1; Totzke, F.2; Schächtele, C.2; Kubbutat, M. H. G.2; Kunick, C.1 1 Institut für Medizinische und Pharmazeutische Chemie, TU Braunschweig, Beethovenstraße 55, 38106 Braunschweig, Germany 2 ProQinase GmbH, Breisacher Straße 117, 79106 Freiburg, Germany Chromosomal rearrangements of anaplastic lymphoma kinase (ALK) have been described in several human cancer diseases [1]. Deregulation of ALK was first identified in anaplastic large cell lymphoma (ALCL), which is associated with NPM (nucleophosmin) ALK fusion gene [2]. The similar fusion gene EML4-ALK plays an important role in the development in 5% of all non-small cell lung carcinoma (NSCLC) [3]. In 2011 the first dual ALK/cMet inhibitor Crizotinib was approved. Although Crizotinib shows a high response rate, emerging resistances are observed with a median of 10.5 months [3]. Second-generation ALK inhibitors like Ceritinib and Alectinib address some of the mutations underlying these resistances [4]. Further development of structurally unrelated inhibitors remains a challenging problem. Design by dockingstudies, syntheses, and kinase inhibitory activities of the title compounds will be presented in the poster. Acknowledgments: L.S. is grateful for a stipend funded by the Cusanuswerk, Bonn, Germany. References: 1. Bossi, R. et al.: Biochemistry 2010, 49: 6813-6825. 2. Morris, S. W. et al.: Science 1994, 263: 1281-1284. 3. Roskoski, R. et al.: Pharmacol. Res. 2013, 68: 68-94. 4. Kinoshita, K. et al.: Biorg. Med. Chem. 2012, 20: 1271-1280. POS.127 Design and synthesis of 2-ureidothiophene-3-carboxylic acids as dual bacterial RNAP and HIV-1 RT inhibitors for treatment of patients co-infected with MRSA and HIV-1 Elgaher, W. A.1; Sharma, K. 2; Haupenthal, J. 1; Mély, Y. 2; Hartmann, R. W.1 Department of Drug Design and Optimization, Helmholtz Institute for Pharmaceutical Research Saarland, Campus C2.3, Saarland University, 66123 Saarbrücken, Germany, 2 Laboratory of Biophotonics and Pharmacology, Faculty of Pharmacy, Strasbourg University, 74 route du Rhin, 67401 Illkirch, France 1 Human immunodeficiency virus type 1 (HIV-1) is a retrovirus that targets and destroys the CD4 cells of the immune system. Consequently, it causes the acquired immune deficiency syndrome (AIDS), which is one of the biggest killer infectious diseases worldwide. Even worse, HIV patients are easily attacked by bacteria, e.g. methicillin-resistant Staphylococcus aureus (MRSA) [1]. Current antiretroviral therapy (ART) is a combination of at least three antiretroviral drugs to allow efficient suppression of virus replication. However, in this context viral resistance is presenting a major problem. The treatment of an HIV-1/MRSA coinfection is even more critical. Administration of antibacterial agents should consider the ongoing prevalence of resistant bacteria in HIV patients [2], as well as potential interactions between ART drugs and the antibacterial agents [3]. This prompted us to develop novel compounds with both antiretroviral and antibacterial activities. This was achieved via exploiting, for the first time, the mechanistic function similarity between the RNA polymerase (RNAP) switch region and the non-nucleoside reverse transcriptase inhibitor (NNRTI) binding site. Starting from our bacterial RNAP inhibitors [4,5], we succeeded in developing potent RT inhibitors while retaining or enhancing the RNAP inhibitory activity following a structure-based drug design approach. A SAR study revealed structural features necessary for RT inhibition. The novel compounds showed good antiviral (in cellulo assay) and antibacterial activities (against MRSA) in the low micromolar range, along with low or no cytotoxicity. Furthermore, the mode of action of our novel inhibitors was studied regarding RT, revealing a non-competitive inhibition mechanism similar to nevirapine. References: 1. Hidron, A. I. et al.: Infect. Drug Resistance 2010, 3: 73−86. 2. Diep, B. A. et al.: Ann. Intern. Med. 2008, 148: 249−257. 3. Piscitelli, S. C.; Gallicano, K. D.: N. Engl. J. Med. 2001, 344: 984−996. 4. Sahner, J. H. et al.: Eur. J. Med. Chem. 2013, 65: 223−231. 5. Elgaher, W. et al.: RSC Adv. 2014, 4: 2177−2194. POS.128 A direct Enzyme-Linked Immunosorbent Assay (ELISA) for the quantitative evaluation of Janus Kinase 3 (JAK3) inhibitors Bauer, S. M.1; Gehringer, M.1; Laufer, S. A.1 1 Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmaceutical Sciences, Eberhard Karls University of Tuebingen, D-72076 Tuebingen, Germany Janus kinases (JAKs) are a group of non-receptor tyrosine kinases involved in the signal transduction processes of cytokines, growth factors and interleukins. Thus, the four JAK family members JAK 1,2,3 and TYK 2 are potential targets for treatment of inflammatory diseases as well as oncological disorders [1-3]. Being restricted to the hematopoietic lineage, JAK 3’s limited tissue expression makes it an attractive drug target for the treatment of inflammation with limited side effects. Several JAK inhibitors are currently under investigation in clinical trials and the pan-JAK inhibitor Tofacitinib has recently been approved by the FDA [4]. With Ruxolitinib a JAK 1 and JAK 2 selective inhibitor for the treatment of myelofibrosis was approved by the EMA [5]. Even though being a promising target, no selective JAK 3 inhibitor is available for therapy until now. In an academic environment, the availability of a robust, precise and costefficient test system is the prerequisite for the performance of any medicinal chemistry program. For identification of novel JAK 3 inhibitors and determination of structure activity relationships, a direct enzymelinked immunosorbent assay was developed. Being applicable in a standard laboratory, the main advantage over existing methods is the basic equipment requirement as this assay does not need special handling originating from the use of cell cultures or radiometric readout. DPhG Annual Meeting 2015 Conference Book • 161 POSTERS In contrast to earlier methods relying on two-step procedures, our assay uses a single peroxidase conjugated antibody to quantify and detect substrate phosphorylation. This optimized assay features a limit of detection of 0.032 µg/mL, a wide dynamic range of 0.638 OD units and an appropriate Z’ factor of 0.66. The determined IC50 value for Tofacitinib was 3.5±0.6 nM (n = 18). For investigation of structure activity relationships, the assay is readily discriminating the effects on kinase inhibition even within a series of structurally similar molecules and is thus suited for lead optimization. When screening our Tofacitinib-derived compound library, the reported ELISA allowed the determination of inhibitory potencies in the low nanomolar range which were subsequently confirmed by commercial radiometric and cellular assays. As a result of our research in this area, we recently described a new class of highly potent tricyclic JAK inhibitors with superior selectivity compared to Tofacitinib [6]. The developed ELISA primarily addresses researchers within the academic setting facing the task to identify lead structures and explore structure activity relationships of JAK 3 inhibitors at reasonable costs. Acknowledgments: The authors want to thank L. Fischer and K. Bauer for successful laboratory co-work during the establishment of this assay. Special thanks to Jun.-Prof. Dr. Pierre Koch and Dr. Peter Keck for their help, support and advice. References: 1. Pesu, M. et al.: Immunol. Rev. 2005, 203: 127 - 142 2. Kim, B.H. et al.: Br. J. Haematol. 2010, 148: 132-143. 3. Haan, C.; et al.: Chem. Biol. 2011, 18: 314-323. 4. O'Shea, J.J.: Ann. Rheum. Dis. 2013, 72: 111-115. 5. Tefferi, A. et al.: Blood 2012, 119: 2721 – 2730. 6. Gehringer, M. et al.: ChemMedChem. 2014, 9: 277-281. POS.130 Thieno[3,2-d]pyrimidin-2-amine derivatives as inhibitors of anaplastic lymphoma kinase Immer, M.1,2; Wölfel, S.1; Preu, L.1; Totzke, F. 3; Schächtele, C. 3; Kubbutat, M. H. G.3; Kunick, C.1,2 1 Institut für Medizinische und Pharmazeutische Chemie, TU Braunschweig, Beethovenstraße 55, 38106 Braunschweig, Germany 2 Center of Pharmaceutical Engineering (PVZ), TU Braunschweig, Franz-Liszt-Straße 35A, 38106 Braunschweig, Germany 3 ProQinase GmbH, Breisacher Straße 117, 79106 Freiburg, Germany The tyrosine kinase anaplastic lymphoma kinase (ALK) is a well known target in various cancer diseases. ALK was identified for the first time as a part of the nucleophosmin (NPM)-ALK fusion protein, which was detected in 60% of anaplastic large cell lymphoma (ALCL) patients [1]. The fusion gene echinoderm microtubule-associated protein like 4 (EML4)-ALK was identified in approximately 5% of all non-small-cell lung cancer (NSCLC) patients [2]. In 2011, Crizotinib was approved by the FDA as the first ALK inhibitor. In 2014 two other ALK inhibitors, Alectinib and Ceritinib, were approved [3,4]. Due to numerous mutations of ALK and thereby acquired resistance the further development of new ALK inhibitors is of great interest. In this poster, we present results of docking experiments of thieno[3,2-d]pyrimidin-2-amine derivatives in the ATP binding pocket of ALK, as well as the syntheses and the ALK inhibitory activity of these compounds. Acknowledgments: M.I. is grateful for a stipend in the framework of „SynFoBiA“, a cooperative research progam funded by “VW-Vorab”. References: POS.129 Probing the preferred binding orientation of the axially oriented phenyl moiety of 1,3-dioxane NMDA receptor antagonists by conformational restriction 1. Morris, S. W. et al.: Science 1994, 263: 1281-1284. 2. Shaw, A. T. et al.: N. Engl. J. Med. 2013, 36: 2385–2394. 3. Kinoshita, K. et al.: Biorg. Med. Chem. 2012, 20: 1271-1280. 4. Shaw, A. T. et al.: N. Engl. J. Med. 2014, 370: 1189-1197. Asare-Nkansah, S.1; Wünsch, B.1 Institut für Pharmazeutische und Medizinische Chemie, Corrensstraße-48, D-48149 Münster, Germany. POS.131 1 The N-methyl-D-aspartate (NMDA) receptor is a ligand-gated ion channel that plays a prominent role in various central nervous system (CNS) events such as development of neurons, synaptic plasticity, memory and learning [1]. The NMDA receptor is highly permeable to Ca2+ ions, and under pathological conditions, elevated intracellular Ca2+ ion concentrations up to cytotoxic levels contribute partly to neuronal death (excitotoxicity. The NMDA receptor is consequently implicated in diseases of the CNS (Alzheimer’s disease, Parkinson’s disease, Huntington’s disease and epilepsy), which are associated with excitotoxicity [2]. Therefore, the NMDA receptor represents a potential therapeutic target for the development of innovative drugs for the treatment of neurological disorders. The 1,3-dioxolane derivatives dexoxadrol (1) and etoxadrol (2) have high NMDA receptor affinity (Figure) with non-tolerable side effects including retrograde amnesia and psychotomimetic effects [3]. Studies performed on 1 and 2 have led to the development of 1,3-dioxane analogs 3 with even higher affinity towards the phencyclidine (PCP) binding site of the NMDA receptor [3,4]. The phenyl moiety in axial orientation at the acetalic C-atom of 3 is postulated as essential for binding with high affinity to the NMDA receptor. The conformationally restricted ligands 4 should therefore give insight into the preferred binding orientation of the phenyl moiety (parallel (3a) or perpendicular (3b)). In this project, the phenyl moiety is fixed in an axial position perpendicular to the 1,3-dioxane ring (4). Figure: Development of novel 1,3-dioxane based NMDA receptor antagonists References: 1. Ono, S. et al.: Chem. Pharm. Bull. (Tokyo) 2002, 50: 966–8. 2. Flores-Soto, M. E. et al.: Neurologia 2012, 27: 301–10. 3. Aepkers, M.; Wünsch, B.: Bioorg. Med. Chem. 2005, 13: 6836–49. 4. Utech, T.; Köhler, J.; Wünsch, B.: Eur. J. Med. Chem. 2011, 46: 2157-69. 162 • DPhG Annual Meeting 2015 Conference Book The Trypanothione Synthetase ATP Binding Site: Flexibility and Similarity to Other ATP Binding Proteins Ehrt, C.1; Brinkjost, T.1,2; Koch, O.1 1 Faculty 2 Faculty for Chemistry and Chemical Biology, TU Dortmund, Germany of Computer Science, TU Dortmund, Germany Trypanothione synthetase (TryS) catalyzes the two-step biosynthesis of trypanothione, which is a key intermediate in trypanosomatid parasites of the species Trypanosoma and Leishmania. Thus, TryS is an attractive drug target to cope with neglected diseases like Chagas disease, leishmaniasis, or African trypanosomiasis that affect approximately 15 to 20 million people worldwide. Interestingly, paullones, a chemical class of potent kinase (GSK-3β and different cyclin-dependent kinases) inhibitors, were shown to inhibit TryS [1]. Based on a comparison of available structures, the binding of paullones to TryS seems to be in accordance with the principle of ligand-sensing cores [2]. That is, the spatial arrangement of secondary structure elements around the ATP binding sites of TryS and kinases is quite similar, independent of the overall fold, which indicates binding of similar scaffolds. Unfortunately, the available Leishmania major TryS X-ray structure (pdbid 2vps) was solved without substrates and an important loop region of the ATP grasp fold is missing. The structure of a related GSP synthetase from Escherichia coli (pdb‑id 2io7) led to a complete model of LmTryS, containing the substrate ATP. Exhaustive MD simulations confirmed this model and revealed that the presence of ATP leads to a partial closure of an associated β-sheet over the bound triphosphate. This validated model gave us the possibility to model structures of TryS from other pathogenic species of Trypanosoma and Leishmania and to compare their ATP binding sites with respect to rational drug design. Furthermore, the conformational space of this highly flexible binding site was elucidated with the help of accelerated MD simulations. As we recently developed a reasonably fast method to search for common ligand-sensing cores in a set of different protein structures based on graph comparisons, we used this method to compare the ATP binding sites of different kinases and our modelled TryS structures from various species. We searched for ATP binding proteins in the Protein Data Bank and compared them not only against representative TryS structures, but also against each other. Structural similarities between DRUG DESIGN/MEDICINAL CHEMISTRY different TryS structures and certain kinases can be shown by only taking into account the protein backbone. A structural comparison of the involved secondary structure elements and the residues which are part of the common ligand-sensing core revealed that, although the backbone architecture is quite similar in both proteins, the underlying residues are quite different regarding physicochemical properties. Known highly potent inhibitors of kinases with the highest similarity to TryS were extracted from the ChEMBL database, clustered and afterwards docked into different TryS ATP binding sites from the MD simulations to find potential candidates for biochemical testing and further modification with regard to selectivity. Here, we present this workflow as a new method to exploit the huge knowledge about kinase inhibitors for the treatment of neglected diseases and facilitate drug discovery for novel targets. We will also show the results of the comparison of ATP binding protein structures against each other and discuss how this can help to extend our knowledge with regard to offtarget effects and structural classification. References: 1. Koch, O.: Drug Discovery in Infectious Diseases (Wiley-VCH) 2013: 429-443. 2. Koch, O. et al.: Future Med. Chem. 2011, 3: 699-708. POS.132 A lovely Liaison: Privileged Scaffolds and conserved Structural Elements in Proteins Humbeck, L.1; Koch, O.1 1 TU Dortmund, Otto-Hahn-Straße 6, 44221 Dortmund, Germany The term “privileged scaffolds” is often used for multiple molecules that show bioactivity on different targets but consist of the same scaffold [1]. Within proteins, conserved structural elements can also be found in different proteins, ranging from conserved motifs that interact with specific functional groups to similar spatial arrangements of secondary structure elements around the ligand binding site (the “ligand-sensing core”) in proteins with different folding patterns that can bind similar scaffolds [2]. Information about similar ligand-sensing cores can be useful for rational identification of new lead structures [3] or predicting polypharmacology [2]. Chemical compound databases like DrugBank (http://www.drugbank.ca/) or ChEMBL (https://www.ebi.ac.uk/chembl/) contain a huge amount of data about molecules and their bioactivity on different protein targets. Therefore, we decided to develop a Python based tool for knowledge discovery to get new insights into the relationship of privileged scaffolds and conserved structural elements in proteins. The main idea of this data mining approach is the identification of scaffolds that bind to different and unrelated protein targets for analyzing potential conserved structural elements. In a first step, a command line version of Scaffold Hunter [4] is used to reduce all molecules in a database to their containing scaffolds. The second step analyzes the sequence similarity of protein targets of all molecules sharing a common scaffold. Only protein targets with identity below 40% are regarded as unrelated. The last step visualizes the results for an in-depth analysis. We will present the overall workflow and the result of an exhaustive chemogenomics analysis of the DrugBank. Around 1500 scaffolds were identified that are active against different protein targets. An analysis of one example already ended up in a new ligand-sensing core that is shared between four different protein targets and can help to identify new lead structures for the respective targets. To prove the hypothesis of relationships between privileged scaffolds and conserved structural elements in proteins (the ligand-sensing cores) we also present the results of a high-throughput screen (HTS) against new inhibitors of one protein of the novel ligand-sensing core, which were similar to known ligands of one of the other proteins of the ligand-sensing core. Acknowledgments: We thank the BMBF (Grant No. 1316053) and DFG (“Algorithms for Big Data” SPP 1736) for funding. References: 1. Welsch, M. E.; Snyder, S. A.; Stockwell, B. R.: Curr. Opin. Chem. Biol. 2010, 14(3): 347-361. 2. Koch, O.: Fut. Med. Chem. 2011, 3(6): 699-708. 3. Willmann, D. et al.: Int. J. Cancer. 2012, 131(11): 2704-2709. 4. Klein, K. et al.: Mol. Inf. 2013, 32: 964–975. POS.133 Discovery of the first small-molecule CsrA-RNA interaction inhibitors as potential anti-infectives using surface plasmon resonance and fluorescence polarization-based screening Maurer, C. K.1,*; Fruth, M.1,*; Empting, M.1; Herrmann, J.1,2; Müller, R.1,2; Dersch, P.3; Hartmann, R. W. 1,4 1 Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Saarland University, Campus C2.3, 66123 Saarbrücken, Germany 2 German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, 30625 Hannover, Germany 3 Helmholtz Centre for Infection Research (HZI), Inhoffenstraße 7, 38124 Braunschweig, Germany 4 Department of Pharmaceutical and Medicinal Chemistry, Saarland University, Campus C2.3, 66123 Saarbrücken, Germany *These authors contributed equally to this work. Bacteria use global regulators to adapt gene expression in response to environmental changes. For instance, the regulator protein CsrA affects translation and stability of messenger RNA by binding to the ribosome binding site of target trancripts [1]. CsrA is widespread among bacteria and is highly conserved in structure and RNA recognition [2]. As global regulator, it controls numerous physiological processes involved e.g. in virulence, metabolism, motility, and biofilm formation [3]. Notably, a csrA knockout mutant of the enteropathogen Yersinia pseudotuberculosis exhibited attenuated virulence in a murine infection model (P. Dersch, unpublished data). Therefore, we considered CsrA as promising target for development of novel anti-infectives. To date, no low-molecular-weight inhibitor of the CsrA-RNA interaction has been described. Thus, for the discovery of such inhibitors, we followed two strategies, a screening and a ligand-based approach. During primary screening, compounds binding in a concentrationdependent manner to Yersinia pseudotuberculosis CsrA should be identified using surface plasmon resonance technology (SPR). Secondary screening based on a fluorescence polarization (FP)-based competition assay should filter out CsrA binders that could inhibit the CsrA-RNA interaction. From a library comprising 712 small molecules, 76 CsrA binders were identified, 4 out of which inhibited the CsrA-RNA interaction. Additionally, screening of a library consisting of 259 myxobacterial metabolites yielded 4 CsrA-RNA interaction inhibitors. Hit validation with the FP assay revealed dose-dependent inhibition with low micromolar Ki values for the 7 studied screening hits. The most promising compound exhibited a Ki of 2 µM. For the ligand-based strategy, short oligonucleotides of varying length and nucleic acid backbone derived from the highly conserved RNA binding motif were systematically studied [4]. In the FP assay, a GGA-motif with RNA-backbone turned out to be the most active ligand-derived inhibitor so far with a Ki value of 48 µM. The identified hits will be further characterized regarding their binding behaviour by SPR and regarding their cellular activity in virulence assays with different bacterial species such as Yersinia pseudotuberculosis and Pseudomonas aeruginosa. References: 1. Romeo, T.: Mol. Microbiol. 1998, 29(6):1321-30. 2. Dubey, A. K.et al.: RNA 2005, 11(10):1579–1587. 3. Timmermans, J.; Van Melderen, L.: Cell Mol Life Sci. 2010, 67(17):2897–2908. 4. Lapouge, K. et al.: RNA Biol. 2013, 10(6): 1030–1041. POS.134 Shifting Selectivity: From JAK1/2 to JAK3 inhibition via click chemistry Forster, M.1; Gehringer, M.2; Bauer, S. M.1; Laufer, S. A.1 1 Institute of Pharmacy, Medicinal Chemistry, Eberhard-Karls-Universitaet Tuebingen, Auf der Morgenstelle 8, 72076 Tuebingen, Germany 2 Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Vladimir-Prelog-Weg 1-5/10, 8093 Zurich, Switzerland The Janus kinase family (JAK) is consisting of four members, JAK1-3 and TYK2. Belonging to the non-receptor tyrosine kinases they associate with cytokine receptors and mediate extracellular signals via the phosphorylation of STAT-proteins (signal transducer and activator of transcription). Whereas the other members of the JAK family are expressed ubiquitously, JAK3 is predominantly found in cells of the haematopoietic system where it has a crucial function in the maturation of T-cells, B-cells and natural killer cells. Patients with a malfunction of JAK3 develop the phenotype of severe combined immunodeficiency (SCID) and are lacking T-lymphocytes and NK-cells. The restriction of its DPhG Annual Meeting 2015 Conference Book • 163 POSTERS function to the immune system suggests that JAK3 inhibition is a promising therapeutic strategy for the treatment of autoimmune diseases, inflammation and allograft rejection [1]. On the other hand it is not finally proven that solely JAK3 inhibition results in an immunosuppression [2]. Therefore highly selective JAK3 inhibitors are needed to further investigate this question. POS.136 Novel ´butterfly` derivatives of FTY720 reduce symptoms in active experimental autoimmune-induced encephalomyelitis (EAE) in mice Zivkovic, A.1; Imeri, F.2; Fallegger, D.2; Schwalm, S.3; Blankenbach, K.3; Engelhardt, B.4; Meyer zu Heringdorf, D.3; Pfeilschifter, J.3; Huwiler, A.2,3; Stark, H.1 1 Institute of Pharmaceutical and Medicinal Chemistry, Heinrich Heine University, Universitaetsstrasse 1, 40225 Duesseldorf, Germany 2 Institute of Pharmacology, University of Bern, CH-3011 Bern, Switzerland; 3 Pharmazentrum Frankfurt/ZAFES, University Hospital, Goethe University Frankfurt am Main, Germany; 4 Theodor-Kocher Institute, University of Bern, CH-3012 Bern, Switzerland. Ruxolitinib is a JAK-Inhibitor with preference for JAK1/2, which is approved for the treatment of myelofibrosis [3]. In our attempt to create new JAK-inhibitors with an improved selectivity towards JAK3, we used Ruxolitinib as scaffold and made a bioisosteric replacement of the pyrazole ring by a triazole. While the synthesis and derivatization of the pyrazole moiety would be a multi-step procedure, the corresponding triazoles are readily accessible via the copper catalyzed azide-alkyne cycloaddition [4] (CuAAC) and can be synthesized in an almost combinatorial fashion. In this study a library of triazole-containing Ruxolitinib derivatives were synthesized and tested for their ability to inhibit JAK3. A chosen subset of the synthesized inhibitors was also tested in a selectivity assay including JAK1-3 and TYK2 to further investigate the selectivity within the Janus kinase family [5]. References: 1. Cornejo, M. G.; Boggon, T.J.; Mercher, T.: Int. J. Biochem. Cell. Biol. 2009, 41: 2376-2379. 2. Haan, C. et al.: Chem. Biol. 2011, 18(3): 314-323. 3. Mesa, R. A.; Yasothan, U.; Kirkpatrick, P.: Nat. Rev. Drug Discov. 2012, 11: 103-104 . 4. Rostovtsev, V. V. et al.: Angew. Chem. Int. Ed., 2002, 41: 2596-2599. 5. Gehringer, M.; Forster, M.; Laufer, S.A.: ACS Comb. Sci. 2015, 17(1): 5-10. POS.135 Synthesis and Antiparasitic Activity of Aromatic Bisamidines Sauer, B.1; Robaa, D.1; Schmidt, M.1; Pierrot, C.3; Bouchut, A.3; Andrews, K.2; Khalife, J.3; Sippl, W.1 1 Martin-Luther-University Halle-Wittenberg, Wolfgang-Langenbeck-Str.4, 06120 Halle, Germany Tropical Parasitology Lab, Eskitis Institute for Drug Discovery, Don Young Road, Griffith University, Nathan, Queensland, Australia 4111 3 U1019-CNRS UMR 8204, Univ. Lille Nord de France, Institut Pasteur de Lille, 1, Rue du professeur Calmette, 59019 Lille, France 2 Malaria belongs to the most important tropical diseases and is subject of various scientific activities concerning drug development. However, there is a need of new drug scaffolds due to resistance problems. In 1977 the aromatic bisamidine furamidine was initially developed as an antiprotozoal agent against trypanosomes [1]. Subsequently, furamidine and its prodrug pafuramidine (DB289) were subjected to clinical trials for the treatment of the sleeping sickness but liver and kidney toxicity led to their discontinuation[2]. Despite this fact the potency against Plasmodium falciparum (IC50: 15.5 nM) [3] identifies furamidine as a promising lead scaffold. The present work aimed for the synthesis of a compound series based on the furamidine core structure. A set of furamidine based bisamidines was prepared introducing modifications on the furan core structure as well as on the aromatic amidine moiety. Subsequently the toxicity of derived compounds against Plasmodium falciparum and different human cell lines was determined ex vivo uncovering structureactivity relationships. References: 1. Das, B. P.; Boykin, D. W.: J. Med. Chem. 1977, 20: 531–536. 2. Wenzler, T. et al.: Antimicrob. Agents Chemother. 2009, 53: 4185–92. 3. Soeiro, M. N. et al.: Expert Opin. Investig. Drugs 2005, 14: 957–972. 164 • DPhG Annual Meeting 2015 Conference Book The sphingosine-1-phosphate (S1P) receptor subtype 1 (S1P1) is a novel therapeutic target for the treatment of multiple sclerosis. FTY720 (fingolimod) is the first functional modulator of S1P1 that is approved for oral treatment of relapsing-remitting multiple sclerosis. In this study we have developed two novel derivatives of FTY720, ST-968 and ST-1071, having an oxazolo-oxazole structure, named as butterfly motive, which proved to be active in intact cells without the necessity of prodrug activation. A straightforward synthesis route for the butterfly derivatives has been developed [1,2]. Both compounds triggered S1P1 and S1P3, but not S1P2, receptor internalisation as a result of receptor activation when using a GFP-coupled receptor constructs transfected into HEK293 cells. Furthermore, in the human endothelial cell line EA.hy 926, ST-968and ST-1071-triggered within minutes a rapid and dose-dependent activation of the classical p42/p44-MAPK cascade which was blocked by pertussis toxin and the S1P1/S1P3 antagonist VPC23019, but not by the S1P2 receptor antagonist JTE-013, suggesting the involvement of a Go/i protein and the S1P1 or S1P3 receptor subtypes. Functionally, ST-968 and ST-1071 acted similar to FTY720 to abrogate S1P-triggered chemotaxis of various immune cells including mouse splenocytes, mouse T cells and of the human monocytic cell line U937. Additionally, endothelial cell permeability, which was enhanced by the pro-inflammatory stimuli TNFα and LPS, was normalized in a dosedependent manner by either S1P, FTY720, ST-968 or ST-1071. All compounds also reduced TNFα-induced ICAM-1 and VCAM-1 mRNA expression, but restored TNFα-mediated downregulation of PECAM-1 mRNA expression. In the in vivo setting, application of ST-968 or ST1071 to mice at a single dose of 1mg/kg resulted in a significant reduction of blood lymphocytes by 70% after 24h. The Multiple Sclerosis (MS) mouse model on active experimental autoimmune encephalomyelitis (EAE) showed that both compounds significantly reduced the clinical symptoms of EAE comparable to that of FTY720 either by prophylactic or therapeutic treatment. References: 1. Stark, H. et al.: PCT WO 2012 131096 A1 (publ. date: October 4, 2012). 2. Zivkovic, A.; Stark, H.: Tetrahedron. Lett. 2010, 51: 3769-3771. POS.137 Antiseizure properties of chlorophenoxy aminoalkyl derivatives Łażewska, D.1; Kuder, K.1; Stark, H.2; Kieć-Kononowicz, K.1 1 Department of Technology and Biotechnology of Drugs, Jagiellonian University Medical College, ul. Medyczna 9, 30-688 Krakow, Poland 2 Institute of Pharmaceutical and Medicinal Chemistry, University Düsseldorf, Universitätsstraße 1, 40225 Düsseldorf Germany , Epilepsy is the most common brain disorder and affects about 1% of the world human population. Despite the availability of many various antiepileptic drugs, about 30% of all seizures are resistant to these drugs. Therefore there is still strong need for new anticonvulsants with higher level of potency and lower levels of toxicity. This work is a part of our research for potential anticonvulsant agents among histamine H3 receptor ligands. A series of chlorophenoxy aminoalkyl derivatives were evaluated for anticonvulsant activity and neurotoxicity according to the standard protocols within Anticonvulsant Screening Program at the NIH/NINDS Bethesda (USA). Pharmacological tests included maximal electroshock (MES) and subcutaneous pentetrazole seizure threshold (scMet) assays as well as neurotoxicity (TOX) evaluation in mice and/or rats after intraperitoneal (i.p.) administration and/or in rats after oral (p.o.) administration. For some DRUG DESIGN/MEDICINAL CHEMISTRY compounds good protection against MES induced seizures was observed. However neurotoxicity was also detected. The most promising results were obtained for 4-chlorophenoxyhexyl derivatives. Acknowledgments: This project was financed by a grant from the National Science Center based on decision No DEC-2011/02/A/NZ4/00031. of (auto)inflammatory diseases [1], cancer [2] and neurodegenerative diseases [3]. Although, many p38α MAP kinase inhibitors have been developed, there is still an urgent need to generate successful clinical candidates with improved in vivo efficacies and reduced side effects [4]. We recently published series of dibenzosuberone and dibenzooxepinone inhibitors with outstanding activity in biological systems and low ATP competitiveness, combined with excellent selectivity against p38α MAP kinase [5,6]. POS.138 Characterization of the HPA-1 polymorphism by MD simulations and FRET measurements Pagani, G.1; Ventura Pereira, J. P.2; Homeyer, N.1; Stoldt, V. R.2; Scharf, R. E.2; Gohlke, H.1 1 Dept. of Pharmaceutical and Medicinal Chemistry, Heinrich Heine University, Universitätsstr. 1, 40225 Düsseldorf, Germany 2 Dept. of Experimental and Clinical Hemostasis, Hemotherapy, and Transfusion Medicine, Heinrich Heine University Medical Center, Moorenstr. 5, 40225 Düsseldorf, Germany The human platelet integrin αIIbβ3 mediates platelet adhesion and aggregation; it is essential for hemostasis but can also foster thrombus formation [1]. αIIbβ3 is polymorphic; its β3 subunit carries the human platelet antigen (HPA)-1. The HPA-1 polymorphism of αIIbβ3 arises from a leucine-to-proline mutation resulting in HPA-1a (Leu33) or its variant isoform HPA-1b (Pro33). This mutation is clinically relevant, since patients with coronary artery disease, who carry the HPA-1b allele, experience their myocardial infarction 5.2 years earlier than HPA-1a/1a patients [2]. HPA-1b is also known as the prothrombotic variant of αIIbβ3, since it was shown to activate platelets enhancing their adhesion and thrombus formation as a consequence of increased classical outside-in signaling. However, the underlying mechanism by which the mutation contributes to the prothrombotic properties of the variant integrin has remained elusive so far. Integrins exist in two main and mutually exclusive conformations: the bent, closed form and the unbent, open structure. Local and global structural rearrangements are required in changing the closed to the open form, thereby leading to integrin activation. In the present study, a combined strategy, integrating large-scale all-atom molecular dynamics (MD) simulations with FRET measurements, was used to characterize the consequences of the Leu33→Pro33 exchange on the structural dynamics of αIIbβ3 at an atomic level. MD simulations of the two αIIbβ3 variants starting from the closed conformation of in total 3 µs length were carried out. For FRET, cyan or yellow fluorescent proteins had been cloned to the C-termini of the αIIb or β3 subunits and expressed on HEK293 cells. Comparative analyses of the MD trajectories revealed that the Pro33Leu mutation gives rise to a local instability that propagates and gradually affects the entire structure, thus leading to the system being globally less stable. FRET measurements confirmed the conformational changes observed in our MD simulations and occurring in the cytoplasmic tails upon integrin activation. MD results supported by experimental findings reveal how a single-point mutation located more than 90 Å away from any ligand binding site in αIIbβ3 can allosterically influence the fine-tuned conformational equilibrium of this protein. References: 1. Kunicki, T.J.; Newman, P.J.: Blood, 1992, 80: 1386-1404. 2. Zotz, R.B. et al.: J. Thromb. Haemost. 2005, 3:1522-1593. POS.139 p38α MAP kinase inhibitors with excellent enzyme and whole blood activity - enhancement of binding interactions between hydrophobic region II and deep pocket Wentsch, H. K.1; Walter, N.1; Mayer-Wrangowski, S. C.2; Rauh, D.2; Laufer, S. A.1 1 Institute of Pharmacy, Department of Pharmaceutical and Medicinal Chemistry, Eberhard Karls Universitaet Tuebingen, Auf der Morgenstelle 8, 72076 Tuebingen, Germany 2 Faculty of Chemistry and Chemical Biology, Technische Universitaet Dortmund, Otto-HahnStrasse 6, 44227 Dortmund, Germany The fundamental role of p38 mitogen-activated protein kinases (MAP kinases) in the biosynthesis of proinflammatory cytokines like Il-1β and TNFα underlines their importance as therapeutic targets for the treatment Figure: Binding mode of compound 1 in p38α MAP kinase (PDB code: 3UVQ) Our lead compound 1 (Figure) showed an excellent IC50 value (IC50 = 1 nM) with respect to p38α MAP kinase but unfortunately a moderate inhibitor activity in a whole blood TNFα release assay (IC50 = 280 nM). One reason might be a short target residence time due to a high ATP concentration and competitive inhibition mechanism. Based on the X-ray structure (PDB code: 3UVQ) [6] we synthesized compounds which form the same interactions to the enzyme and can moreover enhance the interactions between hydrophobic region II and the deep pocket of the enzyme. Both are valid strategies to improve the shortcomings mentioned above. Furthermore we pursued the strategy of a parallel synthesis of a dibenzooxepinone and a dibenzosuberone scaffold both to compare their affinity to the enzyme and their activity in whole blood tests as well as to avoid potential metabolism issues of the dibenzosuberone scaffold. Finally we synthesized a wide range of dibenzepinones following the intentional binding mode and could improve the IC50 value on the isolated enzyme down to the picomolar range and IC50 values in whole blood tests below 50 nM. References: 1. Player, M. R.: Curr. Top Med. Chem. 2009, 9: 598. 2. Tsai, C. J.; Nussinov, R. Semin. Cancer Biol. 2013, 23: 235-42. 3. Anton, R. et al.: PLoS One 2014, 9: e95641.54. 4. Zhang, J. et al.: T. in Pharm. Science 2007, 28: 286-295. 5. Baur, B. et al.: J. Med. Chem. 2013, 56: 8561-8578. 6. Fischer, S. et al.: J. Med. Chem. 2013, 56: 241-53. POS.140 Synthesis of a modified deuterated ω-Acyl Ceramide [EOS] Species and the Impact to the Nanostructure of Stratum Corneum Lipid Model Membranes Sonnenberger, S.1; Eichner, A.1; Lange, S.1; Langner, A.1; Neubert, R. H. H.1; Dobner, B.1 Institute of Pharmacy, Martin-Luther-University Halle-Wittenberg, Wolfgang-Langenbeck-Str. 4, 06120 Halle (Saale), Germany 1 The mammalian skin covers an area of approximately 10% of the body and is, with approximately 10% of the body mass, the largest organ of human being. Most important for the organism is the skin´s barrier function. The skin is divided into three main layers: the subcutis, as the innermost layer, followed by the dermis and epidermis, as the outer layer. The epidermis is additionally divided into further sublayers, in which the Stratum corneum (SC), the outermost layer, plays the crucial role for the skin barrier properties. The SC consists of flattened and keratin-filled dead cells (corneocytes) which are embedded into a multilamellar lipid matrix. The matrix consists of cholesterol (CHOL) and its derivatives such as cholesterol sulfate (ChS), free fatty acids (FFA) and ceramides (CER). As the main fraction of the SC lipids, ceramides play a fundamental role in organisation of the lipid matrix. The ceramides are structurally very heterogeneous. In general, they consist of a fatty acid bonded to the amino group of different sphingoid bases. The fatty acid can exhibit hydroxyl group in alpha or in omega position, whereas the omega hydroxyl can be esterfied to another fatty acid, mainly linoleic acid. The omega hydroxyl fatty acid is, with a chain length about 30 carbon atoms, significantly longer than the non- or alpha-hydroxyl fatty acids with chain length of about 18-26 carbon atoms. Due to this unusual chain length, this ceramide (CER[EOS]) is mainly contributed to the SC´s barrier function. Therefore it is necessary to receive a detailed insight into the molecular arrangement of the lipid matrix organisation. Via neutron DPhG Annual Meeting 2015 Conference Book • 165 POSTERS scattering experiments it is possible to get structural and dynamical information of biological membranes. Neutrons interact with the atomic nucleus and penetrate deep into the membrane. Furthermore, it is possible to distinguish between different isotopes, i.e. hydrogen and deuterium, because of their varying neutron scattering lengths. However, using extracted native lipids, it is difficult to get detailed information because of their complex composition. So, model membranes, based on the native lipid composition were prepared. In the present work we describe the synthetic approaches of a modified deuterated CER[EOS] species (Figure). The derivate consist of sphingosine, 30-hydroxytriacontanoic acid and 10-methylhexadecanoic acid instead of the native bonded linoleic acid. Earlier works already showed, that such a modified ceramide is comparable in its phase behaviour to the native CER[EOS] [1]. Via deuterium labelling, conclusions can be made about the ceramide´s molecular arrangement within the model membrane. For this purpose, 10-D3methylhexadecanoic acid was prepared, coupled with triacontan-1,30diol and after oxidation coupled with sphingosine. First results of neutron scattering experiments show that for the labelled fatty acid varying arrangements are possible. Figure: deuterated methyl branched Ceramide EOS derivative Acknowledgments: We would like to thank Helmholtz-Zentrum-Berlin (HZB, Berlin, Germany) for the allocation of beam time for the neutron scattering investigations. Further we thank Evonik Industries AG (Essen, Germany) for provision of ceramide [AP] C-18. This work was supported by grants of the Deutsche Forschungsgemeinschaft (DFG). References: 1. Engelbrecht et al.: Soft Matter 2011, 7(19): 8998-9011. omega-hydroxy acid can be esterified (EO), e.g. with linoleic acid. The chain length of the acids and the sphingoid bases varies mainly between C18 and C26. Figure: deuterated Ceramide [NP] and [NS] To investigate the arrangement and the composition of the different ceramide species in the lipid matrix, neutron scattering and LC/ESI-MS are commonly used methods [1-3]. Therefore, specifically labelled ceramides with a well-defined structure are needed. For our experiments we used ceramides with a chain length of 18 carbon atoms in the sphingoid base and changed the length of the fatty acid (18, 22 and 24 carbon atoms). In this work we will describe a way to synthesise deuterium labelled non-hydroxy fatty acids at the end of the alkyl chain via the use of LiAlD4. Furthermore, we tried to optimise the coupling of the fatty acid with the sphingoid base and therefore tested different coupling reagents. These results will be additionally presented here. Acknowledgments: We would like to thank Evonik Industries AG (Essen, Germany) and the Deutsche Forschungsgemeinschaft (DFG) for the financial support. References: 1. Sahle, F. F. et al.: Journal of Pharmaceutical and Biomedical Analysis 2012, 60: 7-13. 2. Engelbrecht, T. et al.: Soft Matter 2011, 7: 8998-9011. 3. Schröter, A. et al.: Biophysical Journal 2009, 97: 1104-1114. POS.141 Design and optimization of N-benzyl benzamides: A novel fused scaffold for orally available dual sEH/PPARγ modulators for treatment of metabolic syndrome Proschak, E. 1 1 Institute of Pharmaceutical Chemistry, Goethe-University Frankfurt, Max-von-Laue-Strasse 9, D-60438 Frankfurt a. M., Germany. For abstract see Short Poster Lectures SPL.008 page 105. POS.142 Synthesis of specific deuterated ceramide species [NP] and [NS] with different chain lengths Lange, S.1; Sonnenberger, S.1; Eichner, A.1; Langner, A.1; Neubert, R. H. H.1; Dobner, B.1 1 Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Wolfgang-Langenbeck-Str. 4, 06120 Halle (Saale), Germany The stratum corneum (SC), as the outermost layer of human skin, plays an important role in the homeostasis of mammalians (e.g. regulation of water and the thermal balance). Normally, phospholipids are the main lipid part of biologically membranes, which have to be penetrable for a bearing transport process through them. In contrast to the lipid matrix of the SC, where they are totally lacking, phospholipids are known to keep the membranes fluidity. Compared to that, the ceramides are highly discussed to play a key role to the rigidity of the SC barrier function and further to protecting functions of the skin. For this reason, there is a high interest in the detailed structure of the lipid layers, ceramides are arranging. Next to free fatty acids, cholesterol and its salts, ceramides are the main ingredients of the SC lipid mixture, as they are a group of structurally very heterogeneous sphingolipids. They consists of a long-chain fatty acid (alpha-hydroxy (A), non-hydroxy (N) or omega-hydroxy (O)), which is bond to a sphingoid base (sphingosine (S), dihydrosphingosine (DS), phytosphingosine (P) or 6-hydroxysphingosine (H)). Furthermore, an 166 • DPhG Annual Meeting 2015 Conference Book POS.143 Determining the molecular consequences of clinically relevant glutamine synthetase mutations Frieg, B.1; Görg, B.2; Homeyer, N.1; Keitel, V.2; Häussinger, D.2; Gohlke, H.1 1 Institute for Pharmaceutical and Medicinal Chemistry, Heinrich-Heine-University, Universitätsstr. 1, 40225 Düsseldorf, Germany 2 Clinic for Gastroenterology, Hepatology, and Infectious Diseases, Heinrich-Heine-University, Moorenstr. 5, 40225 Düsseldorf, Germany Glutamine synthetase (GS) catalyzes the ligation of ammonia and glutamate to glutamine under the use of ATP and, thus, essential for nitrogen metabolism [1,2]. Loss of hepatic GS activity has been linked to serious clinical conditions [3]. In particular, two mutations of human GS (R324C and R341C) were connected to congenital glutamine deficiency with severe brain malformations resulting in neonatal death [4]. In a single case known to date, another GS mutation (R324S) was identified in a neurologically compromised patient [5]. However, the underlying molecular mechanisms of GS deactivation by these mutations have not been understood yet. By molecular dynamics simulations, free energy calculations, and rigidity analyses we found that all three mutations influence ATP binding, the first step of GS glutamine formation cycle. In the case of the R324S and R324C mutations, we found a loss of direct salt-bridge interactions with the substrate ATP that cause a deterioration of GS catalytic activity. Remarkably, for the R324S mutant, we observed water-mediated interactions with ATP that reduce this effect and may explain the suggested higher GS residual activity [6]. The R341C mutation destabilizes residue R340 that is important for ATP binding. Furthermore, we predicted the R341C mutant to result in a significant destabilization of helix H8, which should hamper glutamate binding. In order to provide evidence for this, we introduced an additional GS variant through alanine mutagenesis of amino acids interacting with R341, mimicking the loss of interactions in the R341C mutant. After GS overexpression in HEK293 cells, dot-blot analyses revealed that the structural stability of H8 was impaired in the case of the newly introduced GS mutant. This results in a loss of masking of the epitope in the glutamate binding pocket for a DRUG DESIGN/MEDICINAL CHEMISTRY monoclonal anti-GS antibody by L-methionine-S-sulfoximine; in contrast, cells transfected with wild type GS did show the masking. Our analyses reveal complex molecular effects underlying GS deactivation in three clinically relevant mutants. Furthermore, since there is currently no adequate therapy available [6] to treat a glutamine deficiency caused by the R324S mutant, our findings could stimulate the development of ATP binding-enhancing molecules by which the R324S mutant can be “repaired”. Acknowledgments: We are grateful to the ‘‘Zentrum für Informations und Medientechnologie’’ (ZIM) at the Heinrich Heine University for computational support. This work was supported by the Deutsche Forschungsgemeinschaft through the Collaborative Research Center SFB 974 (‘‘Communication and Systems Relevance during Liver Damage and Regeneration’’, Düsseldorf). References: 1. Häussinger, D.: Eur. J. Biochem. 1983, 133(2): 269-275. 2. Häussinger, D.: Biochem. J. 1990, 267(2): 281-290. 3. Qvartskhava, N. et al.: P. Natl. Acad. Sci. USA 2015, 112(17): 5521-5526. 4. Häberle, J. et al.: New. Engl. J. Med. 2005, 353(18): 1926-1933. 5. Häberle, J. et al.: Mol. Genet. Metab. 2011, 103(1): 89-91. 6. Häberle, J. et al.: Orphanet J. Rare. Dis. 2012, 7(48): 1-10. analgesic flupirtine, which on the one hand is a less active opener of potassium channels but on the other hand a clinically valuable drug. However, in some patients even the use of the drug flupirtine rises some concerns related to drug-induced liver injury, most probably because of metabolic toxification and the formation of reactive metabolites. This poses the question whether there is a pattern of cellular reactivity and/or toxicity related to physicochemical properties, i.e. a hierarchy of activity associated with reversible or irreversible covalent interactions with KV7.2/3 channels or other proteins. In order to investigate structureactivity-relationships for this target we synthesized multiple merged structures with the goal of identifying essential moieties of the active molecules aiming for agents with clean toxicity profile and retained activity of the ICA lead compound. The synthesised structures involve carbamate-derivatives of ICA-027243 as well as amide analogs of flupirtine with shifted positions of the pyridine-nitrogen atom. As recent results by our group showed a potential correlation between the oxidizability and the activity of flupirtine and its derivatives [2], the developed compounds are to be tested by cyclic voltammetry. To verify the postulated association the anodic peak potentials are compared to the activities in further studies. POS.144 TopModel: A multi-template meta-approach to Homology Modeling Mulnaes, D.1; Gohlke, H.1 1 Mathematisch-Naturwissenschaftliche Fakultät, Institut für Pharmazeutische und Medizinische Chemie, Heinrich-Heine-Universität, 40225 Düsseldorf, Germany Knowledge of a protein structure is essential to understand its function [1], evolution [2], dynamics [3], stability [4], interactions [5,6] and for datadriven protein- or drug-design [7]. Experimental structure determination rates however are far exceeded by that of next-generation sequencing, resulting in less than 1/1000th of proteins having a known structure. Computational structure prediction seeks to alleviate this problem, and many different methods were developed in the last decade alone. The CASP experiments [8] are widely considered the golden standard for structure prediction benchmarks and have shown the value of consensus- and meta-methods that utilize complementary algorithms. We present a multiple-template meta-approach to structure prediction that combines a broad range of state-of-the-art methods to provide a versatile work-flow and toolbox for structure prediction. Our automated evaluation shows an accuracy competitive with state-of-the-art automated procedures and enables easy re-evaluation/parametrization on any dataset. We anticipate that future integration of new methods will increase the accuracy, sensitivity and applicability of our consensus methods, and that our workflow can be used to evaluate threading software and software for multiple sequence alignments in a consistent manner based on the quality of models produced by the generated alignments. References: 1. Amato, G. et al.: ACS Med. Chem. Lett. 2011, 2(6): 481–484. 2. Lemmerhirt, C. J. et al.: ChemMedChem 2014, 10(2): 368–379. POS.146 Acknowledgments: We thank the Ministry of Innovation, Science and Research of NRW and HHU Düsseldorf for scholarships within the CLIB-Graduate Cluster Industrial Biotechnology. We are grateful to our coauthors in refs. 1 & 5 for fruitful collaborations. Development of a combinatorial chemistry strategy for the synthesis of DNA-encoded libraries References: Škopić, K. M.1; Bugain, O.1; Kalliokoski, T.2; Brunschweiger, A.1 1. Widderich, N. et al.: J. Mol. Biol. 2014, 426(3): 586-600. 2. Prieto, A .I. et al.: Structure 2013, 21(9): 1690-1697. 3. McCammon, J. A.; Gelin, B. R.; Karplus, M.: Nature 1977, 267(1): 585-590. 4. Rathi, P.C.; Hoeffken; H. W.; Gohlke, H.: J. Chem. Inf. Model. 2014, 54(2): 355-361. 5. Gohlke, H. et al.: J. Chem. Inf. Model. 2013, 53(10): 2493-2498. 6. Janin, J.: Protein Science 2005, 14(2): 278-283. 7. Moult, J. et al.: Proteins 2014, 82(S2): 1-6. POS.145 Synthesis and characterization of potassium channel openers with N-benzyl-aminopyridine moiety Bock, C.1; Lemmerhirt, C.1; Link, A.1 1 Institute of Pharmacy, Ernst-Moritz-Arndt-University Greifswald, Friedrich-Ludwig-Jahn-Str. 17, 17489 Greifswald, Germany The small molecule ICA-027243 is a very potent modulator of KV7.2/3 channels but its reactivity-related toxicity in a rat model impedes therapeutic usage [1]. The structure bears some similarities with the 1 Faculty of Chemistry and Chemical Biology, Technische Universität Dortmund, Otto-HahnStraße 6, 44227 Dortmund, Germany 2 Lead Discovery Center GmbH, Otto-Hahn-Straße 15, 44227 Dortmund, Germany The identification of small molecule ligands for proteins of interest is a crucial first step in small molecule probe or drug development programs. The screening of large, pooled DNA-encoded small molecule libraries (DELs) by a conceptually simple selection assay represents a validated technology for the target-based identification of bioactive compounds [1]. We synthesize DNA-encoded screening libraries on “privileged scaffolds“ such as the pyrazolopyrimidine 1. The scaffolds display a functionalization pattern allowing for coupling of these compounds to 5´aminolinker modified DNA and subsequent combinatorial substitution by amide synthesis and Cu(I)-catalyzed alkyne-azide cycloaddition. We selected a set of 150 carboxylic acid and a set of 110 azide building blocks with the help of chemoinformatics in order to optimize diversity of the library (Tanimoto fingerprint analysis), and to control the calculated physicochemical properties of the 16.500-membered library. The aim of the present study was to develop a procedure for parallel, combinatorial synthesis of the library. Whereas 100 of the 110 azides showed excellent conversion (> 90%) to the target triazoles, the coupling efficiencies of the carboxylic acids were very variable. Thus, we decided to couple scaffold DPhG Annual Meeting 2015 Conference Book • 167 POSTERS 1 to 5’-amino-linker modified DNA on solid support, remove the Fmoc group, and introduce the carboxylic acid building blocks in parallel manner on solid support. The DNA-conjugates were then purified by RPHPLC, encoded by T4 ligation, and immobilized on DEAE (diethylaminoethanol) sepharose, an ion exchange resin with excellent DNA bind and release properties [2]. We then introduced the azide building blocks by Cu(I)-catalyzed cycloaddition and were able to extensively wash the resin with EDTA solution to remove copper contaminants prior DNA release. In summary, we have developed an efficient and high-yielding protocol for the combinatorial synthesis of DNA-encoded screening libraries. a) Figure: Development of an encoding strategy for small molecules based on DNA ligation and of the selection assay with encoded model substances and their target proteins. References: 1. Manocci, L. et al.: Chem. Commun. 2011, 47: 12747-12753. 2. Buller F. et al.: Bioconjugate Chem. 2010, 9(21): 1571–1580. POS.148 From stem cell screening towards small molecule tools for in vivo heart regeneration b) Längle, D.1; Flötgen, D.1; Duburs, G.2; Strohmann, C.1; Koch, O.1; Werner, T.3; Hirt, M.3; Schade, D.1 1 Department of Chemistry & Chemical Biology, TU Dortmund, Otto-Hahn-Straße 6, D-44227 Dortmund, Germany 2 Latvian Institute of Organic Synthesis, Aizkraukles 21, Riga LV-1006, Latvia 3 Institute of Experimental & Clinical Pharmacology and Toxicology, University Hospital Hamburg-Eppendorf, Martinistr. 52, D-20246 Hamburg, Germany Figure: Evaluation of combinatorial synthesis strategies to a kinase-directed DEL. References: 1. Buller, F. et al.: Bioconjugate Chem. 2010, 21(8): 1571-1580. 2. Halpin, D. R. et al.: PLoS Biology 2004, 2(7): 1031-1038. POS.147 Encoding small molecule libraries with DNA - and development of a selection assay for DNA-encoded small molecule libraries Jung, K.1; Bugain, O.1; Brunschweiger, A.1 1 Faculty of Chemistry and Chemical Biology, Technische Universität Dortmund, Otto-HahnStraße 6, 44227 Dortmund, Germany The screening of large, pooled DNA-encoded small molecule libraries (DELs) represents a promising technology for the discovery of small molecule ligands at target proteins [1,2] Protein ligands can be identified from these libraries by selection. We use double strand (ds)DNA to encode our small molecule libraries. dsDNA is a rather rigid rod-like structure less likely forming secondary structures that might interfere with the selection assay. We developed an efficient encoding strategy based on T4 DNA ligation in order to record the chemical steps employed in library synthesis. Tetramer overhangs gave high ligation yields, also in two consecutive ligations that are required to encode two building blocks, whereas longer overhangs yielded multiple side products in our hands. To our astonishment a PEG4-linker used as spacer between the small molecule and the DNA inhibited the ligation reaction under standard conditions (16 °C; 16h). To overcome this effect different conditions (temperature, incubation-time, lengths of overhangs, additives to the buffer) were tested. We found out that efficient ligation can be achieved by ligating DNA at 20 °C, for 16h and adding PEG6000 to the ligation buffer. We used the optimized encoding strategy to synthesize small molecule-dsDNA conjugates that we employ to establish a selection assay protocol: desthiobiotin (1), binding to streptavidin; 4sulfamoylbenzoic acid (2), binding to carbonic anhydrase IX; and N1-(2aminophenyl)-N4-terephthalamide (3) binding to HDAC 1. Each DNA has a unique primer region so that for development of a selection assay for DNA-encoded small molecule libraries we can assess conveniently various selection assay conditions by PCR, such as different incubation times, number of washing cycles, different temperatures, ionic strengths of the wash buffer. The newly established selection assay will be used to identify bioactive small molecules from in-house synthesized DNAencoded small molecule libraries. 168 • DPhG Annual Meeting 2015 Conference Book Background: The development of small molecules that control stem cell fate is of tremendous interest for various regenerative medicine applications as it opens up the druggable space [1]. From a high-content screen of 17,000 compounds in mESCs we discovered in the past a novel class of TGF-β inhibitors in the context of cardiac differentiation [2]. A specific subclass of 1,4-dihydropyridines (DHPs) stimulated cardiomyogenesis from murine and human ESCs in stages when uncommitted mesoderm specifies towards a cardiac fate [2,3]. Objective: Since TGF-β is also involved in cardiac remodeling and fibrosis, we propose a dual mode of action for these DHPs as regenerative agents. However, in order to demonstrate proof-of-concept in animal models later in development, the compounds require hit-to-lead optimization. Here, a translational case study illustrates earliest steps in a preclinical drug development campaign for phenotypic screeningderived small molecules. Results: We describe the multidisciplinary medicinal chemistry workflow from ‘screening hit’ to in vivo-suitable pharmacologic tool candidates. Ligand-based (quantitative) structure-activity relationships (SARs), X-ray crystal structure analysis, pathway selectivity and in vitropharmacokinetic profiling provided a solid basis for the development of selective, potent and drug-like lead candidates [4]. Key obstacles that typically limit in vivo applicability, such as poor compound solubility and stability, could be addressed. Moreover, we demonstrated efficacy in an engineered heart tissue (EHT, from neonatal rat cardiomyocytes) as a model of hypertrophy and fibrosis. Additionally, utilizing our ligand-based pharmacophore model, a virtual screening approach gave rise to new chemotypes of TGF-β inhibitors (= scaffold hopping). Conclusions: Systematic, medicinal chemistry-driven efforts led to highly attractive small molecules as novel in vivo pharmacology tool candidates to study heart regeneration and remodeling after myocardial infarction. References: 1. Längle, D. et al.: ACS Chem. Biol. 2014, 9(1): 57-71. 2. Willems, E. et al.: Cell Stem Cell 2012, 11(2): 242-252. 3. Schade, D. et al.: J. Med. Chem. 2012, 55(22): 9946-9957. 4. Längle, D. et al.: Eur. J. Med. Chem. 2015, 95: 246-266. DRUG DESIGN/MEDICINAL CHEMISTRY POS.149 3,4-Bis(indol-3-yl)cyclobut-3-ene-1,2-diones as novel potential antimalarial drugs Lande, D. H.1; Nasereddin, A.2; Dzikowski, R.2; Preu, L.1; Grünefeld, J.1; Kunick, C.1 1 Institut für Medizinische und Pharmazeutische Chemie, Technische Universität Braunschweig, Beethovenstraße 55, 38106 Braunschweig, Germany 2 Department of Microbiology and Molecular Genetics, IMRIC, Hebrew University, Hadassah Medical School, P.O. Box 12272, Jerusalem 91220, Israel Owing to the restricted arsenal of antimalarial drugs and the increasing resistance of the parasite, malaria caused by protozoal parasites of the genus Plasmodium remains one of the most deadly infectious diseases of humans [1]. Annually, more than half a million people of the global population, particularly young children and pregnant women in tropical African countries, are killed by this disease [2]. The urgent need for developing novel potential antimalarial drugs is therefore obvious. Herein we report the syntheses and characterization of 3,4-bis(indol-3yl)cyclobut-3-ene-1,2-diones and their activity profiles against erythrocytic stages of P. falciparum. Furthermore, molecular docking studies in a homology model of PfGSK3, a putative intracellular target for the new compound class, were conducted to explain the observed structure activity relationships. Acknowledgments: This project was funded by the German Federal Ministry of Education and Research (BMBF BioDisc 7; 13GW0024). References: 1. Murray, C. J. L. et al.: J.-Lancet 2012, 379(9814): 413–431. 2. WHO, World Malaria Report 2014. POS.150 Synthesis of functionalized “privileged” scaffolds for DNAencoded libraries Bugain, O.1; Klika Škopić, M.1; Brunschweiger, A.1 1 Faculty of Chemistry and Chemical Biology, Technische Universität Dortmund, Otto-HahnStraße 6, 44227 Dortmund, Germany The screening of large, pooled DNA-encoded small molecule libraries (DELs) represents a validated technology for the target-based discovery of bioactive compounds [1]. DELs are composed of small organic molecules covalently coupled to DNA sequences serving as PCRamplifiable identification bar codes. Small molecule protein ligands can be identified from these libraries by selection. Our aim is to generate DNA-encoded compound libraries based on “privileged scaffolds“ [2]. These scaffolds are overrepresented among bioactive small molecules and therefore attractive core structures for library synthesis. We required the scaffolds to be endowed with a functionalization pattern allowing for coupling to 5´-aminolinker modified DNA and subsequent combinatorial substitution by amide synthesis and alkyne-azide cycloaddition. We designed synthesis strategies that yielded the xanthine (1), pyrazolopyrimidine (2), and benzodiazepine (3, 4, 5) ring systems displaying a carboxylic acid, an Fmoc-protected amine, and a terminal alkyne. Currently, we are using these five compounds to synthesize DNA-encoded libraries by split-and-pool combinatorial synthesis. Each scaffold will yield a final pooled DNA-encoded library encompassing 16,500 compounds. POS.151 Alkoxyurea-based HDAC Inhibitors with quinoline cap groups possess improved activity against chemoresistant cancer cells Stenzel, K.1; Hamacher, A.1; Hansen, F. K.1; Kassack, M. U.1; Kurz, T.1 1 Institut für Pharmazeutische und Medizinische Chemie, Heinrich Heine Universität, Universitätsstrasse 1, 40225 Düsseldorf, Germany Cancer research remains to be one of greatest challenges of our time. Histone deacetylases (HDACs) are linked with different types of cancer and HDAC inhibitors (HDACi) modulate the expression of genes associated with cell cycle, cell differentiation and in addition induce apoptosis in many tumor cells via cell death pathways. Many HDACi demonstrate pronounced cytotoxicity against tumor cells and weaker toxicity on normal cells [1]. Currently four HDACi have been approved for cancer therapy by the FDA. Variation of epigenetic regulation is one of the known causes of abnormal gene expression in a malignant tumor resistant to chemotherapy. Experimental data suggest that the combination of HDACi with established anticancer drugs (e.g. cisplatin) provides synergistic effects in the treatment of hematological and solid tumors, probably generated through HDACi-mediated increased accessibility of DNA [2,3,4]. Isoform-selective HDACi could provide a more effective chemotherapy compared to pan-inhibitors, with fewer side effects and more precise therapeutic properties [5]. Starting from LMK235, a HDACi with HDAC4 and 5 preference [6], we reasoned that the enlargement of the cap group and the connecting unit should lead to a novel type of HDACi with HDAC6 preference. The isoform profiling of the prototype compound 1a (LMK214) confirmed our hypothesis and revealed a pronounced preference for HDAC6 and no inhibition of HDAC4 up to a concentration of 10 µM. A microwave-assisted protocol allowed the systematic variation of the cap moiety. In addition, selected alternative zinc-binding groups (ZBG) were introduced instead of the hydroxamic acid. The synthesized novel inhibitors are characterized by a substituted quinoline or naphthalenyl cap group and an alkoxyurea connecting unit linker region. Figure: Strategy and target compounds The biological evaluation of the target compounds included cellular HDAC and MTT assays on the HNSCC cell line Cal27 and the ovarian cancer cell line A2780 and their cisplatin resistant sublines. Some of the compounds showed similar or improved effects compared to vorinostat on inhibition of cellular HDACs in a whole-cell HDAC assay. Furthermore, incubation with these compounds 48h prior to cisplatin resulted in an enhancement of cisplatin-induced cytotoxicity in the cisplatin resistant sublines. References: 1. Witt, O. et al.: Cancer Lett. 2009, 277: 8–21. 2. Ong, P.-S. et al.: Int. J. Oncol. 2012, 40: 1705–1713. 3. Eckstein, N. et al.: J. Biol. Chem. 2008, 283: 739–750. 4. Mueller, H. et al.: J. Biomol. Screen. 2004, 9: 506–515. 5. Balasubramanian, S.; Verner, E.; Buggy, J. J.: Cancer Lett. 2009, 280: 211–221. 6. Marek, L. et al.: J. Med. Chem. 2013, 56: 427–436. Figure: “Privileged” scaffolds functionalized to furnish DNA-encoded libraries References: 1. Stockwell B. R.: Nature 2004, 432(7019): 846-854. 2. Evans, B. et al.: J. Med. Chem. 1988, 31(12): 2235-2246. DPhG Annual Meeting 2015 Conference Book • 169 POSTERS POS.152 Fighting the Human African Trypanosomiasis – Optimizing an aza-analogous Furamidine by a new prodrug principle elastase, bovine chymotrypsin, bovine trypsin, human factor Xa, human thrombin, human cathepsin L and B) was evaluated by different activity assays and analyzed e.g. with the slow-binding equation. To verify the ability of the probe to target HLE, it was incubated with HLE, subjected to SDS-PAGE and evaluated by in-gel fluorescence detection. Polosek, P.1; Girreser, U.1; Clement, B.1 Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy, CAU Kiel, Gutenbergstraße 76, 24118 Kiel, Germany 1 The Human African Trypanosomiasis (HAT) is known since the beginning of the 20th Century with the first documented case in 1902 [1]. In the 1960’s the HAT seemed to be controlled, but at the beginning of the 21st Century the reported cases increased significantly due to the negligence of the disease [1]. The infection can be separated into 2 stages: the first stage named haemo-lymphatic stage is possible to cure by an early treatment. The second stage called the meningo-encephalitic stage remains hard to treat mainly due to insufficient therapy options (strong adverse effects, difficult application) with an inadequate therapy referring to adverse effects and difficult appliance [1]. The pipeline of available substances to cure the patients is very small and since the admission of nifurtimox and eflornithine as a combination for the second stage of the HAT no substance has been approved for around 20 years [1]. In 2001 pafuramidine maleate (DB 289) began its clinical trials as the first oral applicable substance against HAT [1]. Unfortunately the substance missed its approval due to liver and kidney toxicity in an additional phase Ι trial [1]. Figure: Inactivation of a serine protease (Ser-OH) by the fluorescence-labeled sulfonyloxyphthalimide derivate. Results: A novel probe was designed, synthesized and its spectroscopic properties were determined. The probe was shown to be selective for HLE with an kobs/[I] value of 8360 M-1s-1 and an IC50 value of 12 nM (determined from a reaction time of 1 hour), whereas the other enzymes were inhibited with IC50 values of more the 1 µM. The analysis of the ingel fluorescence revealed a higher detection limit than the common coomassie staining. Conclusion: We obtained a selective and sensitive probe for HLE which is expected to be a powerful tool to study the activity of this enzyme in cell or tissue samples. References: 1. Korkmaz, B. et al.: Pharmacol. Rev. 2010, 62(4): 726-759. 2. Kasperkiewicz, P. et al.: Proc. Natl. Acad. Sci. USA 2014, 111(7): 2518-2523. 3. Neumann, U.; Gütschow, M.: J. Biol. Chem. 1994, 269(34): 21561-21567. Figure: Pentamidine, Furamidine (DB 75), Aza-analogue of Furamidine (DB 829) Based on that structure many new substances were synthesized heading to aza-analogous structures with promising potentiality. So far only substances including the pyridine ring with the nitrogen atom in o-position to the furane ring revealed remarkable results in the cure rate of mice and monkeys in the first stage as well as in the second stage of the HAT [2]. However, the results only appeared by intramuscular application and did not occur by the oral treatment. Therefore it is necessary to exchange the prodrug principle used so far. The prodrug principle reported in the published articles is an O-methylation of the amidoxime [2]. Other prodrug principles are improving the activation of the prodrug, the water solubility and the oral bioavailability [3]. Due to this information a new prodrug of an aza-analogue pentamidine derivation has been developed for oral application. The chosen prodrug-principle is an amidoximeester with a very good solubility which is currently being tested in a mouse model for the second stage of the HAT. References: 1. WHO Technical Report Series 984 2013 2. Thuita et al.: PLoS Negl. Trop. Dis. 2015, 9(2):e0003409. doi: 10.1371/journal.pntd.0003409 3. Kotthaus et al.: ChemMedChem 2011, 6: 2233–2242. doi: 10.1002/cmdc.201100422 POS.154 Synthesis of fluorine-containing PDE10A-Inhibitors as potential Ligands for Positron Emission Tomography (PET) Franz, L.1; Scheunemann, M.2 ; Wagner, S.2; Lang, M.1; Brust, P.2; Briel, D.1 Institute of Pharmacy, University of Leipzig, Bruederstraße 34, 04103 Leipzig, Germany Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Permoserstraße 15, 04318 Leipzig, Germany 1 2 Helmholtz-Zentrum Phosphodiesterases (PDE´s) are second messenger hydrolysing enzymes and important regulators of signal transduction mediated by these molecules. PDE10A, a cAMP and cGMP sensitive hydrolase, is primarily expressed in the striatum and was identified as drug target for the therapy of diverse disorders in the central nervous system (CNS) [1] like schizophrenia or chorea huntington [2]. Recently, 1-arylimidazo[1,5a]quinoxalines have been reported to be potent and selective inhibitors of PDE10A [3]. In terms of a potential use as 18F-labelled PET imaging agent new substituted derivatives were synthesized. It has been shown that the methoxy substituted inhibitors are prone to metabolic oxidation, which leads to a loss of inhibitory potency or ability to cross the blood brain barrier [3,4]. POS.153 A Sulfonyloxyphthalimide Derivate as an Activity-based Probe for Elastase Schulz-Fincke, A. C.1; Tikhomirov, A. S.1,2; Gütschow, M.1 1 Pharmaceutical Institute, Pharmaceutical Chemistry I, University of Bonn, An der Immenburg 4, D-53121 Bonn, Germany 2 Present address: Gause Institute of New Antibiotics, 11B. Pirogovskaya Street, Moscow 119021, Russia Objective: Aberrant expression of human leucocyte elastase (HLE) can lead to degradation of endogenous proteins and extracellular matrix components. Such a scenario is associated with pulmonary diseases and lung cancer [1]. Fluorescence-labeled probes are expected to be very useful for investigating the cell biology and (patho)physiology of HLE [2]. Materials and Methods: The design of the probe is based on a phthalimide structure as a new reactive warhead. It is expected that the nucleophilic active site serine residue of HLE attacks the phthalimide warhead, following by a ring opening and a subsequent Lossen rearrangement. This reaction leads to a covalent modification of the target HLE (Figure) [3]. A polyethylene glycol-derived linker connects the warhead with coumarin 343 which was introduced to visualize the active enzyme. The inactivation of several proteases (HLE, porcine pancreatic 170 • DPhG Annual Meeting 2015 Conference Book To improve the metabolic stability of inhibitors the methoxy function in position 6 was exchanged by chlorine. In the first synthesis step chlorine was introduced at position 6 by electrophilic aromatic substitution. An electron deficient system was generated in step 2 by oxidation of the amine to a nitro function to allow the nucleophilic aromatic substitution of fluorine by 4-methylimidazole in step 3. Afterwards, the amine was recovered by acidic reduction with elementary iron in step 4 and acetylated in step 5. Cyclisation in step 6 was realized by a BischlerNapieralski reaction. The derivatization of the 1-arylimidazo[1,5a]quinoxaline was focused on position 1 and 8. Finally, the fluoropyridinyl-group was introduced by Suzuki-coupling with the corresponding boronic acid at the brominated positions to afford the mono- or disubstituted pyridinyl derivatives. All compounds were DRUG DESIGN/MEDICINAL CHEMISTRY characterized by high performance liquid chromatography, nuclear magnetic resonance spectroscopy and mass spectrometry. It is expected that the new chlorinated derivatives have the same pharmaceutical effects as their methoxy analogues. A 3D-Diverse Fragment Library for Crystallographic Screening and Drug Discovery Acknowledgments: Thanks are to J. Ortwein for HPLC analysis, Dr. L. Hennig for recording and analysis of NMR data and Dr. J. Preidl for LC-MS analysis. Metz, A.1; Huschmann, F. U.1,2; Schiebel, J.1; Mueller, U.2; Weiss, M. S.2; Heine, A.1; Klebe, G.1 References: 1. Liras, S.; Bell, A. S.: Phosphodiesterases and Their Inhibitors (Wiley-VCH) 2014. 2. Schmidt, C. J. et al.: J. Pharm. Exp. Ther. 2008, 325: 681-690. 3. Malamas, M. et al.: J. Med. Chem. 2011, 54: 7621–7638. 4. Schwan, G.: Synthese und in vitro Metabolismus fluorhaltiger PDE10A-Inhibitoren als potentielle PET-Liganden, 2011, University of Leipzig, Germany. POS.155 Structure characterization of 17β-hydroxysteroid dehydrogenase Type 14 and identification of the first inhibitors Bertoletti, N.1; Braun, F.1; Hartmann, R. W. 2; Möller, G.3; Adamski, J.3; Heine, A.1; Klebe, G.1; Marchais-Oberwinkler, S.1 1 Institute for Pharmaceutical Chemistry, Philipps University Marburg, Marbacher Weg 6, 35037 Marburg, Germany 2 Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), 66123 Saarbrücken, Germany 3 Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany 17β-Hydroxysteroid dehydrogenase type 14 (17β-HSD14) is a recently characterized enzyme [1,2], which is mainly located in the brain, liver and placenta. 17β-HSD14 catalyses the oxidation in vitro of estradiol, 5-androstene-3β, 17β-diol and testosterone to estrone, dehydroepiandrosterone and androstedione, respectively, using NAD+ as cofactor. The functional role of this enzyme remains unclear. Administration of estradiol is efficient in the treatment of neuronal diseases like migraine, schizophrenia or Alzheimer disease but is related to severe side effects. We expect that inhibition of 17β-HSD14 will lead to a local increase of estradiol in the brain, which might be beneficial for the treatment of these neuronal disorders or for the prevention of Alzheimer disease. Potent and selective inhibitors are also useful tools to study the role of enzymes in vivo. The goal of this study was to identify the first inhibitors of 17β-HSD14 and optimized their substitution pattern using the 3D structure of the protein/ligand complex. First of all the pure protein was produced in high amount after heterologous expression in E. coli and the purification protocol was established. Then a competitive enzymatic assay was established using the purified enzyme (fluorescence-based assay, following cofactor fluorescence changes). The most interesting inhibitor identified so far in our laboratory was advanced to crystallographic study. The purified enzyme was crystallized as apoenzyme, holoenzyme with NAD+ and in complex with the most interesting inhibitors. The 3D structures of different complexes were resolved and will be presented. These structures will be useful to get insight in to the active cleft and improve the inhibitor potency by structure-based design. References: 1. Lukacik, P. et al.: Biochem. J. 2007, 402(3): 419–427. 2. Marchais-Oberwinkler, S. et al.: J. Steroid Biochem. Mol. Biol. 2011, 125(1-2): 66–82. POS.156 Institute of Pharmaceutical Chemistry, Philipps-University Marburg, Marbacher Weg 6, 35032 Marburg, Germany 2 Helmholtz-Zentrum Berlin, Institute Soft Matter and Functional Materials, Macromolecular Crystallography (HZB-MX) Electron Storage Ring BESSY II, Albert-Einstein-Str. 15, 12489 Berlin, Germany 1 Ligand binding pockets differ regarding their shape and arrangement of potential interaction sites. Thus, we design a general-purpose 3D-diverse fragment library for crystallographic fragment screening that can address a great variety of binding sites while providing particularly suitable starting points for subsequent drug discovery efforts. To facilitate fragment-to-lead evolution after hit identification, this library must adequately cover the accessible chemical space of fragments including substructures of drug-like ligands. A reasonable coverage of chemical space aims at fully exploiting the fact that already a small selection of appropriate fragments (~103 cpds.) covers a much larger proportion of the overall chemical fragment space (~107 cpds.) than a typical high-throughput screening collection (105–106 cpds.) with respect to the drug-sized chemical space (~1063 cpds., MW < 500 Da). Despite the relatively low affinity of fragments to their target, their high ligand efficiency (ΔGbind. divided by number of nonhydrogen atoms) makes them excellent starting points for drug discovery. In addition, fragments often bypass the strict steric requirements for the binding of larger drug-like ligands, thus leading to high hit rates up to 15%. Customarily, different biophysical pre-screening methods are applied, often serially, to limit the number of fragments to a selection manageable by routine X-ray crystallography. However, a comparative study with our 361-fragment library [1] confirmed a limited overlap amongst prescreening methods [2] that also missed many of direct crystallographic screening hits [1]. In contrast, we strive for an appropriate library design and use state-of-the-art virtual screening to equally increase effectiveness and enrichment at no cost of materials. In conjunction with its increased capacity and level of automation this suggests direct crystallographic screening by crystal soaking of a small and preferably target- or structure-based selection of 102–103 fragments as a gold standard. To complement our established in-house fragment library, we now compile a set of 1000 high-quality fragments with properties particularly suited for crystal soaking, immediate fragment-to-lead evolution, prepared for computer-aided subset selection and fragment-to-lead evolution. This expanded library will be part of the Frag2Xtal service facility for crystallographic fragment screening, which will be available at the semi-automated crystallographic BL14.2 at the BESSY II storage ring of the Helmholtz-Zentrum Berlin [3]. Aiming at a representative coverage of chemical space, all sufficiently available and biophysically suitable fragments (> 250,000 cpds. adopting > 1.4·106 conformational and molecular states) were clustered in groups of 3D-similar compounds. To this end, we calculated > 1012 pairwise molecular similarities based on the 3D overlap of volume and interaction features (charges, hydrogen bond donors/acceptors, aromatic ring, etc.) using the ROCS (rapid overlay of chemical structures) method [4] and used these similarities for a hierarchical clustering with the SPARSEHC algorithm [5] at the Marburg HPC cluster MaRC2. Selecting favorable representative fragments from each cluster will allow covering the available chemical space with fragments that are particularly suited for crystallographic fragment screening and as starting points for drug discovery projects. References: 1. Köster, H. et al.: J. Med. Chem. 2011, 54(22): 7784-7796. 2. Schiebel, J. et al.: ChemMedChem 2015, DOI: 10.1002/cmdc.201500267. 3. Mueller, U. et al.: J. Synchr. Rad. 2012, 19(3): 442-449. 4. FastROCS. OpenEye Scientific Software. Santa Fe, NM, USA, URL: www.eyesopen.com. 5. Nguyen, T.-D.; Schmidt, B.; Kwoh, C.-K.: Procedia Comput. Sci. 2014, 29: 8-19. DPhG Annual Meeting 2015 Conference Book • 171 POSTERS POS.157 Convergent synthesis of a linker-connected fluorescent ebselen-coumarin heterodimer Küppers, J.1; Palus, J.2; Giurg, M.2; Skarżewski, J.2; Gütschow, M.1 1 Pharmaceutical Institute, Pharmaceutical Chemistry I, University of Bonn, An der Immenburg 4, D-53121 Bonn, Germany 2 Department of Organic Chemistry, Wrocław University of Technology (A2), Wyspiańskiego 27, 50-370 Wrocław, Poland Objective: Ebselen (1) is a multifunctional drug with a wide range of pharmacological effects that are predominantly due to its interaction with selenoenzymes, e.g. glutathione peroxidase, thioredoxin reductase [1,2]. Such enzymes play an important role in protecting biomembranes and other cellular components from oxidative stress [2]. Fluorescencelabeled probes containing ebselen can be suitable for further biological and medicinal studies, to profile enzyme activities, identify target enzymes and characterize their functions [3]. Synthesis: The synthetic route starts with the procedure for synthesizing ebselen [4]. Anthranilic acid as the starting material was converted into a diazonium salt, which was treated with disodium diselenide to obtain the 2,2´-diselenobisbenzoate (2). After treating 2 with thionyl chloride, a reaction with an appropriate para-substituted aniline derivative with a protected primary aliphatic amine group was performed to give the first component, compound 3. To obtain the PEG linker for the connection of the two components, the amino group of 2-(2-aminoethoxy)ethanole was first Cbz-protected. Afterwards, the hydroxyl group was alkylated with tert.-butyl bromoacetate followed by a Cbz deprotection. The resulting primary aliphatic amine was coupled with the second components, the fluorescent coumarin 343 (4). This was synthesized by submitting 8hydroxyjulolidine-9-carboxaldehyde to a Knoevenagel condensation. In the final steps, both the ebselen derivative 3 and the PEG-coumarin 343 building block were deprotected and the desired probe 5 was assembled. Application: The new probe will be provided to biochemical and pharmacological studies. performed in two steps. First, a clique-detection algorithm is computed which only relies on the information about the pseudocenters that were stored in the database. Second, the cavities are aligned and a score is calculated based on the overlap of the surfaces of the two aligned cavities [2]. There are three applications for binding site comparisons. First the design of novel ligands by retrieving similar ligands (scaffold-hopping) or ligand portions (bioisosteric replacements) from similar pockets. Second the prediction of protein function based on the analysis of active sites by matching well-known sites onto other proteins of unknown properties. The main use, however, is to study ligand selectivity by identifying possible off-targets. In order to characterise similarities of binding sites on a more local level, we developed a strategy to split the cavities into smaller subpockets. Therefore the information about the bound ligands are exploited to generate such subpockets automatically. This is done by decomposing the original ligand into fragments. Every pseudocenter, which is coincides within a predefined radius about the fragment, is selected to define the subpocket. After this process is accomplished, each individual subpocket is used as a separate query for a Cavbase retrieval. In order to validate this approach a well-known example of cross-reactivity has been studied: the approved drug Celecoxib binds to COX-II and CA-II [3]. The result was as follows: (I) an improved similarity score ranked both proteins prominently, (II) among the retrieved proteins another known target of Celecoxib (the PDK-1 protein) was ranked high, (III) and the binding to a previously unknown target was predicted, PPARγ, which was subsequently confirmed experimentally, with an EC50 ≈ 30 µM. This demonstrates that we are able to predict cross-reactivities. Acknowledgments: We thank Prof. Dr. Schubter-Zsilavecz and Christina Lamers (GoetheUniversity Frankfurt, Institute of Pharmaceutical Chemistry) for measuring the PPARγ assay and the Center for Synthetic Microbiology, Marburg (SYNMIKRO) for financial support. References: 1. Hendlich, M.; Rippmann, F.; Barnickel, G.: J. Mol. Graphics Modell. 1997, 15(6): 359–363. 2. Schmitt, S.; Kuhn, D.; Klebe, G.: J. Mol. Biol. 2002, 323(2): 387–406. 3. Weber, A. et al.: J. Med. Chem. 2004, 47(3): 550–557. References: 1. Stoyanovsky, D. A. et al.: ACS Med. Chem. Lett. 2014, 5(12): 1304-1307. 2. Azad, G. K.; Tomar, R. S.: Mol. Biol. Rep. 2014, 41(8): 4865-79. 3. Xu, K. et al.: Chem. Sci. 2013, 4(3): 1079-1086. 4. Palus, J.; Młochowski, J.; Juchniewicz, L.: Polish J. Chem. 1998, 72(8): 1931-1936 POS.159 Development of novel prodrugs for the nitric oxide (NO) precursor Nω-hydroxy-L-arginine (NOHA): A carbohydratebased approach Litty, F.-A.1; Gudd, J.1; Girreser, U.1; Clement, B.1; Schade, D.2 Pharmaceutical Institute, Christian-Albrechts University, Gutenbergstr. 76, 24118 Kiel, Germany 2 Department of Chemistry & Chemical Biology, TU Dortmund, Otto-Hahn-Str. 6, 44227 Dortmund, Germany 1 POS.158 Cross-reactivity prediction using binding site fragmentation Rickmeyer, T.1; Klebe, G.1 Institute of Pharmaceutical Chemistry, Philipps-University Marburg, Marbacher Weg 6-10, 35032 Marburg, Germany 1 Cavbase is a widely used method to quantify the similarity of binding sites across proteins. Surface-exposed binding sites of proteins are detected by the Ligsite algorithm [1]. By use of the aforementioned algorithm a grid-based approach is applied to the protein to identify buried regions on the surface. Subsequently, the physicochemical properties of the cavityflanking residues are encoded into 3D descriptors, the so-called pseudocenters. There are seven different types of pseudocenters covering the following properties: H-bond donor, acceptor, mixed donor/acceptor, aliphatic, pi-pi interactions, aromatic, and metal ion. Following the described procedure, binding sites are stored in a database. The estimation of the similarity between binding sites is 172 • DPhG Annual Meeting 2015 Conference Book Background: Nω-Hydroxy-L-arginin (NOHA) is formed during the physiological generation of nitric oxide (NO) from L-arginine, a process catalysed by nitric oxide synthases (NOSs). In addition to being the most potent substrate for NOSs, NOHA releases NO only at that site where it is ultimately needed as opposed to other NO donors (e.g., nitrates). Furthermore, NOHA is a potent inhibitor of arginases whose overexpression and activity is closely linked to atherosclerotic changes. Hence, NOHA is a promising NO modulator for the treatment of cardiovascular diseases [1,2]. Objective: Since NOHA exhibits a poor chemical and metabolic stability we have devised prodrug concepts for the problematic Nhydroxyguanidine moiety that successfully overcame these liabilities [3]. N- and O-substitution greatly increased the hydrolytic and oxidative stability of the hydroxyguanidine moiety, but in vitro assays revealed that N-carbamates are not bioactivated. Building on our findings that Oacetalic substitution with carbohydrates (R1) furnished surprisingly stable DRUG DESIGN/MEDICINAL CHEMISTRY prodrugs, we next aimed at designing these without carbamate groups. Here, we present both a novel synthetic route and a cell-based assay to quantify cytosolic NO release by automated fluorescence microscopy. Results: We successfully established an orthogonal protecting group (PG) strategy that enabled installing the desired carbohydrate-based prodrug groups and subsequent, gentle deprotection of PGs. Introduction of an allyloxycarbonyl (Alloc)-PG at Nω-position was the key concept for this carbamate-free approach. PG cleavage was performed palladiumcatalyzed without compromising the labile glyosidic bond. Moreover, esterification of the α-carboxylic moiety (R3) was possible to increase the lipophilicity. In addition, we established a 96-well-plate, cell-based fluorescence microscopic imaging assay to demonstrate cellular uptake of the new NOHA prodrugs along with in-cell bioactivation resulting in the release of NO. Briefly, NO was quantified in murine macrophages using DAF-FM-DA after stimulation with LPS. Conclusions: In conclusion, we were able to synthesize a great number of diverse carbamate-free and carbohydrate-based NOHA prodrugs employing an orthogonal PG concept. By fluorescence-based imaging we could show that NOHA as well as most prodrugs entered the cells and increased intracellular NO levels. In fact, we observed a tendency that higher substituted glucose-based prodrugs released NO most efficiently. Moreover, we could show a high chemical stability of the new acetalic NOHA prodrugs further underlining their drug candidate qualities and utility as novel cardiovascular agents. References: 1. Schade, D.; Kotthaus J.; Clement, B.: Pharmacol. Ther. 2010, 126: 279-300. 2. Moncada, S.; Higgs E. A.: Br. J. Pharmacol. 2006, 147: 193-201. 3. Schade, D. et al.: Org. Biomol. Chem. 2011, 9: 5249-5259. POS.160 Canthin-4-one alkaloids and related compounds as a novel chemotype of DYRK1A inhibitors Tremmel, T.1; Meijer, L.2; Bracher, F.1 1 Department 2 ManRos of Pharmacy – Center for Drug Research, LMU Munich, 81377 Munich, Germany Therapeutics, Perharidy Research Center, 29680 Roscoff, Bretagne, France The canthin-4-one alkaloids represent a small class of natural products, with tuboflavine, isotuboflavine and norisotuboflavine being the only known constituents. In previous investigations we worked out the first efficient approach to this ring system, including the total syntheses of tuboflavine and norisotuboflavine, starting from appropriate 1-acyl-βcarbolines and amide acetals [1]. In subsequent investigations we found significant antimicrobial activities of the parent canthin-4-one and 5substituted as well as annulated analogues [2]. This prompted us to work out a novel approach to the canthin-4-one ring system which allows for the introduction of variable substituents at C-6. Readily accessible 1-bromo-β-carboline [3] is converted to 1-ethynyl-βcarboline under Sonogashira conditions and subsequent 1,3-dipolar cycloaddition with nitrile oxides provides 1-isoxazolyl-β-carbolines. Reductive ring cleavage leads to intermediate primary enaminoketones, followed by ring closure to give 6-substituted canthin-4-ones in high yields. Further substitution in position 5 can be achieved easily by iodination followed by palladium catalyzed cross-coupling reactions [4]. Selected canthin-4-ones and some of the synthetic precursors showed interesting inhibitory properties on the protein kinase DYRK1A. POS.161 Biological evaluation of a dual-target PQS-quorum sensing inhibitor that hinders biofilm formation in Pseudomonas aeruginosa Thomann, A.1,+; de Mello Martins, A. G.1,+; Brengel, C.1; Weidel, E.1; Plaza, A.2; Börger, C.3; Empting, M.1; Hartmann, R. W.1,3,4,* Helmholtz Institut for Pharmaceutical Research Saarland, Department of Drug Development and Optimization, Campus C23, 66123 Saarbrücken, Germany 2 Helmholtz Institut for Pharmaceutical Research Saarland, Department of Microbial Natural Products, Campus C23, 66123 Saarbrücken, Germany 3 PharmBioTec GmbH, Sciencepark 1, 66123 Saarbrücken, Germany 4 Pharmazeutische und Medizinische Chemie, Campus C2 , Universität des Saarlandes, 66123 3 Saarbrücken, Germany * Corresponding author; + Authors equally contributed to this work 1 Emergence of Pseudomonas aeruginosa (PA) as a leading cause of nosocomial infections and morbidity in immunocompromised patients has consolidated it in the race for novel antimicrobial compounds [1]. PA infections are notoriously difficult to eradicate due to intrinsic resistance to a variety of available antibiotics. Its distinguished ability to form biofilms amplifies resistance and promotes immune response evasion. The PA quorum-sensing (QS) is a sophisticated network of genome-wide regulation triggered in response to population density. A major component is the pseudomonas quinolone signal (PQS) QS system that regulates the production of several non-vital virulence and biofilm-related determinants [1]. Hence, this QS circuitry is an attractive target for antivirulence agents with lowered resistance development potential. We have developed a dual-inhibitor compound (cmpd. VI) of low molecular weight and high solubility that targets PQS transcriptional regulator (PqsR) and PqsD, a key enzyme in the biosynthesis of PQS-QS signal molecules HHQ and PQS [2,3]. In vitro, cmpd. VI markedly reduced virulence factor production and biofilm formation accompanied by a diminished content of extracellular matrix components. Additionally, co-administration with ciprofloxacin increased susceptibility of PA14 to antibiotic treatment under biofilm conditions. Finally, disruption of pathogenicity mechanisms was also assessed in vivo, with significantly increased survival of challenged larvae in a Galleria mellonella infection model [4]. Favourable physicochemical properties and effects on virulence/biofilm establish a promising starting point for further optimization. In particular, the ability to address two targets of the PQS autoinduction cycle at the same time with a single compound holds great promise in achieving enhanced, synergistic cellular effects while potentially lowering rates of resistance development. References: 1. Tashiro, Y. et al.: Microbes Environ. 2013, 28(1): 13-24. 2. Storz, M. P. et al.: J. Am. Chem. Soc. 2012, 134(39): 16143-16146. 3. Lu, C. et al.: Chem. Biol. 2012, 19(3): 381-390. 4. Hill, L. et al.: Int. J. Antimicrob. Agents. 2014, 43(3): 254-261. POS.162 Allosteric signaling deduced from in silico perturbations on biomacromolecules Pfleger, C.1; Gohlke, H.1 References: 1. Puzik, A.; Bracher, F.: J. Heterocyclic Chem. 2009, 46: 770-773. 2. Puzik, A.; Bracher, F.: Lett. Org. Chem. 2013, 10: 568-572. 3. Bracher, F.; Hildebrand, D.: Tetrahedron 1994, 50: 12329-12336. 4. Tremmel, T.; Bracher, F.: Tetrahedron, 2015, 71: 4640-4646. Institute of Pharmaceutical and Medicinal Chemistry, Heinrich Heine-University, Universitätsstr. 1, 40225 Düsseldorf, Germany 1 Understanding allosteric regulation in biomacromolecules is of great interest for current drug design efforts. Conventionally, models that explain allostery involve a conformational change upon binding of an DPhG Annual Meeting 2015 Conference Book • 173 POSTERS allosteric effector molecule [1,2]. Over the last decades, the view of allostery has been extended to cover the role of dynamics [3]. This type of dynamically dominated allostery can occur in the absence of conformational changes, and thus, is difficult to deduce from static X-ray structures alone. Here, allosteric effectors partially rigidify the biomacromolecule, which then percolates through a network of interactions to distant sites [4]. With respect to this dynamically dominated mechanism, it should be possible to predict putative residues that form an allosteric regulation pathway for signaling between distant sites. For studying dynamically dominated allostery, mechanical perturbation free energies were deduced from rigidity analyses using a constraint network representation of biomacromolecules [5]. The CNA (Constraint Network Analysis) software package has been developed by us to infer biologically relevant characteristics from rigidity analyses, which are essential for understanding the relationship between biomacromolecular structure, (thermos-)stability, and function [6]. Here, CNA was used to analyse changes in stability upon in silico perturbations on ensembles of network topologies, which was quantified by the mechanical perturbation free energies. In order to identify pathways for allosteric signaling, a perresidue decomposition of the mechanical perturbation free energy was performed to predict putative residues that mediate the allosteric signaling between distant sites. We applied CNA on conformational ensembles of PTP1B, LFA-1, and β2AR. Ensembles were derived from MD trajectories of 300 ns length starting from the effector bound state. Each ensemble was perturbed in silico by removing the allosteric effector. We identified multiple residues, which have pronounced mechanical perturbation free energies. Remarkably, these residues form continuous pathways for allosteric signaling connecting residues in the orthosteric site or functionally important residues for allosteric regulation. Being able to identify allosteric signaling and to predict the mechanism of allosteric regulation provides an excellent tool for studying systems with yet unknown allosteric mechanisms. Furthermore, direct assessment of flexibility and rigidity characteristics requires no filtering of spurious correlations, which is needed in the analysis of correlated motions. impermeable cell wall. An analysis of calculated polar surface area and logP indicated an influence of these properties on the minimum inhibitory concentration. Based on computational molecular design, compounds with improved psychochemical properties were designed and synthesized. In fact, these compounds showed an improved activity on mycobacterial growth which underlines the assumption regarding optimized properties. Permeability measurements using a PAMPA-assay also proved an increased permeability and showed the usability as a model system for mycobacterial cell wall permeability. We will present and discuss the design and the results of our improved compounds. In addition, further experiments towards a detailed biochemical characterisation are in progress, and toxicity experiments and tests on infected macrophages are being planned. References: 1. World Health Organization Tuberculosis Programme; http://who.int/tb/ 2. Jaeger, T.; Flohé, L.: Biofactors. 2006; 27: 109-120. 3. Koch, O. et al.: J. Med. Chem. 2013; 56(12): 4849-4859. 4. Koch, O.: A New Player in the Fight against Tuberculosis: Thioredoxin Reductase Inhibitors with High Bioactivity on M. tuberculosis. Talk at “Frontiers in Medicinal Chemistry”, Saarbrücken, Germany, 2011. POS.164 Looking for multi-target acting xanthine derivatives Karcz, T.1; Drabczyńska, A.1; Olejarz, A.1; Köse, M.2; Hinz, S.2; DorozPłonka, A.1; Kubas, B.1; Müller, C. E. E.2; Kieć-Kononowicz, K.1 Department of Technology and Biotechnology of Drugs, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland 2 PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical Chemistry I, An der Immenburg 4, D-53121 Bonn, Germany 1 Bioactive M. tuberculosis thioredoxin reductase inhibitors: An update Triple active compounds that inhibit the monoamine oxidase B (MAO-B), block A2A adenosine receptors (A2A AR) and A1 adenosine receptors (A1 AR) are expected to show synergistic effects in the treatment of Parkinson’s disease or Alzheimer’s disease. Such drugs may simultaneously influence several symptoms of these complex neurodegenerative diseases [1-3]. Previously we described a group of annelated xanthine derivatives which showed good A2A AR and/or A1 AR antagonistic properties [4,5]. Here we focused on compounds with multifunctional activity. New tricyclic xanthine derivatives containing a dopamine moiety were designed and synthesized. In addition analogs lacking the third annelated ring were investigated. All compounds were evaluated in radioligand binding assays for their affinity towards the adenosine receptor subtypes: A1, A2A, A2B and A3. Additionally, compounds were tested at the human MAO-B. In summary, potent dual-target-directed A1/A2A adenosine receptor antagonists which showed MAO-B inhibitory properties in the submicromolar range were identified. Bering, L. 1; Pretzel, J.1; Rudo, A.1; Schneefeld, M.2; Hölscher, C.3; Bange, F.-C.2; Koch, O. 1 Acknowledgments: Partially supported by Polish National Science Center funds, granted on the basis of decision; No: UMO-2012/04/M/NZ4/00219 and COST Action CM1207 (GLISTEN), financed by EU-FP7. References: 1. Monod, J. et al.: J. Mol. Biol. 1965, 12(1): 88-118. 2. Koshland, D. E. et al.: Biochemistry 1966, 5(1): 365-385. 3. Cooper, A.; Dryden, D. T.: Eur. Biophys. J. 1984, 11(2): 103-109. 4. Gohlke, H. et al.: Proteins 2004, 56(2): 322-337. 5. Pfleger, C.: Ensemble-Based Framework for Analyzing Dynamically Dominated Allostery (PhD thesis), Düsseldorf, 2014. 6. Pfleger, C. et al.: J. Chem. Inf. Model. 2013, 53(4): 1007-1015. POS.163 1 TU Dortmund University, Otto-Hahn-Straße 6, 44221 Dortmund, Germany School Hannover, Carl-Neuberg-Straße 1, 30625 Hannover, Germany Center Borstel, Parkallee 22, 23845 Borstel, Germany 2 Medical 3 Research The resurgence of tuberculosis, caused primarily by Mycobacterium tuberculosis, and the appearance of multi-drug resistant and extensively drug resistant M. t. strains strengthen the need for new drugs with alternative modes of action [1]. The interaction between the mycobacterial thioredoxin reductase (TrxR) and its substrate thioredoxin (Trx) is a promising new drug target for the treatment of tuberculosis, since M.t. lacks the common glutathione system and the M.t. TrxR shows a substantial difference in sequence, mechanism and structure to human TrxRs. The TrxR is part of the M.t. antioxidant system that reduces hydroperoxides, contributes to ribonucleotide reduction, and thus guarantees the survival within macrophages [2]. Although the target mechanism is a protein-protein interaction, the first known inhibitors with different scaffolds could be identified using an exhaustive highthroughput docking based on the available TrxR X-ray structures [3]. By means of structure-based design, the activity of the most promising candidate could be increased to an IC50 up to the low nanomolar range that also showed an influence on the growth of M. tuberculosis [4]. In order to further improve the bioactivity of the promising compounds we focused on optimizing the physicochemical properties that are important for permeability, since M. tuberculosis shows an unusual thick and 174 • DPhG Annual Meeting 2015 Conference Book References: 1. Van der Schyf, C.J.: Expert. Rev. Clin. Pharmacol. 2011, 4(3): 293-298. 2. Petzer, J. P. et al.: Neurotherapeutics. 2009, 6(1): 141-151. 3. Pisani, L. et al.: Curr. Med. Chem. 2011, 18(30): 4568-4587. 4. Drabczyńska, A. et al.: Purinergic Signal. 2013, 9(3): 395-414. 5. Drabczyńska, A. et al.: Eur. J. Med. Chem. 2011, 46(9): 3590-3607. POS.165 New P2Y11 ligands: Synthesis and biological activity Müller, D.1; Hongwiset, D.2; Kassack, M. U.1 1 Institut für Pharmazeutische und Medizinische Chemie, Heinrich-Heine Universität Düsseldorf, Universitätsstr. 1, 40225 Düsseldorf 2 Faculty of Pharmacy, Chiang-Mai University, Suthep Rd, Mueang Chiang Mai District, Chiang Mai 50200 The P2Y11 receptor is a GPCR belonging to the nucleotide family of purinergic receptors. During the last decade several pathophysiological processes could be identified, in which the P2Y11 receptor participates. P2Y11 is associated with narcolepsy or different inflammatory diseases like acute myocardial infarction [2,3]. Furthermore, interactions of this DRUG DESIGN/MEDICINAL CHEMISTRY receptor subtype can influence cell proliferation or sensitivity of tumor cell lines against cytotoxic agents [4]. Despite its participation in a variety of pathophysiological processes, only few potent agonists and antagonists of the P2Y11 receptor are known. In addition, there is a lack of selective ligands [5]. To drive the development of potent and selective P2Y11ligands further, our research focused on new non-nucleotide P2Y11 receptor ligands. We synthesized a series of symmetrical and asymmetrical arylureas, starting from commercial and non-commercial building blocks in a four-step sequence. The biological activity of the compounds was determined with a functional calcium assay using 1321N1 cells recombinantly expressing P2Y11 receptors. POS.167 Development and preclinical characterization of partial farnesoid X receptor agonists for metabolic disorders Merk, D. 1 1 Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt, Germany; For abstract see Short Poster Lectures SPL.009 page 106. POS.168 Design, synthesis and preliminary evaluation of chemical tools for target identification of novel TGFβ signaling inhibitors Flötgen, D.1; Längle, D.1; Carrillo García, C.1; Schade, D.1 References: 1 Department of Chemistry & Chemical Biology, TU Dortmund, Otto-Hahn-Str. 6, D-44227 Dortmund, Germany 1. Abbracchio, M. P. et al.: Pharmacol. Rev. 2006, 58(3): 281-341. 2. Mignot, E. et al.: Nat. Genet. 2011, 43(1): 66-71. 3. Erlinge, D. et al.: European Heart Journal 2007, 28(1): 13-18. 4. Hu, J. et al.: J. Cell. Biochem. 2011, 112(9): 2257-2265. 5. Jacobson, K. A. et al.: Drug Discov. Today 2010, 15(13-14): 570-578. POS.166 Development of ligands for the cannabinoid-like orphan receptors GPR18 and GPR55 Kieć-Kononowicz, K.1; Rempel, V.2; Karcz, T.1; Schoeder, C.2; DorozPłonka, A.1; Kaleta, M.1; Müller, C. E.2 Department of Technology and Biotechnology of Drugs, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland 2 PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical Chemistry I, An der Immenburg 4, D-53121 Bonn, Germany 1 GPR18 and GPR55 are orphan G protein-coupled receptors (GPCRs) that interact with certain cannabinoid receptors (CBRs) ligands. GPR18 was reported to be activated by Δ9-tetrahydro-cannabinol (THC). GPR55 was found to be activated by cannabinoid receptor antagonists, including the CB1R inverse agonist - rimonabant. GPR18 was found be one of the most highly upregulated receptors in metastatic melanoma [1]. Activation of GPR18 was reported to induce migration of human endometrial cells. Therefore, GPR18 receptor antagonists may be useful novel therapeutics for the treatment of endometriosis and cancer [2]. On the other hand, GPR55, highly expressed in adrenal glands, central nervous system, certain cancer cell lines and immune cells, was postulated to play a role in mediating inflammatory and neuropathic pain, promoting cancer cell migration and proliferation. Thus, GPR55 receptor antagonists may be useful for the treatment of cancer, neuropathic pain [3]. Recently, several bicyclic imidazole-4-one derivatives were discovered as ligands of GPR18 and GPR55 [4] using β-arrestin translocation assays. Selectivity of these compounds with respect to CB1 and CB2 receptors was also evaluated. In the present study, 3-benzyloxybenzylideneimidazo[2,1-b][1,3]thiazin3-ones (group I) and related 3-benzyloxybenzylideneimidazo[2,1b][1,3]thiazepin-3-ones (group II) were investigated for their interaction with GPR18, GPR55 and CBRs. Selective antagonists for GPR18 as well as for GPR55 were found in group I. In contrast, in group II compounds showed higher affinity for CB1 and CB2 receptors. One of the new compounds exhibited agonistic activity at GPR55 indicating that antagonists for this receptor may easily be converted to compounds that can activate the receptor. Acknowledgments: Partially supported by Polish National Science Center funds, granted on the basis of decision; No: UMO-2013/11/B/NZ7/04865 and COST Action CM1207 (GLISTEN), financed by EU-FP7. References: 1. Qin, Y.; et al.: Pigment Cell Melanoma Res. 2010, 24(1): 207-217. 2. Alexander, S.P.: Br. J. Pharmacol. 2012, 165(8): 2411-2413. 3. Shore, D. M.; Reggio, P.H.: Front. Pharmacol. 2015, 6: 69. 4. Rempel, V. et al.: Med. Chem. Commun. 2014, 5: 632-649. Background: Target identification of small molecules derived from phenotypic screens (forward chemical genetics) is a demanding task but also very rewarding as it may reveal unknown (druggable) targets, thus representing a crucial step in drug development [1]. In our case, a subclass of 1,4-dihydropyridines (DHPs) was discovered from a screen for stimulators of cardiogenesis in murine embryonic stem cells. Mechanistic studies revealed a new mode of TGFβ signaling inhibition via proteasomal degradation of the TGFβ type II receptor [2]. Objectives: Since we have established a sound structure-activity relationship (SAR), 3D-QSAR and pharmacophore model, [3, 4] we had a good picture of where to incorporate reactive functionalities for an affinity-based approach. The desired DHP tool compounds should be potent, selective and carry functional groups for photocrosslinking to the unknown target as well as for fluorescence-based detection. Here, we summarize 1) synthetic routes that were explored to furnish a diverse set of these DHP derivatives, 2) bioactivity as TGFβ inhibitors and 3) initial data on their utility as molecular tools to study cellular processes. Results: We chose to incorporate small-sized, aliphatic diazirines for intracellular photocrosslinking and alkynes for click reaction with azidecontaining fluorophores to enable their detection. DHPs with these groups in 2-, 3- and 7-position were successfully synthesized. In this regard, using a small, iodine-containing diazirine as a building block proved particularly valuable. Importantly, most of these DHP derivatives still potently inhibited TGFβ signaling (IC50 = 0.5-2.0 µM). Moreover, >100 conditions were systematically explored to optimize the click reaction of DHP alkynes with FAM-azide in HEK cells for confocal microscopy, allowing the analysis of the subcellular localization of the labelled DHPs (= click products). Conclusions: A diverse set of DHP tool compounds for target identification could be synthesized and were active as TGFβ inhibitors. According to our preliminary cellular data, they can be used to study colocalization of DHPs with TGFβ type II receptor (mCherry fusion protein). Next steps will include their use to ‘fish’ the target protein by photocrosslinking and proteomic analyses, potentially unravelling a novel (maybe universal) regulatory circuitry of receptor dynamics within the TGFβ superfamily of ligands. References: 1. Ziegler, S. et al.: Angew. Chem. Int. Ed. 2013, 52: 2744–2792. 2. Willems, E. et al.: Cell Stem Cell 2012, 11: 242–252. 3. Schade, D. et al.: J. Med. Chem. 2012, 55: 9946–9957. 4. Laengle, D. et al.: Eur. J. Med. Chem. 2015, 95: 249–266. DPhG Annual Meeting 2015 Conference Book • 175 POSTERS POS.169 α5β1 Integrins are Receptors for Bile Acids with a (Nor-)Ursodeoxycholane Scaffold Bonus, M.1; Sommerfeld, A.2; Häussinger, D.2; Gohlke, H.1 1 Institute for Pharmaceutical and Medicinal Chemistry, Heinrich-Heine-University Düsseldorf, Universitätsstr. 1, 40225, Germany 2 Clinic for Gastroenterology, Hepatology and Infectious Diseases, Heinrich-Heine-University Düsseldorf, Moorenstr. 5, 40225, Germany Integrins are ubiquitously expressed cell adhesion receptors and the most prevalent bidirectional signaling molecules on the cell surface. They are involved in osmosensing in the liver [1,2,3] and facilitate the communication between components of the extracellular matrix and cytoplasmic proteins. Upon activation, integrins undergo large-scale conformational changes from a compact, bent state to an open, extended state [4]. A recent study combined immunofluorescence staining (IFS) experiments and molecular dynamics (MD) simulations to identify tauroursodeoxycholic acid (TUDC) as potent agonist of α5β1 integrins in hepatocytes [5]. Activation of α5β1 leads to choleresis by FAK/cSrc/MAPK dependent signaling events [6,7,8]. TUDC-induced integrin activation and subsequent signaling is sensitive to inhibition by the trihydroxylated taurocholic acid (TC), which tightly binds to α5β1 in MD simulations. However, effects of other bile acids on α5β1 integrin activation have not been investigated at the molecular level. Here we report on combined IFS experiments and MD simulations that indicate that α5β1 integrins are not exclusively activated by TUDC. 24nor-ursodeoxycholic acid (norUDCA), a side chain-shortened homologue of UDCA, induces conformational changes in the βA domain of α5β1 that are similar to the one evoked by TUDC, but overall less pronounced. Conformational changes in simulations of integrin ectodomains bound with the taurine conjugate of norUDCA (TnorUDCA) and glycoursodeoxycholic acid (GUDC) were significantly less pronounced. Unconjugated UDCA, similar to the inhibitory TC, only showed insignificant alterations in the structure of the integrin ectodomain and was considered fully inactive. A ranking based on the extent of structural changes observed during the MD simulations correlates with results from IFS experiments on the efficacy of the bile acids. These results indicate that norUDCA activates α5β1 integrins and that MD simulations are able to predict different degrees of bile-acid induced integrin activation. Minor structural changes in the bile acids strongly influence their efficacy. This holds true for a comparison of TUDC (activating) vs. TC (inhibitory) and norUDCA (activating) vs. UDCA (nonactivating). Acknowledgments We are grateful to the ‘‘Zentrum für Informations und Medientechnologie’’ (ZIM) at the Heinrich Heine University for computational support. This work was supported by the Deutsche Forschungsgemeinschaft through the Collaborative Research Center SFB 974 (‘‘Communication and Systems Relevance during Liver Damage and Regeneration’’, Düsseldorf). References: 1. Häussinger, D. et al.: Gastroenterology 2003, 124(5): 1476-1487. 2. vom Dah,l S. et al.: J. Biol. Chem. 2003, 278(29): 27088-27095. 3. Schliess et al.: J. Biol. Chem. 2004, 279(20): 21294-301. 4. Xiong, J. P. et al.: Science 2001, 294(5541): 339–345. 5. Gohlke, H. et al.: Hepatology 2013, 57(3): 1117–1129. 6. Schliess, F. et al.: Gastroenterology 1997, 113(4): 1306–1314. 7. Beuers, U. et al.: Hepatology 2001, 33(5): 1206–1216. 8. Häussinger, D. et al.: Gastroenterology 2003, 124(5): 1476–1487. POS.170 C2-Linked Dimeric Strychnine Analogs as Bivalent Ligands Targeting Glycine Receptors Zlotos, D. P.1; Banoub, M.1; Holzgrabe, U.2; Breitinger, H.-G.; Breitinger, U.1; Villmann, C.3 Dept. of Pharmaceutical Chemistry, The German University in Cairo, New Cairo City, 11835 Cairo, Egypt 2 Institut für Pharmazie and Lebensmittelchemie, Universität Würzburg, Am Hubland, 97074 Würzburg, Germany 3 Institut für Klinische Neurobiologie, Universitätsklinikum Würzburg, Versbacherstr. 5, 97078 Würzburg, Germany 1 Strychnine (1), the major alkaloid from the plant Strychnos nux vomica, exhibits pharmacological activity at several neurotransmitter receptors. Its most pronounced pharmacological action is a strong antagonistic activity at glycine receptors (GlyRs), which are anionic chloride channels composed of five subunits and linked to hyperpolarisation and inhibition of neuronal firing [1-3]. The glycine receptors are one of the major 176 • DPhG Annual Meeting 2015 Conference Book mediators of rapid synaptic inhibition in the mammalian brainstem, spinal cord, and higher brain centers. Dysfunction of the receptors is associated with motor disorders such as hypereflexia, or some forms of spasticity [4]. To date, strychnine is the ligand displaying the highest activity at recombinant and native GlyRs in binding and functional assays [2,3]. Here, we report the synthesis and pharmacological evaluation of the first series of bivalent ligands 4a-d targeting glycine receptors. Designed as strychnine dimers, the target compounds should be able to bind simultaneously to two subunits of the pentameric receptors causing a possibly stronger inhibition than the monomeric strychnine. N N H O N O2N H H H O N H 2. H2SO4 O H H H N H EtOH O O H N H H O H H H H H O 3 N H2OC-(CH2)n-CO2H n = 2,4,6,8 H 2 strychnine (1) CH2Cl2, EDCI HCl N H2N SnCl2 H 1. HNO3 H H N (CH2)n N H O O N H H N O 4a 4b 4c 4d O n=2 n=4 n=6 n=8 H H H O Acknowledgments: Deutscher Akademischer Austauschdienst (DAAD), Bundesministerium für Bildung und Forschung (BMBF) References: 1. Lynch, J.W.: Physiol. Rev. 2004, 84: 1051. 2. Laube, B. et al.: Trends Pharmacol. Sci. 2002, 23: 519. 3. Jensen, A.A.; Kristiansen, U.: Biochem. Pharmacol. 2004, 67: 1789. 4. Breitinger, H.-G.; Becker, C.-M.: Curr. Pharm. Des. 1998, 4(4): 315-34. POS.171 Human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes: Chemically-defined derivation, genetic manipulation and applications in drug development Rathmer, B.1; Carrillo García, C.1; Greber, B.2; Schade, D.1 Department of Chemistry & Chemical Biology, TU Dortmund, Otto-Hahn-Str. 6, D-44227 Dortmund, Germany 2 Max Planck Institute for Molecular Biomedicine, Von-Esmarch-Str. 54, 48149 Münster, Germany 1 The Nobel Prize awarded discovery that somatic cells can be reprogrammed to an induced pluripotent stem cell-like state and the ability to generate cardiomyocytes (CMs) from these iPSCs using directed differentiation methods have provided a fertile ground for many applications. Such applications are not only of therapeutic nature (= regenerative medicine) but also include technologies for the drug discovery process (= safety pharmacology and toxicology) and disease modeling approaches (= disease-in-a-dish) for basic and applied research in cardiac biology [1-3]. Although current differentiation protocols enable the derivation of CMs from human stem cells in high yields under chemically-defined conditions, there are still several obstacles for the above outlined applications. These include the heterogeneous constitution of hiPSCCMs as they typically consist of a mixture of ventricular, atrial and pacemaker cells. Moreover, the level of maturity of hiPSC-CMs is lower than that of native, adult CMs which may limit some applications. Notably, not much is known about the underlying developmental processes and suitable small molecule modulators are lacking [2,3]. Here, we provide an overview on our hiPSC-based approaches to the discovery and characterization of new small molecular modulators of cardiac differentiation and regeneration. These efforts build on using a robust and efficient chemically-defined differentiation protocol that allows us to establish a platform of new high-content assays [4]. In this regard, we are exploring different methods for generating hiPS reporter cell lines harboring fluorescence reporter cassettes (e.g., bacterial artificial chromosome transgenesis) to determine CM yields, constitution and their proliferative capacity. One aim is, for example, the identification of proproliferative small molecules by ‘forward chemical genetics’. This approach would not only allow the expansion of early, immature CMs for DRUG DESIGN/MEDICINAL CHEMISTRY transplantation but also increase our knowledge of mechanistic cues that direct the postnatal loss of CM proliferation. Moreover, we are using the assay platform to study mechanistic aspects of (new) small molecule Wnt/β-Catenin inhibitors as this pathway is a key player within cardiopoietic differentiation. These studies might be useful to enable cell context-specific Wnt inhibition (safety for in vivo applications) and improving the quality of hiPSC-derived CMs. References: 1. Takahashi, K.; Yamanaka, S.: Cell 2006, 126: 663-676. 2. Schade, D.; Hansen, A.; Greber, B.: Drug Target Review 2015, 2(2): 34-38. 3. Schade, D.; Plowright, A. T.: J. Med. Chem. 2015, in press. 4. Zhang, M. et al.: Stem. Cells 2015, 33: 1456–1469. POS.172 The Crystal Structure of the Nisin Resistance Protein Reveals the Underlying Mechanism of Lantibiotic Resistance Khosa, S.1; Frieg, B.2; Mulnaes, D.2; Kleinschrodt, D.3; Hoeppner, A.4; Gohlke, H.2; Smits, H. J. S.1 Institute of Biochemistry, Heinrich Heine University, Universitätsstr. 1, 40225 Düsseldorf, Germany of Pharmaceutical and Medicinal Chemistry, Heinrich Heine University, Universitätsstr. 1, 40225 Düsseldorf, Germany 3 Protein Production Facility, Heinrich Heine University, Universitätsstr. 1, 40225 Düsseldorf, Germany 4 Crystal and X-ray Facility, Heinrich Heine University, Universitätsstr. 1, 40225 Düsseldorf, Germany 1 2 Institute Lantibiotics are potent antimicrobial peptides and often considered as next generation antibiotics [1]. Some pathogenic bacteria, however, express membrane-associated resistance proteins, which proteolytically inactivate lantibiotics [2]. The most prominent member of lantibiotics is nisin, which contains five specific and crucial lanthionine rings that are the hallmark for its activity as well as recognition [3,4]. We report the first three-dimensional structure of the lantibiotic resistance protein superfamily, namely the nisin resistance protein from Streptococcus agalactiae (SaNsr) at 2.2Å, which is active against nisin [5,6]. It contains three domains, a N-terminal helical bundle, a protease cap domain and a protease core domain, which also harbors the highly conserved TASSAEM region. The active center and the binding site of nisin within SaNsr have been characterized via sitedirected mutagenesis and molecular modeling, respectively. Due to the presence of the lanthionine rings, nisin itself is highly unsusceptible to proteolytic degradation. However, SaNsr takes advantage of the methyllanthionine rings present in nisin for substrate recognition and specificity. This structural information would pave way for designing small molecular compounds inhibiting lantibiotic resistance proteins by which the potency of these fascinating small peptides can be fully explored. Acknowledgments: We thank Lutz Schmitt for fruitful discussions, encouragement, support and invaluable advice. We are grateful to Phillip Ellinger for initiating the project and Andre Abts for stimulating discussions, constant support and ideas. We thank Michael Lenders for help with the MALS measurement. We are grateful to the staff of ESRF ID23-2 and ID29 for support during crystal screening and data collection, especially Christoph Mueller-Dieckmann for his enormous patience, assistance and support. We are thankful to Heinrich Heine International Graduate School of Protein Science and Technology (iGRASPseed) for providing a scholarship to S.K. We are grateful to the “Zentrum für Informations- und Medientechnologie” (ZIM) at the HeinrichHeine-University Düsseldorf for providing computational support. References: 1. Hancock, R. E.; Sahl, H.-G.: Nat. Biotechnol. 2006, 24: 1551-1557. 2. Nawrocki, K. L.; Crispell, E. K.; McBride, S. M.: Antibiotics 2014, 3: 461-492. 3. Sahl, H.-G.; Bierbaum, G. Annu. Rev. Microbiol. 1998, 52: 41-79. 4. Wiedemann, I. et al.: J. Biol. Chem. 2001, 276: 1772-1779. 5. Sun, Z. et al.: Antimicrob. Agents Chemother. 2009, 53: 1964-1973. 6. Khosa, S.; AlKhatib, Z.; Smits, S. H.: Biol. Chem. 2013, 394: 1543-1549. DPhG Annual Meeting 2015 Conference Book • 177 POSTERS POS.175 4.9 Inflammation POS.173 BMP2K is a crucial mediator of inflammatory and angiogenic processes in the human endothelium Bischoff, 1 Institute I.1; Dai, B.2; Strödke, B.3; Bracher, F.3; Fürst, R.1 of Pharmaceutical Biology, Goethe-University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany Biology, Center for Drug Research, University of Munich, Butenandstraße 513, 81377 München, Germany 3 Department of Pharmacy - Center for Drug Research, University of Munich, Butenandstraße 7, 81377 München, Germany 2 Pharmaceutical Chronic inflammation is characterized by constant leukocyte infiltration and angiogenesis in the inflamed tissue, which is the case in various diseases such as rheumatoid arthritis or chronic inflammatory bowel diseases. As permanent leukocyte extravasation leads to severe tissue damage, there is a demand for compounds that promote the inhibition of inflammation and angiogenesis and the discovery of new drug targets. The bone morphogenetic protein-2 (BMP-2)-inducible kinase (BMP2K) might be a novel target of such substances. The goal of this study is to elucidate the role of BMP2K in the endothelium and in leukocytes during inflammation and angiogenesis. In this project C81, an inhibitor of BMP2K, as well as RNAi-based gene silencing was applied to inhibit BMP2K and, therefore, to clarify the role of this kinase during inflammatory processes. Different from prechondroblastic cells, the BMP2K seems not to be regulated by BMP-2 in human umbilical vein endothelial cells (HUVECs) (q PCR). In addition, pro-inflammatory stimuli such as LPS or TNFα did not show any effect on BMP2K regulation on the mRNA level. The inhibition of BMP2K by C81 or gene-silencing reduced the migratory capacity of a human microvascular endothelial cell line (HMEC-1). Also the proliferation of HMEC-1 was reduced after C81 treatment with an IC50 of 6.9 µM. The performance of a tube formation assay using Matrigel demonstrated that the inhibition of the BMP2K by C81 significantly impaired the formation of capillary-like structures in a dose-dependent manner. Interestingly, the analysis of signaling molecules in HUVECs that play a crucial role in cell proliferation (ERK, Akt, JNK, p38) revealed that these pathways are not influenced by C81 treatment or genesilencing of BMP2K (Western blot). Rising concentrations of C81 reduced the TNFα-triggered expression of cell adhesion molecules (ICAM-1, VCAM-1, E-selectin) on the surface of HUVECs (flow cytometry) and on mRNA levels (qPCR). Similar results for surface expressions were detected in BMP2K-silenced endothelial cells. Based on these findings a cell adhesion assay using the monocytic cell line THP-1 and C81-treated or BMP2K-silenced HUVECs was performed. THP-1 showed a significantly decreased adhesion to activated HUVECs. Most notably, C81 or knock-down of BMP2K did not lead to the reduction of TNF-induced IκBα degradation or p65 translocation into the nucleus. The results indicate that BMP2K is crucially involved in the proinflammatory and pro-angiogenic activation of human endothelial cells. Furthermore, these data highlight C81 as a promising tool to interrupt BMP2K-mediated signaling events. Further studies are needed to elucidate the precise role of BMP2K during inflammation and angiogenesis and to clarify the regulatory mechanisms that underlie those processes. Regulatory role of 5-Lipoxygenase-activating protein (FLAP) in 5-Lipoxygenase substrate acquisition, turnover and interaction Gerstmeier, J.1; Newcomer, M.2; Werz, O.1; Garscha, U.1 1 Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-SchillerUniversity, 07743 Jena, Germany; 2Department of Biological Sciences, Louisiana State University, Baton Rouge, Louisiana 70803 Leukotriene (LT) biosynthesis from arachidonic acid (AA) is catalyzed by 5-Lipoxygenase (5-LOX), and proceeds via the intermediate 5hydroperoxyeicosatetraenoic acid (5-HPETE) to LTA4. Among all LOXs, solely 5-LOX catalysis from endogenous substrate requires an essential translocation process from the cytosol or perinuclear region to the nuclear membrane-embedded helper-protein 5-LOX-activating protein (FLAP), upon cell activation. Although structures for both FLAP and 5LOX have been solved, the molecular basis for efficient AA transfer from FLAP to 5-LOX remains elusive, and the “corked” cavity of 5-LOX has led to speculation on how AA can enter the active site. We used sitedirected mutagenesis, coupled with 5-LOX product analysis and fluorescence microscopy techniques in stable transfected HEK293 cells [1] to reveal the entry portal for AA. Our results support a model in which AA gains access to the active site through the plugged “FY” cork. Upon 5-LOX translocation and association with the membrane, the two bulky side chains of the “FY” cork swing out for membrane insertion to uncork the active site. This “uncorking” however must be reversible for full cellular 5-LOX activity as a permanently open cavity, mimicked by alanine mutants of both “corking” residues, completely precludes LT biosynthesis. Of great interest, co-expression of FLAP rescues the 5LOX activity of “cork” mutations. The ability of FLAP to restore AA metabolism to an “uncorked” 5-LOX strongly suggests that a physical protein-protein interaction between 5-LOX and FLAP occurs to complete the 5-LOX active site for efficient substrate acquisition and turnover. In particular, LTA4 formation at the expense of 5-HPETE is aided by the “FY” cork. Finally, distinct proof for the true existence of 5-LOX/FLAP complexes was accomplished by proximity ligation assay (PLA). Unlike GFP-based FRET approaches, PLA probes capture the interacting proteins in their native form. Although the “FY” cork itself is not an interaction site with FLAP, both residues are pivotal for complete translocation capacity of 5-LOX and LTA4 formation. Our findings highlight the significance of FLAP in 5-LOX substrate acquisition and conversion, and offer novel perspectives to control of subcellular localization and activity of 5-LOX. References: 1. Gerstmeier, J. et al.: Biochim. Biophys. Acta. 2014, 1840(9): 2961-2969. POS.176 In situ forming gel devices as local depot therapeutic for Rheumatoide Arthritis Mohammadi, M.1; Abebe, D.2; Li, Y.1; Kandil, R.1; Xie, Y.1; Fujiwara, T.2; Merkel, O. M.1 1 Department of Pharmaceutical Sciences, Wayne State University, Detroit, MI 48201 of Chemistry, The University of Memphis, 213 Smith Chemistry Building, Memphis, Tennessee, 38152, USA 2 Department POS.174 Time-resolved in situ assembly of the 5-lipoxygenase / 5lipoxygenase-activating protein complex in primary human leukocytes Garscha, U. 1; Gerstmeier, J. 1; Werz, O. 1 1 Chair of Pharmaceutical/ Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller University Jena, Philosophenweg 14, 07743 Jena, Germany For abstract see Short Poster Lectures SPL.010 page 107. 178 • DPhG Annual Meeting 2015 Conference Book More efficient, novel anti-inflammatory therapies with reduced side effects are needed to treat Rheumatoid arthritis (RA), a chronic and disabling autoimmune condition that affects about 1% of the population in developed countries [1]. Even though a multitude of cell types is involved in RA inflammation, it is agreed upon that macrophages play a central role in the pathophysiology of RA [2]. Locally implantable, targeted, macrophage-specific RNA interference (RNAi)-based therapies could therefore revolutionize RA therapy. INFLAMMATION Three-layered micelles (3LM) entrapping and encapsulating nucleic acids were formed from triblock copolymers of PLLAPEI-PLLA and PLLA-PEG-PLLA in a three-step procedure outlined in upper Figure [3]. Their structure and DNA entrapment in the core were determined by staining DNA with silver nitrate and TEM detection [4]. Hydrodynamic diameters and zeta potentials were measured by dynamic light scattering and laser Doppler anemometry. DNA release in neutral and acidic pH was detected by modified SYBR Gold assays [3]. For targeting of activated macrophages, folic acid (FA) was attached to the PEGchain of a PLLA-PEG diblock affording PLLA-PEG-FA. Subsequently, 3LM were formed with PLLA-PEG-FA in the outer polymer shell. RAW264.7 cells were activated with LPS [5] or left resting. One day after the activation, the cells were treated with targeted and non-targeted 3LM loaded with fluorescently labeled DNA. The uptake of 3LM or PEI/DNA polyplexes was quantified by flow cytometry. Thermoresponsive hydrogels were obtained by stereocomplexing 3LM which contain PLLA-PEG-PLLA in the outer core with PDLA-PEG-PDLA which is shown in lower Figure [6]. The core-corona structure and efficient DNA entrapment in the core were confirmed by TEM. The sizes were found to be less than 200 nm, and the encapsulation efficiency of DNA was optimized based on the ratio of the PEI block in PLLA-PEI-PLLA per DNA [3]. 3LM were stable at neutral pH but released DNA in an acidic environment [3]. 3LM were efficiently targeted to activated macrophages by blending PLLA-PEG-FA and PLLA-PEG-PLLA in the outer layer, while non-targeted micelles or PEI polyplexes were not efficiently taken up. Stereocomplexes of 3LM form hydrogels above their phase transition temperature. References: 1. WHO, Chronic diseases and health promotion in: Chronic rheumatic conditions 2015, Geneva. 2. Gordon, S.;Taylor, P. R.: Nat. Rev. Immunol. 2005, 5: 953-964. 3. Abebe, D. G. et al.: Macromolecular Bioscience 2015, 15(5): 698-711. 4. Zheng, M. et al.: ACS Nano 2012, 6: 9447-9454. 5. Funk, J. L. et al.: Atherosclerosis 1993, 98: 67-82. 6. Abebe, D.G.; Fujiwara, T.: Biomacromolecules 2012, 13: 1828-1836. POS.177 Establishment of a cell-based model to study LTC4synthase inhibitors independent on exogenous LTA4 supply Liening, S.1; Scriba, G.1; Werz, O.1; Garscha, U.1 1 Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-SchillerUniversity of Jena, Philosophenweg 14, 07743 Jena, Germany Cysteinyl-leukotrienes (cys-LT) are powerful pro-inflammatory mediators that cause bronchoconstriction in anaphylaxis and asthma. They are formed via the 5-lipoxygenase (5-LOX)/leukotriene C4-synthase (LTC4S) pathway in immunocompetent cells. 5-LOX metabolizes arachidonic acid to the unstable leukotriene A4 (LTA4) that is subsequently conjugated with glutathione (GSH) to leukotriene C4 (LTC4) by LTC4-S, an integral membrane protein that belongs to the superfamily of membraneassociated proteins in eicosanoid and glutathione metabolism (MAPEG). The tripeptide side chain of LTC4 is cleaved in two successive steps to form LTD4 and LTE4. Cys-LTs are recognized by GPCRs and cys-LT receptor antagonists have mainly been developed to diminish the inflammatory action of cys-LTs. Besides the receptors, LTC4-S might be an additional interesting target to intervene with cys-LTs. However, structural similarities to other MAPEGs and the lack of an appropriate test system have hampered the possibility to reveal and develop selective LTC4-S inhibitors. The main objective of our work was to establish a cell-free and a cellbased assay system for LTC4-S to evaluate putative LTC4-S inhibitors. Therefore HEK293 cells that lack expression of 5-LOX and other MAPEGs were transfected with cDNA of LTC4-S, and stably LTC4-Sexpressing colonies were selected. From these cells, LTC4-S containing microsomes were prepared and incubated with LTA4 methyl ester (LTA4me). LTC4-me was analyzed and quantified by LC-MS/MS and 2.5 µg of microsomes formed around 7 ng of LTC4-me. MK886 as a known LTC4S inhibitor reduced LTC4-me synthesis with an IC50 value of 3.5 µM. However, a major drawback of the cell free assay is the need to supply expensive and unstable LTA4-me. In order to circumvent this problem, we stably co-expressed the LTA4-forming 5-LOX together with LTC4-S in HEK293 cells. Stimulating these cells with Ca2+-ionophore and 20 µM exogenous arachidonic acid, around 300 ng of LTC4 could be detected. MK886 inhibited the LTC4 synthesis with an IC50 value of 3.1 µM. Since HEK293 cells are void of other MAPEG enzymes like 5-LOX-activating protein (FLAP) or microsomal prostaglandin E2 synthase-1 (mPGES-1), the reduced formation of LTC4 by putative inhibitors can be exclusively deduced from LTC4-S intervention. Summarizing, we established a cell-free and cell-based HEK293 system in order to evaluate putative LTC4-S inhibitors. Especially, the cellular system is advantageous since LTA4 as LTC4-S substrate is generated in situ. Additionally, this cell system can be applied to investigate LTC4-S functionality, as site-directed modifications can be studied in a cellular environment. POS.178 Structure-dependent potentiation of the complement inhibiting C1 inhibitor by glycosaminoglycans and other sulfated glycans Alban, S.1; Schoenfeld, A.-K.1 1 Pharmaceutical Germany Institute, Christian-Albrechts-University, Gutenbergstr. 76, 24146 Kiel, Introduction: Activation of the human complement system as part of the innate immunity is associated with many diseases like rheumatoid arthritis or age-related macular degeneration. Hence, reduction of overwhelming activity displays a promising therapeutic strategy. Heparin and other sulfated glycans (SG) are known inhibitors of complement activation. One assumed mechanism is the potentiation of C1 inhibitor (C1inh), an endogenous regulator of the complement system [1]. Objectives: We examined series of SG differing in their degree of sulfation (DS) and their molecular weight (MW) for their C1inh potentiating effect to evaluate structure-activity relationships. The latter could be helpful to develop optimized complement modulators. Materials and Methods: The effect of C1inh in presence and absence of SG on the activity of C1s esterase was measured by a chromogenic substrate assay. We examined the C1inh modulating effects of following SG: (1) seven heparins with MW ranging from 3 to 15 kDa, (2) further partially chemically modified glycosaminoglycans, e.g. heparan sulfates and danaparoid, (3) two series of high-MW (≈ 160 kDa) and low-MW (≈ 19 kDa) semisynthetic β 1,3 glucan sulfates, and (4) three algaederived SG. Results: None of the tested SG inhibited C1s esterase directly. The heparins (DS ≈ 1.1) and the algal SG (DS ≈ 0.6) exhibited a C1inh potentiating effect depending on their MW. On the contrary, the MW turned out to be not important for compounds with a DS above 1.3. For the low-MW β-1,3-glucan sulfates and glycosaminoglycans a DSdependent potentiation of C1inh could be observed, whereas the highMW β-1,3-glucan sulfates led to a potentiation independent of their DS. Even in case of a low DS (≈ 0.7) the high-MW compound exhibited a strong C1inh potentiating effect, whereas the low-MW β 1,3 glucan sulfate was inactive. Conclusion: The C1inh potentiating effect of SG correlated with both increasing DS and MW, whereby a high DS showed to compensate a low MW and, inversely, a high MW a low DS. Acknowledgments: The study was financially supported by the European Fishery Fund (EFF) of the European Union and the Ministry of Agriculture, Environment and Consumer Protection Mecklenburg-Vorpommern References: 1. Rajabi, M. et al.: Biochim. Biophys. Acta. 2012, 1820(1): 56-63. DPhG Annual Meeting 2015 Conference Book • 179 POSTERS POS.179 Design, synthesis and structure-activity relationship of Nphenylbenzenesulfonamides as dual 5-lipoxygenase and microsomal prostaglandin E2 synthase-1 inhibitors As lead structures aspirin, a covalent COX inhibitor, and indomethacin, a competitive COX inhibitor, have been selected. The cluster substituted a (chloro)phenyl ring. Cheung, S.-Y.1; Hanke, T.1; Fischer, K. 2; Listing, M. 2; Temml, V.3; Schuster, D.3; Werz, O.2; Schubert-Zsilavecz, M.1 1 Goethe University of Frankfurt am Main, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany University of Jena, Philosophenweg 14, 07743 Jena, Germany 3,4 Institute of Pharmacy/Pharmacognosy3 or Pharmaceutical Chemistry4, Leopold-FranzensUniversity of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria 2 Friedrich-Schiller Prostaglandins (PGs) and leukotrienes (LTs) are powerful bioactive lipid mediators that have a large number of biological actions in the human body [1,2]. The common precursor of PGs and LTs is arachidonic acid (AA). The 5-lipoxygenase (5-LO) and the microsomal prostaglandin E2 synthase-1 (mPGES-1) are both enzymes which are involved in the AA cascade. The 5-LO is the initial enzyme which catalyzes the transformation of AA to LTs; whereas the mPGES-1 is responsible for the conversion of PGH2 into PGE2 which is one of the most prominent mediators of inflammation, pain, and fever. A novel pharmacological approach for anti-inflammatory therapy is the dual inhibition of 5-LO and mPGES-1. In contrast to the traditional NSAIDs, the dual inhibition of PGs and LTs might be superior over single interference with PGs in terms of anti-inflammatory effectiveness as well as regarding reduced side effects [3]. In this study we wanted to explore the structure-activity relationship of Nphenylbenzensulfonamide derivatives as dual 5-LO/mPGES-1 inhibitors. The lead structure of this series was 4-(N-octyl-4-methylbenzenesulfonamido)-benzoic acid (compound 1, see Figure below), which was originally identified in a virtual screening approach by Waltenberger, B. et al. [4]. For this compound, a facile three-step synthesis was developed and structural optimization was carried out in three directions, while maintaining the central Nphenylbenzenesulfonamide scaffold (see Figure below). Molecular docking studies supported the importance of the sulfonamide moiety. Furthermore, we were able to identify moieties with beneficial or detrimental impact on the overall potency. Asborin, the carborane analogue of aspirin showed a reduced COX inhibition, but proved to be a potent aldo/keto reductase 1A1 inhibitor (IC50=1.4µm) instead. The carborane moiety increased the acetylating activity followed by decreased selectivity [2,3,4]. Indoborin, the ocarborane analogue of indomethacin, remained a potent COX inhibitor, but only against the COX-2 isoform (IC50=3.7µm), which is the favorite COX drug target [5]. Further esterification to the corresponding indoborin methyl ester gave a highly active and selective COX-2 inhibitor (IC50=0.08µm). Acknowledgments: I thank the Beck-Sickinger group (Leipzig), Gust group (Innsbruck), HeyHawkins group (Leipzig), Marnett group (Nashville) for the opportunity to perform the experiments and the Studienstiftung des deutschen Volkes for financial support. References: 1. Scholz, M. et al.: Chem. Rev. 2011, 111: 7035-7062. 2. Scholz, M. et al.: ChemMedChem 2009, 4: 746-748. 3. Scholz, M. et al.: ChemMedChem 2011, 6: 89-93. 4. Scholz, M. et al.: Eur. J. Med. Chem. 2011, 46: 1131-1139. 5. Scholz, M. et al.: Bioorg. Med. Chem. 2012, 20: 4830-4837. POS.181 Structure-Activity Relationships and Development of Novel Aminothiazole-Comprising 5-Lipoxygenase Inhibitors Kretschmer, S. B. M.1; Woltersdorf, S.1; Rödl, C. B.1; Vogt, D.1; Stark, H.1,2; Steinhilber, D.1; Hofmann, B.1 1 Goethe University, Institute of Pharmaceutical Chemistry, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany; 2 Heinrich Heine University, Institute of Pharmaceutical and Medicinal Chemistry, Universitätsstraße 1, 40225 Düsseldorf, Germany; References: 1. Funk, C. D.: Science 2001, 294(5548): 1871−1875. 2. Samuelsson, B.; Morgenstern, R.; Jakobsson, P.J.: Pharmacol. Rev. 2007, 59(3): 207–224. 3. Koeberle, A.; Werz, O.: Curr. Med. Chem. 2009, 16(32): 4274–4296. 4. Waltenberger, B. et al.: J. Med. Chem. 2011, 54(9): 3163–3174. POS.180 ortho-Carborane as Pharmacophore in Cyclooxygenase Inhibitors Scholz, M. S.1 1 Pharmaceutical Institute, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany Artificial building blocks, such as boron clusters, are innovative moieties in extending the chemical space used in drug discovery. Boron forms various cluster types which are often unstable under aqueous conditions. However, C2B10-carborane-clusters, with two carbon atoms at selected vertex positions, are sufficiently stable fragments. Among those, orthocarborane reveals a unique electronic composition and a spherical geometry which occupies double the volume of a rotating phenyl ring [1]. In order to study the applicability of ortho-carborane as pharmacophore it was integrated into well-investigated cyclooxygenase (COX) inhibitors. 180 • DPhG Annual Meeting 2015 Conference Book Acute inflammation is a physiological response to injury, aimed at removing the noxae to restore homoestasis. In contrast, excessive inflammation can be seen as a unifying component in many chronic diseases [1]. Leukotrienes (LTs) herein play an important role, as these lipid mediators are chemotactic for leukocytes (namely LTB4) and increase vasopermeability of small blood vessels (LTC4) [2]. In this respect, LTs are associated with diseases like asthma, allergic rhinitis, rheumatoid arthritis, atherosclerosis and certain types of cancer [3]. 5-Lipoxygenase (5-LO) as the key enzyme of LT biosynthesis is a viable and well studied target. Nonetheless, currently there is only one approved drug on the market (Zileuton, Zyflo®, FDA approval) which usage is limited, partly due to hepatotoxic adverse effects [4]. 2Aminothiazoles may fill this gap, since certain derivatives present well balanced compounds with high potency and appropriate ADMET characteristics. Previous studies focused on the impact of small variations preserving a basic scaffold (ST-1083) on 5-LO inhibition and related targets [5,6]. In this study, however, we focused at assessing the underlying structureactivity relationships on 5-LO inhibition while extending the 2aminothiazole scaffold in order to achieve novel superior lead structures. ST-1853 represents such a novel lead structure with a high potency (IC50 = 50 nM in isolated intact polymorphonuclear leucocytes), appropriate specificity as well as non-cytotoxic behavior. We present a successful lead optimization for further investigations as novel antiinflammatory drug. Acknowledgments: This work was supported by Else Kröner-Fresenius-Stiftung, TRIP, LOEWE, OSF, Fonds der Chemischen Industrie and DFG INST 208/664-1. References: 1. Han, J.; Ulevitch, R. J.: Nat. Immunol. 2005, 6(12): 1198–1205. 2. Rådmark, O. et al.: Biochim. Biophys. Acta 2015, 1851(4): 331-339. 3. Peters-Golden, M.; Henderson W. R.: N. Engl. J. Med. 2007, 357(18): 1841-1854. 4. Steinhilber, D.; Hofmann, B.: Basic Clin. Pharmacol. Toxicol. 2014, 114(1): 70–77. 5. Suh, J. et al.: Chem. Biol. Drug. Des. 2012, 80(1): 89-98. 6. Rödl, C. B. et al.: Eur. J. Med. Chem. 2014, 84: 302-311. INFLAMMATION POS.182 Filaggrin Deficiency Triggers the Invasion of CD4+ T cells via TSLP Secretion Wallmeyer, L.1; Hedtrich, S.1 1 Institute of Pharmacy, Pharmacology and Toxicology, Freie Universität Berlin, Königin-LuiseStraße 2+4, 14195 Berlin, Germany Introduction: Loss-of-function mutations in the filaggrin gene (FLG) are a major predisposing factor for atopic dermatitis (AD), but also immunologic mechanisms are involved in its pathophysiology. The release of cytokines such as IL-4, IL-13, IL-31, TNF-alpha and cellular crosstalk between keratinocytes and lymphocytes are important promoters for the manifestation and maintenance of AD. Moreover, thymic stromal lymphopoietin (TSLP) is highly expressed by keratinocytes in lesional skin of AD patients and stimulates the differentiation of naïve CD4+ cells into Th2 cells which contribute to the induction of allergic inflammation [1]. Hence, to study the effects of CD4+ T cell migration caused by a lack of FLG in more detail, a FLG knockdown skin model (FLG-) was established [2,3]. After T cell supplementation, the expression of TSLP and skin barrier proteins as well as the barrier function of the skin models was investigated. Methods: Normal (FLG+) and FLG deficient (FLG-) skin models were generated according to previously published procedures [2]. Naïve CD4+ T cells were generated from human peripheral blood mononuclear cells by negative selection, activated with anti-CD3/CD28-beads and thereafter, added to the skin models for 2 days at day 12 of tissue cultivation. Subsequently, the skin models were analyzed for protein expression of FLG, involucrin (IVL) and TSLP using immunofluorescence staining. To analyze the influence of Th2 cytokines on T cell migration and differentiation, the skin models were pre-incubated with IL-4 and IL13 (30 ng/ml) for 2 days before T cell supplementation. To investigate the effect of TSLP secretion on T cell invasion, skin models were preincubated with TSLP neutralizing antibody (20 µg/ml). Skin absorption studies were performed with the radioactive labeled standard compound testosterone to assess the skin barrier function. Results and Discussion: Interestingly, FLG deficiency alone triggered the migration of CD4+ T cells into the dermal equivalent of the skin models. In contrast, no immune cell migration was observed in FLG+, but the stimulation with IL-4 and IL-13 stimulated migration of T cells into FLG+ and FLG-. Immunofluorescence staining showed significantly increased TSLP levels in FLG-, FLG- supplemented with CD4+ cells and in FLG+ supplemented with CD4+ cells, although in the latter no immune cell migration was detected. A compensatory upregulation of IVL in FLG- was observed, which was leveled out after T cell contact. Furthermore, only due to the presence of CD4+ T cells underneath the construct, without direct contact, the expression of FLG and IVL in FLG+ were reduced. The influence of T cells led to a reduced barrier function of the skin models. Interestingly, after inhibition of TSLP secretion no T cell migration was observed. Our data show, that an increase of TSLP due to the lack of FLG plays a potential role for the migration of T cells into dermal tissue. As there are no dendritic cells presenting in the skin model, our data suggest a direct link between TSLP and stimulation of T cells without involvement of dendritic cells. Acknowledgments: Financial support by the foundation SET (Foundation for the Promotion of Alternate and Complementary Methods to Reduce Animal Testing) is gratefully acknowledged. References: 1. Soumelis, V. et al.: Nat. Immunol. 2002, 3: 673-80. 2. Küchler, S. et al.: Altern. Lab. Anim. 2011, 39: 471-80. 3. Vávrová, K. et al.: J. Invest. Dermatol. 2014, 134: 746-53 predisposing factor for the manifestation of AD [1]. Aside from barrier deficiencies, AD is characterized by over-shooting Th2-mediated inflammatory processes and impaired innate immunity such as altered expression of antimicrobial peptides (AMP) [2]. The Th2 cytokines IL-4 and IL-13 significantly contribute to the pathogenesis of AD, but their effects on the skin barrier and particularly the interdependencies with FLG deficiency are not yet fully understood. A deficient skin barrier appears to upregulate AMPs [3] and absence of FLG increases the expression of other structural proteins such as involucrin [4]. In this study, the influence of FLG knockdown on the expression of the human β-defensins 1-3 and skin barrier proteins under inflammatory conditions was evaluated. Methods: Normal (FLG+) and FLG deficient (FLG-) skin models were generated according to previously published procedures [5]. In primary human keratinocytes gene knock down was induced by transfection with FLG specific siRNA. Starting at day 10 of cultivation, the skin models were exposed to IL-4 and/or IL-13 (30 ng/ml) for 4 days. Subsequently, protein expression was determined using western blot analysis and immunostaining; gene expression was quantified via RT-PCR. The skin models were further investigated for morphology (H&E staining) and release of the proinflammatory cytokines IL-6 and IL-8 via ELISA. To unravel the mechanism of increased AMP expression, primary human keratinocytes were pre-incubated with IL-1R antagonist (10 µg/ml), IL-6 and TLR-2 neutralizing antibodies (5 µg/ml), respectively. Results and Discussion: Histological examination revealed a thickening of the viable epidermis in the skin models following IL-4 and IL-13 treatment. FLG knockdown amplified this effect (FLG+ 91.0 ± 13.8 µm vs. FLG+/IL4/13 138.1 ± 10.7 µm and FLG- 108.7 ± 14.4 µm vs. FLG-/IL-4/13 164.2 ± 11.0 µm). Cytokine specific increased levels of IL-8 and IL-6 in FLG- models indicate altered susceptibility to inflammatory stimuli. Additionally, we observed a compensatory upregulation of involucrin in the FLG- models, which was considerably disturbed by IL-4 and IL-13 exposure. Furthermore, cytokines significantly reduced the expression of FLG and IVL in normal skin models. Most interestingly, we detected significantly higher expression of HbD2 and HbD3 in FLG- models. This was especially interesting because HbD2 and HbD3 are known to be upregulated through bacteria or inflammation but not by a genetic defect. Since HbDs stimulate the release of proinflammatory cytokines, these results might explain the more severe AD phenotype in patients with FLG mutations [6]. Pre-incubation with an IL1R antagonist significantly diminished the upregulation of HbD2 and HbD3 indicating an IL-1 mediated mechanism. Levels of the constitutively expressed HbD1 were unaffected. In conclusion, FLG deficiency leads to a counterregulation of skin barrier proteins, which is disturbed by Th2 derived cytokines, and, secondly, to an altered immune response indicated by an upregulation of HbD2 and HbD3. Acknowledgments: Financial support by the Collaborative Research Center 1112 for the project C02 is gratefully acknowledged. References: 1. Palmer, C.N. et al.: Nat. Genet. 2006, 38(4): 441-6. 2. Kopfnagel, V. et al.: Curr. Opin. Allergy Clin. Immunol. 2013, 13(5): 531-6. 3. Ahrens, K. et al.: J. Invest. Dermatol. 2011, 131(2): 443-52. 4. Presland, R.B. et al.: J. Invest. Dermatol. 2000, 115(6): 1072-81. 5. Küchler, S. et al.: Altern. Lab. Anim. 2011, 39(5): 471-80. 6. McAleer, M.A.; Irvine, D.A.: J. Allergy Clin. Immunol. 2013, 131(2): 280-91. POS.184 Impact of torsion of N,4-diaryl-1,3-thiazol-2-amines on 5lipoxygenase inhibitory potency Woltersdorf, S.1; Kretschmer, S. B. M.1; Rödl, C. B.1; Vogt, D.1; Steinhilber, D.1; Hofmann, B.1; Stark, H.1,2 1 Institute POS.183 Filaggrin deficiency alters the innate immune response in a 3D skin model Hönzke, S.1; Schäfer-Korting, M.1; Hedtrich, S.1 1 Institute for Pharmaceutical Sciences, Pharmacology & Toxicology, Freie Universität Berlin, Königin-Luise-Straße 2+4, 14195 Berlin, Germany Introduction: Atopic dermatitis (AD) is a chronic, relapsing inflammatory skin disease which is characterized by an impaired skin barrier function. In 2006, mutations in the filaggrin gene (FLG) were identified as a major of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, D60438 Frankfurt am Main, Germany 2 Institute of Pharmaceutical and Medicinal Chemistry, Heinrich Heine University Düsseldorf, Universitätsstrasse 1, D-40225 Düsseldorf, Germany Leukotrienes (LTs) are important lipid mediators derived from polyunsaturated fatty acids playing a key role as regulators in immunity and inflammatory processes and thereby influencing acute as well as chronic diseases, e.g. asthma, allergic rhinitis, cardiovascular disease and certain types of cancer [1]. The LT biosynthesis is initiated by the 5lipoxygenase enzyme (5-LO). It catalyzes the conversion of free arachidonic acid to LTA4 which is subsequently converted into further LT subtypes [2]. Up to date, there is only one approved direct 5-LO inhibitor in clinical use: Zileuton. It acts by chelating the catalytic iron in the active DPhG Annual Meeting 2015 Conference Book • 181 POSTERS site of the enzyme. However, this drug exhibits non-optimal pharmacodynamic and pharmacokinetic profiles. Therefore, novel potent 5-LO inhibitors are of great interest for an anti-inflammatory therapy [3]. Starting from the well-known 5-LO inhibitor SKI-II [4], we prepared a series of N,4-diaryl-1,3-thiazol-2-amines with bulky substituents preferring a twisted conformation at 4-position of the heterocycle (R2) and investigated their influence on inhibition of human 5-LO activity. The impact of the torsion was addressed by additional substitution at the 5position (R1) influencing especially the cytotoxicity profile. The chemical structure of the thiazole-2-amine scaffold was further optimized at positions R3 maintaining high 5-LO inhibitory activity and selectivity. OH R3 S NH R1 N SKI-II R3 S NH R2 Cl OH N Substituents R1 - R2 for torsion Substituens R3 for efficacy From this series we could deduce the importance of bulky substituents and torsion on 5-LO inhibitory potency. With compound ST-1906 (4-((4(2,4-dichlorophenyl)-5-methylthiazol-2-yl)amino)-2,6-dimethylphenol) we present the most potent derivative of this series. It blocks the 5-LO activity with an IC50 value of 0.05 µM (0.033 - 0.067 µM) and demonstrates no signs of cytotoxicity. Acknowledgments: This work was supported by Else Kröner-Fresenius-Stiftung, TRIP, LOEWE, OSF and Fonds der Chemischen Industrie. References: 1. Gualde, N et al.: Trends. Mol. Med. 2008, 14(10): 461-9. 2. Dennis, EA et al.: J. Lipid. Res. 2009, 50(6): 1015-38. 3. Steinhilber, D.; Hofmann, B.: Basic Clin. Pharmacol. Toxicol. 2014: 114(1): 70–77. 4. Suh, J et al.: Chem. Biol. Drug. Des. 2012, 80(1): 89-98. . 182 • DPhG Annual Meeting 2015 Conference Book CLINICAL PHARMACY 4.10 Clinical Pharmacy POS.185 Investigation of altered organ blood flow on carvedilol disposition in adult and paediatric chronic heart failure patients by using PBPK modelling Rasool, M. F.1,2; Khalil, F.1; Läer, S.1. 1 Institute of Clinical Pharmacy and Pharmacotherapy, Heinrich-Heine University, 40225 Düsseldorf, Germany of Pharmacy, Bahauddin Zakariya University Multan, 60800, Pakistan. 2 Department Background: The reduction of organ blood flows in chronic heart failure (CHF) can significantly affect the clearance of drugs with high hepatic extraction. Physiologically based pharmacokinetic modelling (PBPK), due to its ability to incorporate the pathophysiological changes in different diseases can be used in CHF to predict clearance of drugs with high hepatic extraction as carvedilol. Methods: A PBPK model based on cytochrome-P450 clearances was developed. The Developed model was evaluated in healthy adults and after incorporation of reduced organ blood flows in adult CHF patients, the evaluated adult CHF model was scaled to paediatric CHF patients using population based simulator Simcyp®. A two-fold error range for the ratios(Obs/Pred) of the pharmacokinetic parameters was used for model evaluation. Results: The prediction results were within the 2-fold error range. The CL/F ratio(Obs/Pred) was clearly improved after incorporation of reduced organ blood flows in adult CHF patients. In paediatrics CHF patients, improvement in predictions were seen only in adolescents above 17 years of age, staged with NYHA system of classification. Conclusion: There was a clear link between reduced organ blood flows and reduced carvedilol clearance in adult patients with CHF. It was suggested that Ross scoring system in paediatrics was not well correlated with organ blood flow reductions as the NYHA classification system. Due to the mechanistic nature of the developed PBPK model, it can be extended to other drugs with high hepatic extraction. Acknowledgments: The research leading to these results has received funding from the European Union Seventh Framework Programme (FP7/2007-2013) under grant agreement n°602295 (LENA) and from the Faculty Development Program Bahauddin Zakariya University Multan, 60800, Pakistan. POS.186 Evaluation of a regulatory-compliant bioanalytical setting suitable to determine pharmacokinetics and pharmacodynamics of enalapril in Phase II/III studies in all paediatric age groups Burckhardt, B. B.1; Tins, J.1; Läer, S.1 1 Institute of Clinical Pharmacy and Pharmacotherapy, Heinrich-Heine-University, Universitaetsstr.1, 40225 Düsseldorf, Germany Background: Clinical investigations on pharmacokinetics and pharmacodynamics are highly required to improve paediatric pharmacotherapy. The trial-related blood loss should not exceed 3% of the total blood volume within 4 weeks [1]. Prior to this investigation bioanalytical assays in serum, urine and saliva were developed to meet the challenging ethical and analytical burdens especially in neonates and infants. The applicability of the tailored paediatric assays has not been shown and is recommended prior the assays are applied to the vulnerable paediatric population. Objective: To evaluate the applicability and reliability of a new tailored bioanalytical setting for pharmacokinetic and pharmacodynamic determination of drugs acting on the RAA system in paediatric patients. Invasive and non-invasive collection procedures need to be considered. Methods: A proof-of-concept study in 22 healthy adults was conducted by applying the developed low-volume bioanalytical setting. Utilizing HPLC-MS/MS, the bioanalytical assays determined enalapril and enalaprilat concentrations in 50 µL serum as well as 100 µL urine and saliva. Changes of humoral parameters of the RAA system were measured by five immunological assays. Drug concentrations in serum were determined 31 times during the first 24 hours after drug administration while blood for humoral parameters were sampled 10 times. Additionally, the applicability of the low-volume assays was assessed by a regulatory-compliant Phase I study in 24 healthy volunteers analyzing urine and serum concentrations of enalapril and enalaprilat. Results: The proof-of-concept study made very dense and reliable individual concentration-time profiles of the drugs and the humoral parameters available. Using the low-volume assays revealed comparable pharmacokinetic results of enalapril and enalaprilat in serum and urine when compared to data published in literature. Changes in the humoral parameters are also similar and prove the applicability of the bioanalytical platform. Applying the developed low-volume bioanalytical assays allows to determine 6 drug concentrations and 2 times the measurements of changes in the renin angiotensin aldosterone system in newborns at a single day without infringing ethical recommendations. The conducted investigations on non-invasive sampling approaches in saliva appeared inappropriate to replace invasive pharmacokinetic sampling of enalapril. Conclusion: The applicability of the child-appropriate bioanalytical platform was proven in adults and it can be applied in upcoming paediatric Phase II and Phase III studies of the LENA (Labeling of Enalapril from Neonates up to Adolescents) project. Meaningful concentration-time profiles of the cardiovascular drug and their effect on humoral parameters can be obtained even in neonates without infringing ethical recommendations on trial-related blood loss. The obtained pharmacokinetic and pharmacodynamic dataset within the proof-ofconcept study allows to validate physiological-based computer models that are of increasing interest for an optimal planning and conduction of paediatric clinical trials. Acknowledgments: The research leading to these results has received funding from the European Union Seventh Framework Programme (FP7/2007-2013) under grant agreement n°602295 (LENA). References: 1. European Commission, Ethical considerations for clinical trials on medicinal products conducted with the paediatric population 2008. POS.187 Thrombosis prophylaxis in high-risk patients: Is the recommended dosage sufficient? An observational study on pharmacokinetics of Enoxaparin Beheiri, S. N.1,2; Wähnert, D.2; Schulz, D.3; Völler, S.1; Hempel, G.15 WWU Münster Institut für Pharmazeutische und Medizinische Chemie- Klinische Pharmazie, Corrensstraße 48, 48149 Münster, Germany 2 Universitätsklinik Münster- Klinik für Unfall-, Hand- und Wiederherstellungschirurgie, AlbertSchweitzer-Campus 1, 48149 Münster, Germany 3 Universitätsklinik Münster- Klinik für allgemeine Orthopädie und Tumororthopädie, AlbertSchweitzer-Campus 1, 48149 Münster, Germany 1 Background: Deep vein thrombosis is one of the most serious complications in musculoskeletal surgery. The prevalence in critically ill patients is reported up to 60% [1]. Patients after major operations like spinal surgery, endoprosthesis as well as pelvic fractures exhibit a significant elevated risk. Obese patients also belong to the high-risk collective without reference to the type of surgery. As opposed to this, the presence of renal failure entails the danger of accumulation of the anticoagulative agent and raises the risk of bleeding complications. In clinical routine, all patients receive a fixed dose of 40 mg enoxaparin, as recommended in the prescribing information. We hypothesized that distinct patients may have insufficient thrombosis protection by the actual dose recommendations. The aim of this study is to evaluate the effectiveness of the recommended dosage of enoxaparin using antifactor-Xa activity as a surrogate-parameter. Materials and Method: A prospective case controlled study was designed. 117 patients were included and divided in into three groups (Group I (n=43): high- risk patients, <100 kg, eGFR >60 ml/min; group II (n=62): >100 kg, eGFR >60 ml/min; group III (n=12): <100 kg, eGFR 3050 ml/min). Postoperatively, all patients received enoxaparin 40 mg once daily. Anti-factor-Xa activity was measured four and twelve hours (peak and trough level) after application of the third dose to verify if the target area of 0.1-0.4 I.E./ml was reached. Dose adaption was conducted in case of insufficient anti-factor-Xa activity and the activity was checked again later. Furthermore, a population pharmacokinetic model using NONMEM was applied using the anti-factor-Xa activity data of all patients. This model was used to prove that dose adaptation results in reaching target range. DPhG Annual Meeting 2015 Conference Book • 183 POSTERS Results: After 4 h, adequate anti-factor-Xa activity was found only in group I (median 0.20 I.E./ml). 28.5% of all anti-factor-Xa peaks of the obese patients (body weight 114 kg) reached the level of 0.1 I.E./ml (median 0.13 I.E./ml). In group III (median eGFR 45.2 ml/min) all peak levels reached the target area but were significantly increased (median 0.29 I.E./ml). The median activity of <0.1 I.E./ml after 12 h in all groups is not sufficient for prophylaxis. In patients with peak levels of <0.1 I.E./ml after 4 h, the dosage was increased. If the trough level after 12 h was <0.1 I.E./ml, enoxaparin was administered twice daily. In all patients with adapted dose, anti-factor-Xa activity was controlled again. Measured trough levels do not indicate accumulation of enoxaparin (group I: Figure). There was no increase in bleeding complications after adaptation of the dosage schedule. A two-compartment model was adequate for describing the enoxaparin kinetics. Body weight, body mass index and renal clearance were the most influencing covariates for enoxaparin clearance (25.6 ml/h/kg). Interindividual variability was found to be 25.9%. The simulation of antifactor-Xa activities for the adapted dose schedule demonstrates that activities in the target range are achieved. final concentration of 2∙106 CFU/mL in the CC. Next (t=0 h), LEV was added to the CC mimicking an i.v. bolus injection. Samples (350 µL) were drawn at predefined points of time(-2, 0, 0.5, 1, 1.5, 2, 3, 4, 6, 20, 24 h). The experiments were performed at least in duplicate and included a growth control without addition of antibiotic as positive control. The quantification of LEV was performed with a developed, validated fluorimetric assay and for the bacteria the droplet plate assay was used [3]. The simulations of LEV concentration-time profiles were realised with Berkeley Madonna™. Figure: Schematic illustration of the dynamic IVIM [2] Conclusion: Our study demonstrates that anticoagulation with enoxaparin once daily without consideration the patients’ weight and renal function does not achieve sufficient anti-factor-Xa levels in all patients. Consequently, a possible increased risk of thrombosis may exist despite guideline-oriented anticoagulation with enoxaparin. Acknowledgments: Klinik für Unfall-, Hand- und Wiederherstellungschirurgie der Universitätsklinik Münster, Klinik für allgemeine Orthopädie und Tumororthopädie der Universitätsklinik Münster References: 1. Geerts, W. H. et al.:. N Engl J Med. 1994, 331: 1601-6. POS.188 Dynamic in vitro infection model enables mimicking of in vivo levofloxacin PK profiles showing eradication of E. coli infection Bartels, I. K.1; Goebgen, E. B.1; Kloft, C.1 Results: After single i.v. bolus administration the measured maximum LEV concentrations were 8.74±2.08 µg/mL (n=6). The shape of the curve showed an exponential decrease with a half-life of 7.0 h. Concentrations below 1.0 µg/mL were reached after 20 to 22 h. The profile of the measured data were comparable to the previously simulated concentration-time profiles of LEV. For time-kill curves with E. coli (n=1) bacterial samples >3 h after start of LEV exposition were below the limit of quantification. The growth control, however, shows a logarithmic increase of bacteria up to 6 h followed by a constant conc. of bacteria of 2∙109 CFU/mL over time. Discussion/Conclusion: The dynamic IVIM allowed to mimic in vivo concentration-time profile in plasma of LEV after i.v. bolus administration. Therefore, implementation of the PK properties of drugs in in vitro investigations on the PD in clinically relevant scenarios are feasibible to e.g. to detect subtherapeutic antiinfective therapies before clinical trials are performed. Especially investigations on the development of resistance, induced by e.g. drug concentrations below the minimum inhibitory concentration (MIC) at the end of dosing intervals, could be impeded. Due to the neglected immune system the results of the presented IVIM can give a first idea of the PD what can be expected in vivo and need further investigations in e.g. clinics. As proof-of principle, the investigated drug concentration could successfully eradicate E. coli after 3 hours treatment. Further investigations should include bacteria with higher MICs in which sub-therapeutic concentrations at the target side are more likely. References: 1. http://www.rxlist.com/levaquin-drug/clinical-pharmacology.htm (Accessed: 23.06.2015) 2. Michael, J.: Pharmacodynamic in vitro studies contributing to the rational use of linezolid in infections by vancomycin resistant Enterococcus faecium. 2011. 3. Schwalbe, R.; Steele-Moore, L.; Goodwin, A. C.: Antimicrobial susceptibility testing protocols (CRC Press) 2007. Dept. of Clinical Pharmacy and Biochemistry, Institute of Pharmacy, Freie Universitaet Berlin, Kelchstr. 31, 12169 Berlin, Germany 1 Objectives: In order to determine the pharmacodynamic (PD) effect of antibiotics against clinically relevant bacterial strains, often the simpler static in vitro infection models (IVIMs) with a constant unchanging drug concentration are used. As a possibility to allow for a better characterisation of the PD considering in vivo pharmacokinetic (PK) properties of the drug, dynamic IVIMs were developed, which include changing drug concentrations over time. For the antibiotic levofloxacin (LEV), a treatment option for e.g. urosepsis caused by Escherichia coli (E. coli), the maximum plasma concentration after single i.v. administration of 750 mg is 11.5±4.0 µg/mL. With the half-life of 7.5±1.6 h a simulation of a concentration-time profile is possible [1]. The aims of these investigations were (i) mimicking the in vivo plasma concentration-time profile of LEV with a dynamic IVIM [2] and (ii) determining the PD resulting from the applied PK in time-kill experiments with E. coli. Methods: The dynamic IVIM used for the investigations was previously presented [2]. Briefly, pumps managed in- and outflow to the central heatable compartment (CC) (see Figure).The flow rate of the pumps was calculated based on the in vivo half-life of LEV and the volume of the CC (90 mL) and set to 0.15 mL/min. In a pre-incubation step (2 h) E. coli (ATCC 25922) was grown in cation-adjusted Mueller Hinton Broth to a 184 • DPhG Annual Meeting 2015 Conference Book POS.189 Analysis and optimization of the medication of multimorbid patients on acute geriatric wards - A prospective intervention study (OptiMe) Nachtigall, A.1; Heppner, H. J.2; Thürmann, P. A.1 1 Clinical Pharmacology, Faculty of Health, Department of Human Medicine, HELIOS Clinical Center, 42283 Wuppertal, Germany 2 Geriatric Medicine, Faculty of Health, Department of Human Medicine, HELIOS Clinical Center, 58332 Schwelm, Germany Background: Demographic change is a frequently discussed topic due to the challenges that health care professionals have to face whilst dealing with elderly patients. Geriatric patients are characterized by multiple morbidities like congestive heart failure, diabetes, osteoarthritis, hypertension etc. The guideline-adapted treatment of numerous diseases inevitably leads to polypharmacy [1]. This causes drug related problems (DRP) and drug-drug interactions (DDI), which are often unpredictable and can therefore contribute to adverse outcomes such as hospitalizations. As only few clinical trials exist regarding elderly people taking multiple drugs, evidence-based decision-making is often CLINICAL PHARMACY impossible. Therefore the cooperation between health care professionals like pharmacists and physicians is essential to provide an effective and adequate health care. Objective: The aim of this project is the optimization of pharmacotherapy during hospitalization on a geriatric ward in order to improve the elderly patients’ outcomes. The short-term objective is the implementation of a clinical medication management by a pharmacist to detect drug related problems (DRP). Primary endpoint is the percentage of patients having a DRP defined as a) prescription of potentially inappropriate medication (PIM, PRISCUSlist), b) drug-drug interaction (DDI) from “contraindicated” to “concurrent application not recommended”, c) presence of an adverse drug reaction and d) prescription without indication. Methods: For this prospective, controlled, interventional study 400 patients will be enrolled: 200 patients in the intervention ward (A) and another 200 in the control ward (B) will be recruited on the Geriatric ward (HELIOS clinic Schwelm, North Rhine-Westphalia, Germany) during a one year period (January – December 2015). The allocation to ward (A) or (B) is done study-independently in the administration department of the clinic, resulting in a quasi-randomization. Inclusion criteria are the prescription of at least five medications, age of 70 years or above and signed informed consent. Each patient from ward (A) will be paired with one patient from side (B) having the same sex and age ±5 years. Data are recorded in an ACCESS database (Microsoft 2013) specifically developed for this project, where drugs are coded according to the ATC code, adverse drug reactions according to the WHO SOC and their severity using CTCAE criteria. Analysis and screening of prescribed drugs will be performed by using the Medication Appropriateness Index (MAI) [2] and the PRISCUS-List [3] (screening for potentially inappropriate medication (PIM)). Drug-drug interactions will be checked using the ABDA module for DDIs. Functional scores for mobility and cognition and the preferences of the patient - identified by a questionnaire – complete the tools for decision-making. In both groups, the patient’s medication will be documented at admission and discharge, but only in the intervention group drug related problems will be communicated to the physicians. All suggestions of the pharmacist will be documented and coded as well as their acceptance by physicians. Results: The prevalence of DRP is expected to be reduced from approx. 50% to 20% in the intervention group and - considering the learning bias and the routine work - to 30% in the control group. The study protocol was approved by the ethics committee of the University Witten/Herdecke. So far, 106 patients in the intervention and 103 patients in the control group have been included. First experience shows the feasibility of the intervention. Acceptance rate and cooperativeness are high, whereas the daily lack of time of physicians is a limiting factor. Acknowledgments: This study was supported by HELIOS Kliniken GmbH, grant ID 063614 and by Robert Bosch Stiftung. References: 1. Boyd, C. M. et al.: JAMA. 2005, 294(6): 716-24. 2. Hanlon, J. T. et al.: J Clin Epidemiol. 1992, 45 (10): 1045–51. 3. Holt, S.; Schmiedl, S.; Thürmann, P. A.: Dtsch Arztebl Int. 2010, 107(31-32): 543–51. POS.190 Development of a drug-drug interaction registry based on notifications collected in german community pharmacies Braun, C. A.1; Woltersdorf, R.1; Simons, S.2,3; Klahn, D.4; Jaehde, U.1 1 Institute of Pharmacy, Clinical Pharmacy, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany 2 MVDA e.V., Kirschbaumweg 23, 50996 Cologne, Germany 3 Apotheke am Stadttor, Werdohler Straße 4-6, 58809 Neuenrade, Germany 4 LINDA AG, Emil-Hoffmann-Straße 1a, 50996 Cologne, Germany Background: In their daily practice community, pharmacists deal with a multitude of drug-drug interaction (DDI) notifications. Purpose: The aim is to develop a DDI register that continuously collects all DDI notifications in German community pharmacies including how pharmacy staff manages DDI. This can help to estimate the incidence and relevance of DDI in community pharmacies. A DDI register also provides a basis for advanced education of the pharmacy staff in order to improve the quality of DDI management. Methods: We perform our evaluation in cooperation with the LINDA AG, a network of community pharmacies in Germany. In participating pharmacies the pharmacy software automatically generates datasets of all detected DDI notifications, which are sent to a DDI register. A statistical analysis is performed regarding the total amount of DDI notifications, the distribution of the degree of severity, and the incidence of distinct DDI. Results: In a pilot study, we detected more than 490,000 DDI notifications in 74 community pharmacies within four months. Preliminary analysis revealed that about 5% (23,000) of these notifications were due to severe DDI. QT interval prolonging drugs, psychotropic drugs, sympathomimetics, and antihypertensives represent classes of drugs most often involved in severe DDI notifications. Conclusions: Our preliminary analysis indicates the high frequency of detected potential DDI in German community pharmacies but does not reflect their actual relevance. Therefore, we developed a standardized electronic system for documentation of DDI management for further analysis. POS.191 The standardized medication plan – First results from the MMP16 consortium Abbas, Z.1; Schächtele, S.2; Griebel, L.3; Prokosch, H. U.3; Dörje, F.4; Maas, R.2; Fromm, M.2; Dormann, H.5; Friedland, K.1 1 Molecular and Clinical Pharmacy, Department of Chemistry and Pharmacy, FriedrichAlexander-University Erlangen/Nürnberg, Cauerstr.4, 91058 Erlangen, Germany 2 Clinical Pharmacology, Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-University/ Erlangen-Nürnberg, Fahrstr. 17, 91054 Erlangen, Germany ³ Institute for Medical Informatics, Department of Medical Informatics, Friedrich-AlexanderUniversity Erlangen-Nürnberg, Wetterkreuz 13, 91058 Erlangen-Tennenlohe, Germany 4 Pharmacy Department, Erlangen University Hospital, Erlangen, Palmsanlage 3, 91054 Erlangen, Germany 5 Department of Emergency Medicine Hospital Fürth, Jakob-Henle-Straße 1, 90766 Fürth, Germany Side effects and medication errors are frequent causes for hospital admissions. One reason for these mistakes are information gaps in the actual medication of the patient. As an answer to solve these problems, a standardized medication plan was developed by the German ministry of health. This plan is currently tested in three model regions regarding comprehensibleness and acceptance by patients but also by health care providers such as physicians and pharmacists. Our consortium MMP16 consists of 10 medical practices and 10 corresponding community pharmacies in Nürnberg and Fürth which will use the standardized medication plan over 2 years. Our consortium also involves clinicians, clinical pharmacologists, clinical pharmacists, scientists in health economy and medical computer scientist. Together, we developed questionnaires to address the acceptance of patients of the standardized medication plan. In addition, we will analyze the completeness and the update of the medication plans by health care providers using the computer platform MediTalk. Our questionnaires are divided in different parts, they start with demographic questions about the age, sex, education and mother tongue. Then questions regarding the general attitude to medication are included, which are mostly about the importance of medication and the patients’ knowledge about their medication. Adherence is measured using the 8-item-Moriskyquestionnaire. Finally, the questionnaire contains several questions about the medication plan itself as well as the patient’s expectations regarding the effect of having a medication plan for himself but also for the health care providers. To evaluate the ease of use and the comprehensibility, a pretest including 13 patients was conducted in a community pharmacy. Interestingly, 23% of the asked patients do not know the name of the active compound of their medication. Therefore, the question was changed asking for the drug name and not for the active compound. Several other points were also changed improving the ease of use of the questionnaires. In a next step, the questionnaires will be filled in by 120 patients using the standardized medication plan. Acknowledgments: We thank the BMG for funding the MMP16 consortium. DPhG Annual Meeting 2015 Conference Book • 185 POSTERS POS.192 Medication review for geriatric patients in nursing homes Bitter, K.1; Pehe, C.2; Krüger, M.3; Heuer, G.3; Quinke, R.3; Jaehde, U.1 1 Institute of Pharmacy, Clinical Pharmacy, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany 2 AOK Rheinland/Hamburg, health insurance, Kasernenstr. 61, 40213 Düsseldorf, Germany 3 Pharmacists’ Association North Rhine, Tersteegenstr. 12, 40474 Düsseldorf, Germany Background: Drug-related problems (DRP) are common in nursing home residents due to polymedication [1]. Community pharmacies supplying drugs to nursing homes may play an important role in detecting and solving DRP in nursing home residents. Objectives: This project aims to evaluate whether a simple medication review, solely based on the patient’s medication and performed by community pharmacists, can enhance the medication safety of nursing home residents. Methods: At the beginning, participating pharmacists attended a special training focusing on pharmacotherapy and adverse drug events in the elderly. Patients at the minimum age of 65 years insured by AOK Rheinland/Hamburg (AOK) and regularly taking at least five drugs per day were invited to participate. The AOK provided prescription data of these patients to the pharmacies where the current medication and further information from the nursing home, e.g. dose regimens, were added. If necessary, unclear or false medication data of the nursing homes was corrected. Based on the medication list, pharmacists performed a simple medication review according to a specific guideline. The detected and solved DRP were counted. Results: So far, we tested the feasibility of this intervention in a pilot study including five community pharmacies. The medication of 28 patients was surveyed. In 89% of the cases, the pharmacists added further medication data to the provided prescription data. Concerning the nursing homes’ medication data, 18 DRP were detected including e.g. poor documentation of dosage forms or dose regimens or even missing documentation of drugs. In average, the pharmacists identified two DRP per patient. Most frequent were drug-drug-interactions (33%) of which 40% were considered as relevant for the residents’ medication safety. 39% of the patients took drugs considered as potentially inadequate in the elderly. The pharmacists gave recommendations to the physicians if necessary. However, the acceptance rate by the general practitioners was 15% only. Conclusions: A simple medication review performed by pharmacists seems to be feasible and can be performed on the basis of a complete drug anamnesis. Acknowledgments: We thank all participating pharmacies for the excellent collaboration. References: 1. Gurwitz, J. H. et al.: Am. J. Med. 2005, 118: 251-8. In an ongoing clinical trial, microdialysis is performed in the ISF of subcutaneous tissue of neonates treated with VAN. Prior to clinical trials, an in vitro microdialysis characterisation of the particular drug is crucial in order to set optimal settings for a consistent in vivo relative recovery (RR); being a prerequisite for calculation of ISF concentration. Methods: In vitro characterisation of VAN was performed with a standardised microdialysis system [2] and as in the clinical trial with CMA 63 catheters (membrane length 10 mm, molecular weight cut-off 20 kDa). VAN concentration ranged from 10 to 100 µg/mL; the surrounding medium was heated to 37 °C to mimic body temperature. Three different settings were investigated: (i) Ringer’s solution (RS) as perfusate and surrounding medium, (ii) RS as perfusate and phosphate buffered saline (PBS) as medium and (iii) PBS for both, medium and perfusate. The surrounding medium mimicked the ISF. RR was determined for a flow rate of 1.0 µL/min and pH values of 7.0 and 7.4 (for PBS) in both, delivery (i.e. VAN diluted in RS or PBS in the perfusate) and recovery (i.e. VAN diluted in PBS or RS in the surrounding medium) experiments, to quantify and assess equality of RR in both permeation directions. Five replicates were taken in each experiment. An HPLC assay for the quantification of VAN from microdialysate samples was developed and validated according the EMA guideline [3]. Results: RS used as perfusate and PBS as medium (ii) resulted at pH 7.0 in a consistent RR in both experiments over the entire concentration range. At pH 7.4, RR showed lower recovery values compared to delivery but less variable than if PBS was used for both. Setting (i) leads to higher RR in delivery than in recovery and so the RR values were not equal in both directions. In setting (iii) the RR for delivery was consistent over the investigated concentrations and pH range, recovery experiments showed high variations (CV ≤ 23.2%). Conclusion: Based on RR findings, use of RS as perfusate and PBS as surrounding medium led to a consistent RR in in vitro investigations. This setting is also the most similar to the setting in the clinical trial. Equality of RR in both directions could not be proven in vitro and reasons have to be further investigated. In upcoming investigations alternative buffers to PBS will be examined to improve the setting. References: 1. Plock et al.: Biomed. Chromatogr. 2005, 19: 237–44. 2. Simmel, F.; Kloft, C.: Int. J. Clin. Pharmacol. Ther. 2010, 48(11): 695–704. 3. European Medicines Agency (EMA): Guideline on bioanalytical method validation 2012 (http://www.ema.europa.eu/docs/en_GB/document_library/Scientific_guideline/2011/08/WC500 109686.pdf; 29.06.15) POS.194 Variability of meropenem serum concentrations in intensive care patients Ehmann, L.1,2; Zander, J.3; Zoller, M.4; Minichmayr, I. K.1,2, Kloft, C.1 1 Dept. of Clinical Pharmacy and Biochemistry, Institute of Pharmacy, Freie Universitaet Berlin, Kelchstr. 31, 12169 Berlin, Germany Graduate Research Training program PharMetrX, Germany 3 Institute of Laboratory Medicine, Hospital of the Ludwig-Maximilans-University of Munich, Marchioninistr. 15, 81377 Munich, Germany 4 Dept. of Anaesthesiology, Hospital of the Ludwig-Maximilans-University of Munich, Marchioninistr. 15, 81377 Munich, Germany 2 and POS.193 Optimisation of microdialysis of vancomycin in neonates by in vitro experiments Burau, D.1; Fürtig, M.-A.1; Schröpf, S.2; Kloft, C.1 1 Dept. of Clinical Pharmacy and Biochemistry, Institute of Pharmacy, Freie Universitaet Berlin, Kelchstr. 31, 12169 Berlin, Germany 2 Dept. of Pediatrics, Dr. von Haunersches Kinderspital, Lindwurmstraße 4, 80337 Munich, Germany Background: Vancomycin (VAN) is used as an antibiotic agent against infections with gram-positive bacteria especially methicillin resistant Staphylococcus aureus (MRSA) e.g. sepsis. In order to achieve therapeutic VAN concentrations, the pharmacokinetics (PK) has to be well characterised. Extrapolation of PK parameters from adults to other patient populations, e.g. neonates, may increase the risk of therapy failure (underdosing) or adverse effects (overdosing). But especially in neonates, in whom infections may easily be life-threatening, PK studies are difficult to perform due to the ethical reasons and limited availability of blood volume. An alternative, minimally invasive method is the microdialysis technique which benefits from sampling without collecting body fluids. In the particular interstitial fluid (ISF) of the target site, a catheter consisting of a semipermeable membrane is inserted, allowing measurements of unbound and therefore effective drug concentrations continuously [1]. 186 • DPhG Annual Meeting 2015 Conference Book Background and Objectives: Meropenem (MER) is a broad spectrum carbapenem antibiotic frequently used for the treatment of severe nosocomial infections in intensive care patients. In these patients, high pharmacokinetic variability has been shown, which increases the risk of overdosing on the one hand and underdosing or resistance development on the other hand, potentially leading to therapy failure [1]. MER undergoes primarily renal elimination and its activity is linked to time above minimum inhibitory concentration (MIC) [2]. The objective of the present work was to assess serum concentrations after standard doses of MER in a critically ill patient population with respect to effective concentrations. Methods: A monocentric prospective observational study was conducted in an ICU at the University Hospital of Munich in intensive care patients with severe infections. For this analysis, a subpopulation of 39 patients without renal replacement therapy and extracorporeal membrane oxygenation was chosen. 1000 mg (n=38) or 2000 mg (n=1) MER were administered every 8 hours as intravenous short-term infusions (30 min). Multiple serum samples were taken over 4 days and quantification of MER was performed using liquid chromatography tandem mass spectrometry (LC-MS/MS). Besides, different patient factors including e.g. age, weight, BMI, disease scores (APACHE II Score, SOFA Score) CLINICAL PHARMACY and laboratory values (e.g. IL-6, CRP, CD64, creatinine clearance (CLCR)) were determined. An exploratory statistical and graphical data analysis was performed using R 3.2.0. In order to determine the variability of MER serum concentrations, minimal concentrations (Cmin) were compared. To assess whether effective serum concentrations were reached, minimal concentrations of all 4 study days were investigated and compared to non-species-related MIC breakpoints (EUCAST: S/I breakpoint: 2 mg/L, I/R breakpoint: 8 mg/L [3]). Results: Patient characteristics of the examined population were shown to be highly variable. 22 male and 17 female patients with a median age of 58 years (range: 29-84 years) and a median body weight of 70 kg (range: 44.0-140 kg) were analysed. The majority of the patients suffered from sepsis (82.1%), which was most frequently caused by pneumonia (78.1% of sepsis patients) or peritonitis (15.6% of sepsis patients). The patients showed considerable variability in the creatinine clearance (median: 81.0 mL/min, range: 19-229 mL/min) and in IL-6 serum concentrations (median: 100 pg/mL, range: 24.0-10096 pg/mL). High inter-patient variability was also observed for MER minimal concentrations, which differed up to a factor of 1000 (Cmin: median: 2.52 mg/L range: 0.03-30.4 mg/L). Not only variability between the patients was detected, but also within individuals, between the single study days. During the 4 study days, 79.5% of the patients showed at least one Cmin measurement below the S/I breakpoint. In 25.8% of those, even all Cmin values were below 2 mg/L. Comparing the Cmin measurements with the I/R breakpoint, 87.2% of the patients showed at least one Cmin measurement lower than 8 mg/L during the study period. In 67.6% of those, Cmin was always below this breakpoint. In a first investigation to explain the high observed variability in the MER concentrations, a hyperpolic relationship between Cmin concentrations of MER and CLCR was found. Conclusion: High variability of MER serum concentrations was observed in intensive care patients, potentially exposing them to the risk of subtherapeutic levels. Based on the clinical data, a population PK model will be developed to describe the concentration-time profiles in the population as well as variability between patients and study days. Ultimately, a covariate analysis will be performed to identify further patients factor explaining the large variability in the population and to assess whether dose adjustments are required in intensive care patients. References: 1. Gonçalves-Pereira, J.; Póvoa, P.: Crit. Care 2011, 15(5): 1-17. 2. Nicolau, D .P.: CID 2008, 47: 32-40. 3. http://www.eucast.org/clinical_breakpoints (date of access: 25 June 2015) POS.195 Population pharmacokinetics of Voriconazole in pediatric cancer patients, taking the inflammatory status into account München, S. E.1; Pieper, S.2; Kirchhefer, U.3; Müller, C.4; Moskovits, J.5; Lehrnbecher, T.5; Groll, A. H.2; Hempel, G.1 Institut für Pharmazeutische und Medizinische Chemie, Klinische Pharmazie, Westfälische Wilhelms-Universität Münster, Corrensstr. 48, 48149 Münster, Germany 2 Klinik für Kinder- und Jugendmedizin, Pädiatrische Hämatologie und Onkologie, Universitätsklinikum Münster, Albert-Schweizer-Str. 33, 48149 Münster, Germany 3 Medizinische Fakultät, Institut für Pharmakologie und Toxikologie, Westfälische WilhelmsUniversität Münster, Domagkstr. 12, 48149 Münster, Germany 4 Institut für Pharmakologie, Universitätsklinikum Köln, Gleueler Str. 24, 50931 Köln, Germany 5 Zentrum der Kinder und Jugendmedizin, Hämatologie und Onkologie, Universitätsklinikum Frankfurt am Main, Theodor-Stern Kai 7, 60590 Frankfurt, Germany 1 Due to underlying condition and treatment, immunocompromised pediatric patient populations such as pediatric cancer patients are at high risk to develop invasive fungal infections (IFIs). One option to treat and prevent IFIs is the antifungal triazole voriconazole. The pharmacokinetic properties of voriconazole show a high inter-individual variability as well as a high intra-individual variability, which is even more pronounced in the pediatric population [1]. Furthermore, pharmacokinetic properties differ in pediatrics and adults. Previous studies demonstrated that voriconazole is eliminated in adult patients following Michaelis-Menten type pharmacokinetics. However, at adult standard doses of 3-4 mg/kg BID, the elimination of voriconazole in pediatric patients seems to be linear with a lower voriconazole exposure at similar weight-based doses. Thus, to reach adequate plasma concentrations in pediatric patients, doses have to be increased, eventually leading to non-linear pharmacokinetics [2]. One reason for the high intra-patient variability might be the acute level of inflammation which is reflected by using C-reactive protein (CRP) plasma concentrations. Statistical evaluation has confirmed a linear relationship of CRP plasma concentrations and voriconazole trough concentrations in adults [3]. We therefore analyzed the correlation of voriconazole trough concentrations with the CRP in a cohort of immunocompromised pediatric patients. A linear regression analysis with 14 pediatric cancer patients, 13 years old or younger, confirmed the linear relationship of CRP plasma concentrations and voriconazole trough concentrations (p<0.001). A 1 mg/L increase in CRP resulted in an 0.018 mg/L increase in voriconazole trough concentrations (95% CI: 0.011 – 0.026 mg/L). Thus, 49.7% of the variance in the trough concentration can be explained by CRP plasma concentrations. For this study, patient data from therapeutic drug monitoring of voriconazole has been collected since 2002 and collection is ongoing. With this data a pediatric model for voriconazole using a nonlinear mixedeffect modelling approach (NONMEM) was created regarding certain covariates such as CRP levels representing the inflammatory status. A two-compartment model with combined linear and non-linear elimination is used. The aim of this study is to better characterize pharmacokinetic processes of voriconazole in pediatric patients, with special regards to those patients under 2 years of age and to determine if current dosing regimens are the most appropriate and also applicable for this very young patient group. References: 1. Pieper, S et al.: J. Antimicrob. Chemother. 2012, 67(11):2717-24. 2. Karlsson, M. O. et al.: Antimicrob. Agents Chemother. 2009, 53(3):935-44. 3. Van Wanrooy, M. et al.: Antimicrob. Agents Chemother. 2014, 58(12):7098-101. POS.196 High predictability of plasma lacosamide and no differences by different age and gender through normalization processes Schaefer, C.1,2; Cawello, W.1; Andreas, J. O.1 1 UCB Pharma, Monheim, Germany of Düsseldorf, Institute Clinical Pharmacy and Pharmacotherapy, Düsseldorf, Germany 2 University Rationale: Lacosamide (LCM) is a newer antiepileptic drug indicated as adjunctive therapy for partial-onset seizures (POS) in adults. The objective of this analysis was to assess the effect of age and gender on the pharmacokinetic (PK) parameters of LCM through normalization processes and to evaluate if PK of LCM is different over age or influenced by gender and enforces considerations about therapeutic regimes. Methods: Data were extracted from two Phase I clinical pharmacology studies in healthy subjects. First study included healthy Caucasian women (18-40 years, oral LCM 400mg/day) whereas the second study included healthy male and female subjects (>65 years, 200mg/day) and healthy male subjects (18-45 years, 200mg/day). The PK parameters, area under the concentration time curve over a dosing interval at steady state (AUCtau,ss) and maximum measured concentration in a dosing interval at steady state (Cmax,ss) were determined by non-compartmental methods. AUCtau,ss and Cmax,ss values were normalized by body weight, height , fat free mass (FFM), lean body weight (LBW), and by volume of distribution (Vd). FFM, LBW and Vd were approximated through empiric equations.For AUCtau,ss and Cmax,ss statistical comparison of different groups were performed after log-transformation based on an analysis of variance (ANOVA). Results were compared to results of a population pharmacokinetic (POP PK) study of LCM in patients with POS. Results: A total of 66 healthy subjects were included and stratified by age group and gender. AUCtau,ss and Cmax,ss tended to show higher values in females compared to males and also in elderly vs. young subjects. Differences between groups were smaller when data were normalized by the above mentioned parameters. Before normalization higher AUCtau,ss and Cmax,ss values in females compared with males in both age groups (older and younger) were found. The relative bioavailability for AUCtau,ss and Cmax,ss concerning age and gender effects showed that normalization by Vd led the 90% confidence intervals of the ratio for AUCtau,ss and Cmax,ss for age and gender comparisons to fall within the range 80-125%. Same was true for Cmax,ss normalized by LBW or FFM. The POP PK study in adults with POS DPhG Annual Meeting 2015 Conference Book • 187 POSTERS conformed these results and concluded that inter-individual variability of Vd can be explained to a large extent by differences in gender and body composition. Conclusion: Pharmacokinetic evaluations in healthy subjects and in patients with POS revealed that almost all differences in LCM PK parameters observed between young and elderly or between male and female subjects can be explained by body weight or differences in Vd. Results are confirmed by the results of the POP PK study. Disclosure: Work was funded by UCB. All authors are employees of UCB Pharma. References: The poster was presented at the AES in Seattle 2014. The corresponding abstract is published in Epilepsy.Curr. 2015 15(s1):319. POS.197 Reliability of a European survey on the pharmacological management of paediatric heart failure Makowski, N.1; Castro Diez, C.1; Khalil, F.1; Läer, S.1 1 Institut für Klinische Pharmazie und Pharmakotherapie Heinrich-Heine-Universität Düsseldorf, Universitätsstraße 1, 40225 Düsseldorf, Germany Background: Little evidence is at present available to help clinicians guide decisions when tackling the pharmacological management of paediatric heart failure (HF). As a consequence, therapeutic strategies are largely supported by adults’ data extrapolation and own expertise. The variability in drug treatment routines across Europe is expected to be high. Nevertheless, there are no epidemiological data that describe the current situation. Survey research is a form of scientific inquiry that merits rigorous design and analysis in order to allow gathering reliable and unbiased data [1]. In this regard, different strategies need to be implemented during the design and conduction of the study. Making sure that the questions are designed in a clear and unambiguous way is a crucial aspect of the survey development procedure and a key point to guarantee the reliability of the survey instrument. Aim: To assess the reliability of the questionnaire used for a survey developed in the context of the LENA (Labeling of Enalapril from Neonates up to Adolescents) project to characterise the different therapeutic strategies for the management of paediatric HF that are currently practiced across Europe. Methods: A test-retest strategy was chosen to assess the reliability of the questionnaire. This method is used to evaluate whether the same question posed to the same individuals yields consistent results at different times [1]. The answers of a panel of six experts of 3 countries were analysed for this purpose. The questionnaire was tested in October 2014 and retested in April 2015. Individual answers given by each participant were compared for consistency. Consistency was defined for single choice questions (SQC) and open questions (OQ) as an identical answer in test and retest. For multiple choice questions (MCQ) consistency was calculated as percentage of identical answer options selected by each participant per MCQ. Results: Reliability was assessed for 44 questions (considering subquestions). Twenty-two were SCQ, 10 OQ and 12 MCQ. For the 22 SCQ, on average 76% (33-100%) of the participants gave a consistent answer. For 6/22 questions, all participants were consistent. For the OQ on average 65% (25-100%) of the participants gave a consistent answer. In two out of ten questions, all experts gave a consistent answer. For the MCQ, physicians gave a consistent answer with an average of 83% (6892%). Conclusions: No formal cut-off values exist to judge the acceptable level of reliability of questionnaires within the scientific community. The calculated values for the SCQ (76%), OQ (65%) and MCQ (83%) seem to be similarly high. It has to be taken into consideration that the long period of time (6 months) between the test and retest, rapidly changing knowledge in medical science, and also increased awareness to the subject triggered by the survey could have contributed to the fact that full consistency was not reached. Acknowledgments:The research leading to these results has received funding from the European Union Seventh Framework Programme (FP7/2007-2013) under grant agreement n°602295 (LENA) References: 1. Burns, K. et al.: CMAJ 2008, 179(3): 245-252. 2. Andres, L.: DESIGNING & DOING SURVEY RESEARCH (SAGE) 2012. 188 • DPhG Annual Meeting 2015 Conference Book POS.198 Compatibility analysis of propofol – Optimization of the drug treatment safety Gersonde, F.1; Kunze, T.1; Eisend, S.2, Haake, N.3 1 Department of Clinical Pharmacy, Pharmaceutical Institute, Christian-Albrechts-University Kiel, Gutenbergstraße 76, 24118 Kiel, Germany 2 Dispensary University Hospital Schleswig-Holstein, Campus Kiel, Arnold-Heller-Straße 3, 24105 Kiel, Germany 3 Department of Intensive Care, imland Hospital Rendsburg, Lilienstraße 20-28, 24768 Rendsburg, Germany Infusion therapy is an essential part in the treatment of intensive care patients. Due to increasingly complex therapy standards a simultaneous application of multiple drugs through a central multi-lumen catheter is unavoidable [1]. This entails the risk of physicochemical and chemical incompatibility reactions. One standard sedative used on the intensive care unit is propofol. Because of its physicochemical and optical properties propofol poses a special risk in identifying stability problems and incompatibilities. Additionally, only limited compatibility data are available [2,3]. The purpose of this work is the optimization of the drug treatment safety in a cardiovascular intensive care unit in preventing drug incompatibilities of propofol with other analgetic and sedative drugs. On the cardiovascular intensive care unit documented propofol drug combinations were narrowed down to practice-oriented combinations of propofol 2% with clonidine, midazolam, sufentanil, remifentanil, piritramid, (S)-ketamine, lormetazepam, γ-hydroxybutyric acid and dexmedetomidine which were diluted with sodium chloride 0,9% to standardized concentrations. Mixtures at a ratio of 1:1, 1:10 and 10:1 were stored at room temperature for 7 days. Samples were taken at defined points of time. The physical and the emulsion stability in particular were determined by pH value, zeta potential and globule size distribution measurements using light backscattering as well as dynamic light scattering and laser diffraction. Analyses on crystal and microbiological growth give additional information about the stability. The chemical stability determination is carried out by high performance liquid chromatography (HPLC). The light backscattering and zeta potential analyses resulted in three stability groups for the 1:1-mixtures in which the group with the most destabilization phenomena consisted of propofol, remifentanil and lormetazepam. All other mixtures remained stable over a defined period of time. The results of the globule size distribution measurements of the 1:1, 1:10 and 10:1 mixtures using dynamic light scattering and laser diffraction showed that each combination of propofol with remifentanil, piritramid and γ-hydroxybutyric acid is instable. No crystal and bacterial growth as well as no stability-relevant pH-shift could be detected. The HPLC data indicate a chemical stability of all previously tested propofol drug (1:1) combinations. Evidence for incompatibilities and compatibilities of propofol with analgetic and sedative drugs could be obtained. Through further investigations the drug treatment safety should be increased. References: 1. Kanji, S. et al.: Crit. Care Med. 2010, 38(9): 1890-1898. 2. Michaels, MR.; Stauffer, GL.; Haas DP.: Ann. Pharmacother. 1996, 30: 228-232. 3. Lilley, E. M. M. et al.: Anaesthesia 1996, 51: 815-818. BIOTECHNOLOGY 4.11 Biotechnology POS.199 Modification of glyco-engineered living cells by copper-free (SPAAC) and copper catalyzed azide alkyne cycloaddition (CuAAC) Gutmann, M.1; Memmel, E.2; Braun, A.1; Seibel, J.2; Meinel, L.1; Lühmann, T.1 1 Institute 2 Institute for Pharmacy and Food Chemistry, University of Würzburg, Germany for Organic Chemistry, University of Würzburg, Germany Introduction: The surface of cells consists of a variety of functional elements, including a cell and site specific glycocalyx. These glycan structures impact the function, stability and localization of proteins on the cell surfaces or critically control cell-cell recognition processes [1]. Azide modified monosaccharides can be incorporated into the glycocalyx, and are thereby accessible for e.g. Cu(I)-catalyzed azide alkyne cycloaddition (CuAAC) and strain-promoted alkyne-azide cycloadditions (SPAAC) [2,3]. This study addresses (i) the temporal design space within which the chemistries can be performed without jeopardizing cell toxicity, (ii) the effectiveness of cell surface decoration through the azide functionlized glycocalyx and (iii) the cellular sequestration of fluorescence dyes coupled onto the cell surfaces by means of these chemistries. Materials and Methods: An azide-functionality was introduced onto the surface by means of a glyco-engineering step using tetraacylated-Nazidoacetylglucosamin (Ac4GlcNAz) [2]. Cell toxicity of the CuAAC was assessed by FACS measurements of propidium iodide (PI) and fluorescein diacetate (FDA) stained cells and qPCR analysis of apoptotic and anti-apoptotic maker genes. The Intensity of CuAAC compared to SPAAC was measured after surface staining with the fluorescent dye sulfo-Cy5-alkine by flow cytometry (FACS) [4]. The sequestration of the surface coupled fluorescent dye was assessed by following the residence time through confocal laser scanning microscopy (CLSM). Results and Discussion: qPCR analysis of all conditions did not induce mRNA levels of apoptotic (Caspase-2, Bcl 2 associated X protein, apoptotic protease activating factor I) and anti-apoptotic (B cell lymphoma 2) maker genes after 5 and 20 minutes using best conditions (50 µM CuSO4; 250 µM Tris[(1-benzyl-1H-1,2,3-triazol-4yl)methyl]amine (THPTA); 2,5 mM C6H7NaO6) verified by FACS analysis. In a series of experiments, the cell surface of azido-modified NIH3T3 and Freestyle 293-F cells was decorated by means of glycoengeneering followed by CuAAC or SPAAC reaction using alkyne-functionalized fluorescent dyes. Images of fluorescence labelled cells show a high degree labelling of the cell membranes (Figure A) with decrease in the fluorescence over time (Figure B) (12 hours). Flow cytometry analysis of Freestyle 293-F cells shows an increased degree of labelling with a fluorescent dye over time in both reaction-types (CuAAC and SPAAC) with reduced background noise and efficiency regarding SPAAC. CuAAC is an acceptable chemistry when applied to living cells in a strictly controlled time frame. Exposure to Cu+ as required for the reaction may not exceed 20 minutes and covalently coupled molecules (as assessed for a fluorescent dye) remain detectable on cellular surfaces for 12 hours following the coupling. SPACC shows advantages in form of lower toxicity and less unspecific binding but leads to lower coupling efficacy. POS.200 Spatially controlled decoration of biologics using enhanced green fluorescent protein as model protein Wurzel, J.1; Lühmann, T.1; Meinel, L.1 1 Institute for Pharmacy and Food Chemistry, University of Würzburg, Germany Introduction: Replacing existing amino acids (AA) by unnatural AA (uAA) allows for an efficient introduction of one or more novel and unique functional group(s) into a protein. The resulting proteins can be decorated solely at the site of the uAA with small chemical drugs, other peptides or proteins, or polymers with unprecedented spatial control and homogenous product outcome. Therefore, this genetic engineering strategy is instrumental in addressing the heterogeneous product outcome of current, chemical decoration strategies of biologics. In this study, we demonstrate two different methods for spatially controlled N-terminal decoration using eGFP as a model protein, (#1) a copper-catalyzed azide-alkyne cycloaddition (CuAAC) or (#2) by factor XIIIa (FXIIIa) mediated acyl-transfer. Biotin (as an example for a low molecular weight molecule) and a 10 kDa polyethylene glycol (PEG) polymer were deployed as decorating molecules. Materials and Methods: eGFP analogues were profiled for subsequent CuAAC by introducing the uAA propargyl-L-lysine (Plk) into the protein at position 4 (L4Plk) deploying E. coli with an expanded genetic code [1,2]. In an alternative approach, a specific amino acid sequence (NQEQVSPL), derived from alpha-2-plasmin inhibitor (a2PI) serving as a high affinity target for the transglutaminase FXIIIa, was integrated [3,4]. The factor XIIIa mediated acyl-transfer requires a second peptide (FXIIIapeptide: Ac-FKGG-PEG6-C-NH2) [5], which was synthesized by solid phase peptide synthesis. Azido functionalized Biotin or PEG (required for decoration strategy #1; CuAAC click chemistry) and maleimide/acrylamide functionalized Biotin or PEG (required for decoration strategy #2; the biotin/PEG was coupled to cysteine of the FXIIIa-peptide through a Michael addition) were purchased. A2PI-eGFP and Plk-eGFP were expressed in E.coli BL21 (DE3) and purified by ion affinity chromatography. All species were characterized by MALDI-MS, ESI-LC-MS/MS and HPLC and fluorometric characterization. Results and Discussion: A2PI-eGFP and Plk-eGFP were expressed in reasonable yields (30 and 2 mg/L, respectively) and purities of approximately 95%. Each expressed mutant displayed typical fluorescent properties identical to wild-type eGFP. The synthesis of FXIIIa-peptide, A2PI-eGFP-biotin/PEG and Plk-eGFP-biotin/PEG constructs was successfully performed and characterized. Both strategies (#1 CuAAC; #2 Transglutaminase) proved successful for efficient, rapid, and site directed decoration of eGFP. The technique is introducing tight spatial control for the decoration of biologics translating into unprecedented homogenous product outcome of the decorated species for future pharmaceutical application. References: 1. Eger, S. et al.: Methods Mol. Biol. 2012, 832: 589-596. 2. Nguyen, D. P. et al.: J. Am. Chem. Soc. 2009, 131: 8720. 3. Patterson, J. et al.: Mater. Today 2010, 13(1-2): 14–22. 4. Früh, S. M. et al.: Chembiochem. 2014, 15(10): 1481-6. 5. Hu, B. H.; Messersmith, P. B.: J. Am. Chem. Soc. 2003, 125(47): 14298-9. POS.201 Figure: 5 min CuAAC reaction (50 µM CuSO4; 250 µM THPTA; 2,5 mM C6H7NaO6) of NIH 3T3 cell membrane with a red fluorescent refers to Sulfo-Cy5-alkyne and blue fluorescent to Dapi. (A) Image after 30 minutes (B) Image after 12 hours References: 1. Freeze, H. H. et al.: Nature Rev. 2006, 7: 537-551. 2. Homann, A. et al.: Beilstein J. Org. Chem. 2010, 6: 24. 3. Memmel, E. et al.: ChemComm (Cambridge, England). 2013, 49(66): 7301–3. 4. Hong, V. et al.: Bioconjugate Chem. 2010, 8(8): 1620-34. Site-directed immobilization and bioresponsive release of anticatabolic agents for muscle regeneration Braun, A.1; Ritzer, J.1; Gutmann, M.1; Ebert, R.2; Jakob, F.2; Lühmann, T.1; Meinel, L.1 1 Institute for Pharmacy and Food Chemistry, University of Würzburg, Germany Center for Musculoskeletal Research, Würzburg, Germany 2 Orthopedic Introduction: Progressive loss of skeletal muscle mass, strength and function during aging (sarcopenia), poses a major threat to independence DPhG Annual Meeting 2015 Conference Book • 189 POSTERS and quality of life in the elderly. Therefore, developing therapeutic interventions to stop its progression is of great interest. In our strategy we target myostatin, a potent negative regulator of myogenesis that is highly enriched in skeletal muscle of sarcopenic patients, with peptide antagonists [1]. Intending to control the release of this biologic in case of local tissue inflammation during the progression of the disease, we immobilize the peptides by means of protease-degradable linkers on the surface of nanoparticles. In this way, we create a bioresponsive release system that is sensitive to the upregulation of matrix metalloproteinases in case of local inflammation triggering the release of the biologic from the particulate carrier. Materials and Methods: The myostatin inhibitors and protease-sensitive linker sequences were manufactured by solid phase peptide synthesis [2] with a propargyl-modified glycine analogue introduced into the sequence in case of the myostatin inhibitor and an azido-homoalanine in case of the linkers for the conjugation via click chemistry [3]. RP-HPLC and MALDI-MS were deployed for verification of purity, and potency was profiled by a luciferase-based reporter gene assay and by C2C12 myoblast differentiation ability. Particle preparation was performed by reacting alkyne-functionalized poly methyl methacrylate (PMMA) nanoparticles with the bi-azido-functional MMP-sensitive linker in presence of 0.25 mM copper(I)-catalyst and 0.5 mM THPTA, followed by reaction with the fluorescent dye DBCO-PEG4-carboxyrhodamine and fluorescence-labelled anticatabolic peptide, respectively. Release experiments were performed using 900 ng/mL of MMP-1, -8 and -9 at 37 °C for a period of 20 hours, during which several readings were conducted in order to obtain a release profile [4]. Results and Discussion: Successful synthesis and purification of the functionalized peptides was demonstrated by mass spectrometry and HPLC. The luciferase assay and C2C12 differentiation assay confirmed the inhibiting activity on myostatin signalling and comparison of the potency of alkyne-functionalized and original peptides resulted in similar outcome. The protease-sensitive linker functionalized for conjugation with the anticatabolic peptides was optimized accordingly to ensure a high percentage of proteolytic cleavage (≥ 96%) by incubation with elevated concentration of each of the proteases MMP-1, -8 and -9. The successful coupling of the linker to alkyne-functionalized PMMA particles was indicated by labelling with the fluorescent dye DBCO-PEG4carboxyrhodamine. Fluorescence intensity of the supernatant after cleavage with MMPs was analyzed by RP-HPLC with fluorescence detection and confirmed the successful cleavage of the proteasesensitive linker from particle surface. In conclusion, a potent site-specifically modified myostatin inhibitor with retained activity was developed for advanced controlled release approaches. The immobilization through protease-sensitive linkers on nanoparticles demonstrated the ability for surface decoration and created a system responding to local tissue inflammation. This approach is further profiled for boosting muscle function and regeneration in ongoing studies e.g. by combining the anticatabolic agent with growth factors like IGF-1. Acknowledgments: The financial support from the Bavarian Research Foundation (FORMOsA grant) is gratefully acknowledged. References: 1. Tsuchida, K. et al.: Current Opinion in Drug Discovery & Development, 2008, 11: 487-94. 2. Han, H. et al.; 2004, US 2004/0181033 A1. 3. Hong, V. et al.: Angewandte Chemie- International Edition. 2009, 48: 9879-83. 4. Steinhagen, M. et al.: ACS Applied Materials & Interfaces. 2014, 6: 5891-99. POS.202 MiR-CLIP – a chemical biology strategy for the direct identification of microRNA targets enabled by chemically synthesized microRNA probes Brunschweiger, A.1,2,5; Imig, J.1,5; Brümmer, A.3; Guennewig, B.1; Mittal, N.3; Kishore, S.3; Tsikrika, P.1; Gerber, A. P.4; Zavolan, M.3; Hall, J.1 1 Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland; of Chemistry and Chemical Biology, TU Dortmund University, Dortmund, Germany; 3 Computational and Systems Biology, Biozentrum, University of Basel, Basel, Switzerland; 4 Department of Microbial and Cellular Sciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey, UK. 5 Contributed equally. 2 Faculty Identifying the interaction partners of noncoding RNAs is essential for elucidating their functions. The chemical synthesis of site-specifically modified microRNAs [1,2] enabled a chemical biology strategy for the 190 • DPhG Annual Meeting 2015 Conference Book direct identification of their RNA targets, their “targetome”. We synthesized (pre-)miRNAs modified with a psoralen photo linker and biotin affinity handle to capture their targets in cells in an approach, termed microRNA crosslinking and immunoprecipitation (miR-CLIP) [3]. Photo-crosslinking coupled to a tandem purification strategy consisting of Argonaute 2 immunopurification followed by streptavidin affinity purification of mirCLIP probe-linked RNAs provided selectivity in the capture of targets, which were identified by Illumina sequencing. miRCLIP with pre-miR-106a, a miR-17-5p family member, identified hundreds of putative targets in HeLa cells, many carrying conserved sequences complementary to the miRNA seed but also many that were not predicted computationally. MiR-106a overexpression and inhibition experiments confirmed that miR-CLIP captured functional targets, including H19, a long noncoding RNA that is expressed during skeletal muscle cell differentiation. We showed that miR-17-5p family members bind H19 in HeLa cells and myoblasts. During myoblast differentiation, levels of H19, miR-17-5p family members and mRNA targets changed in a manner suggesting that H19 acts as a ‘sponge’ for these miRNAs. The miR-CLIP protocol was successfully applied to identify targets of further microRNAs, e.g. let-7g. Figure: Schematic presentation of miR-CLIP, a chemical biology approach for the identification of interaction partners of microRNAs. Acknowledgments: This work was supported with two grants by the Swiss National Science Foundation: CRS205321_124720 and CRSII3_127454 (joint Sinergia grant). References: 1. Rebhan, M. A.; Brunschweiger, A.; Hall, J.: Chembiochem 2013, 14(16): 2091-2094. 2. Pradère, U. et al.: Angew. Chem. Int. Ed. 2013, 52(46): 12028-12032. 3. Imig, J. et al.: Nat. Chem. Biol. 2015, 11: 107-115. POS.203 Defined immobilisation of interleukin-4 (IL-4) for spatial controlled M2 macrophage polarization Lühmann,T.1; Spieler V. 1; Werner, V. 1; Meinel, L. 1 1 Institute for Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany For abstract see Short Poster Lecture SPL.011 on page 108. POS.204 Screening of nanoparticles for drug-delivery across the blood-brain barrier using autodisplayed LRP1 IV-domain on E. coli Fenoy, C. G.1; Raudszus, B.2; Nienberg, C.1; Langer, K.2; Jose, J.1 1 Institute of Pharmaceutical and Medicinal Chemistry, PharmaCampus, Westfälische WilhelmsUniversität Münster, Corrensstraße 48, Münster, 48149, Germany 2 Institute of Pharmaceutical Technology and Biopharmacy, PharmaCampus, Westfälische Wilhelms-Universität Münster, Corrensstraße 48, Münster, 48149, Germany The blood-brain barrier (BBB) surrounding the central nervous system (CNS) protects it from widely different potential hazards. Due to its complex structure the pass of compounds across the BBB is highly restricted. This is an important drawback for the therapy of diseases like brain tumors or Alzheimer´s disease. The high molecular weight drugs used are normally not able to reach its target specifically and in large amounts, without altering the integrity of the BBB [1]. Many specific transport systems, like LPR1 (Low-density lipoprotein receptor-related protein-1), contribute to the flow of substances between bloodstream and the CNS. LRP1 has diverse biological functions, including ApoE binding BIOTECHNOLOGY and endocytosis. Nanoparticles modified with ApoE, loaded with a drug of interest, could bind to LRP1. The whole system will be internalized and the drug could pass though the BBB, thereby reaching its target [2]. Previous studies showed the high binding affinity of ApoE to the fourth binding domain of LRP1 (LRP1-IV) [3]. In this work, LRP1-IV was expressed on the surface of E. coli using Autodisplay technology. For this purpose an artificial gene for a fusion protein was constructed. The fusion protein consisted of an N-terminal signal peptide directing the protein across the inner membrane of E. coli, the LRP1-IV domain, and the Cterminal autotransporter facilitating the transport of LRP1-IV to the surface of the cell. Once LRP1-IV is expressed on the surface, whole cells can be used for binding studies with ApoE, avoiding cumbersome protein purification [4]. Surface display of LRP1-IV was verified by western blot of outer membrane preparations and flow cytometry of whole cells with a specific LPR1 antibody. Flow cytometric analysis also indicated that cells displaying LRP1-IV bind purified ApoE3 and ApoE3 labeled with PromoFluor-NHS 633 Our next step will be to test the binding of ApoE3 modified nanoparticles to surface displayed LPR1-IV. These nanoparticles could be used as carriers across the BBB for drugs which normally are not able to cross this barrier [5,6]. The assay established in this study allows further investigation relevant for the development of nanoparticles, since binding affinities between ApoE3 variants and LRP1-IV or variants thereof could be easily quantified. References: 1. Abbott, N. J.: J. Inherit. Metab. Dis. 2013, 36: 437-449. 2. Wagner, S. et al.: PLoS One 2012, 7: e32568. 3. Neels, J. G. et al.: J. Biol. Chem. 2009, 274: 31305-31311. 4. Jose, J.; Meyer, T. F: Microbiol. Mol. Biol. R. 2007, 71: 600-619. 5. Michaelis, K. et al.: J. Pharm. Exp. Ther. 2006, 317: 1246-1253. 6. Zensi, A. et al.: J. Control. Release 2009, 137: 78–86. DPhG Annual Meeting 2015 Conference Book • 191 POSTERS POS.206 4.12 Other topics POS.205 Rapid bacterial cell counting method: Proof-of-principle with a batch culture of E. coli Goebgen, E. 1 Dept. B.1; Wicha, S. G.1; Kloft, C.1 of Clinical Pharmacy and Biochemistry, Institute of Pharmacy, Freie Universitaet Berlin, Kelchstr. 31, 12169 Berlin, Germany Objectives: Quantification of bacteria is one of the key steps in in vitro infection models and performed for different purposes, e.g. determination of antibiotic treatment efficacy or bacterial resistance. The objective of the current investigations was to use a recently developed and validated electronic cell counting method [1] to quantify bacterial concentrations of a growing culture of Escherichia coli (E. coli). Furthermore, the droplet plate counting method as the “gold standard” was to be performed in parallel to enable a comparison between the two methods. Methods: The in vitro experiments were performed under static conditions in a cell-culture flask (batch culture), filled with 9.5 mL cationadjusted Mueller Hinton Broth (CA-MHB). Colonies from a freshly prepared subculture of E. coli (ATCC 25922) dispensed in 0.9% NaCl and adjusted to McFarland 0.5 were added to the culture flasks to obtain an inoculum of 106 CFU/mL after a pre-incubation step of 2 hours. Samples were taken either directly after the start of the incubation “lagexperiments” or after the pre-incubation step of 2 hours “logexperiments”. The predefined sampling time points were the first sample (t=0), 1, 2, 4, 6, 8, 10 and 24 hours after the first sample. Bacterial concentrations of the samples were measured using both the electronic cell counting and the droplet plate counting method; both methods being explained elsewhere [1, 2]. All experiments were performed at least in triplicate. For reasons of comparability the results of the electronic cell counting method were transformed from count/mL to colony forming units (CFU)/mL, using a strain-specific defined calibration function (y=0.166x+1951). Results: Bacterial concentrations in CFU/mL versus the time after start of incubation (TASI) showed a comparable curve shape for both cellcounting methods, with a slightly higher bacterial concentration (<1 decade) for the electronic cell counting method. This difference was visible until TASI=6 hours or TASI=4 hours for lag- or log-experiments, respectively. The delay in bacterial replication in the first 2 hours, visible as a slight increase of half a decade in the first 2 hours, was more pronounced when performing the “gold standard” as quantification method (“gold standard” vs. electronic cell counting: 5∙105 to 106 CFU/mL vs. 9∙105 to 9∙107 CFU/mL). From the maximum bacterial concentration 109 to 1010 CFU/mL at TASI=6 hours, the bacterial concentration stabilised in a “plateau phase” between 109 to 1010 CFU/mL until TASI=12 hours. At the end of the incubation period (TASI=24 or 26 hours for lag- or log-experiments, respectively), the bacterial concentration reached a value of ≤109 CFU/mL. Conclusions: The newly validated electronic cell counting method for E. coli successfully quantified samples from a batch culture. Comparison with results determined with the “gold standard” of bacterial quantification showed good agreement between the two different measurement methods. Furthermore, the shape of the bacterial concentration-time profile showed the expected shape with the different phases typical of batch cultures: delay in bacterial growth (lag-phase), exponentially growing bacteria (log-phase), dynamic equilibrium between growing and less viable bacteria (stationary phase), decline of viable cells (net death phase) [3]. Therefore, the electronic quantification method seemed to be a promising alternative to the time-consuming “gold standard”, which includes an incubation step of at least 20 hours for E. coli. With a measuring time of approximately 10 s, the electronic cell-counting method could streamline the investigations on antibiotic efficacy in in vitro infection models if a quantification step of bacteria is included. Further investigations on the influence of the diameter evolvement, especially in the first two hours of incubation (lag-time), on the number of measureable cells shall be the next steps. References: 1. Goebgen, E.B.; Wicha, S.G.; Kloft, C.: 25th ECCMID, Copenhagen, Denmark. 2015: EV0532 2. Schwalbe, R.; Steele-Moore, L.; Goodwin, A.C.: Antimicrobial susceptibility testing protocols (CRC Press) 2007. 3. Monod, J.: Selected Papers in Molecular Biology (Academic Press, Inc.) 1978. 192 • DPhG Annual Meeting 2015 Conference Book Animal substances in the materia medica of the 18th century Krafczyk, K.1; Friedrich, C.1 Institute for the History of Pharmacy, Universität Marburg, Roter Graben 10, 35037 Marburg, Germany 1 Animal derived remedies played an important role in the history of the materia medica from ancient times until the end of the 18th century. In the 17th century the pharmaceutical use of animals reached heyday, because of the ‘Signaturenlehre’ and the publication of the ‘Heylsame Dreck-Apothecke […]’ (Healing dirt pharmacy) by Christian Franz Paullini (1634–1712) in 1696. During our studies we examined three important pharmacopoeias of the 18th century to depict the postulated decline of the pharmaceutical use of animals. In the Dispensatorium Brandenburgicum (1698) 54 different animals were used to obtain remedies. Because of the lexical scope of the Pharmacopoea Wirtenbergica (1741) we could even prove use of 78 animals. In the Pharmacopoea Borussica (1799) only 21 animal derived remedies were detected. Consequently, as it is shown by the above numbers, various animals have been pharmaceutically used. One of them was the red coral (Corallium rubrum), also referred to as the precious coral. We present preliminary results of our study about the typical therapeutic indications of this cnidarian in the 18th century. It was used for example as an antiepileptic, heart strengthening and hemostatic remedy. Furthermore, it was applied against stomach complaints, which can be explained by its high content of calcium carbonate, absorbing excess gastric acid. This poster will illustrate the great variety of animals used as remedies and describe the pharmaceutical use of the red coral in the 18th century. References: 1. Friedrich, C.; Müller-Jahncke, W.-D.: Geschichte der Pharmazie. Bd. 2 - Von der Frühen Neuzeit bis zur Gegenwart (Govi-Verlag) 2005. 2. Schneider, W.: Lexikon zur Arzneimittelgeschichte. Sachwörterbuch zur Geschichte der pharmazeutischen Botanik, Chemie, Mineralogie, Pharmakologie, Zoologie. Bd. 1 - Tierische Drogen (Govi-Verlag) 1968. 3. Schindler, H.; Frank, H.: Tiere in Pharmazie und Medizin. 50 Einzeldarstellungen (Hippokrates-Verlag) 1961. POS.207 Implementation of a GxP compliant quality system encompassing all consortium members of the FP-7 funded LENA Project Ciplea, A. M.1; Kleine, K.2; Burckhardt, B. B.1; Läer, S.1; Breitkreutz, J.3; Špatenková, L.4; Klingmann, I.4 1 Department of Clinical Pharmacy and Pharmacotherapy, Heinrich-Heine-University, Universitätsstr.1, 40225 Düsseldorf, Germany 2 Simply Quality – Dr. Karl Kleine, Johannes-Damrich-Str.4, 82362 Weilheim i. OB, Germany 3 Ethicare GmbH, Am Fliederbusch 2, 45721 Haltern am See, Germany, 4 Pharmaplex bvba, Avenue Saint-Hubert 51, 1970 Wezembeek-Oppem, Belgium Background: The LENA (Labeling of Enalapril from Neonates up to Adolescents) project has been initiated to improve the healthcare of children with heart failure by an enalapril orodispersible mini-tablet. The LENA consortium combines academic clinical research centres, SMEs (small and medium-sized enterprises) and a patient/parent advocacy organisation. The objective of the project requires to comply with respective GxP regulations like Good Manufacturing Practice (GMP), Good Clinical (Laboratory) Practice (GCP/“GCLP” [1]) and Good Vigilance Practice (GVP). The project team is comprised of sub-teams experienced in paediatric clinical practice, medicines development, clinical research and project management, but not all team members work in comprehensive quality framework. Aim: To establish a well-documented, efficient quality system applying a new approach for ensuring quality in all trial aspects by combining existing organisation-related quality system elements of the project partners with newly developed SOPs and overarching, integrating trialspecific elements to ensure a reliable quality environment for the LENA Phase I clinical trial. Methods: Based on the network-structure of the project organisation, a strategy based on a team approach with joint responsibilities for the quality conduct of the project was pursuit, forming a QM Team consisting of the project leader, the leaders for pharmaceutical and clinical OTHER TOPICS development and an external quality expert. The team compiled a quality manual and an organisational chart displaying the sub-teams and their responsibilities. Another responsibility of the team was the integration of existing SOPs and Work Instructions as well as the creation of studyspecific procedures presented in “Manuals” and furthermore the verification of appropriate qualification of all staff involved in the project through CVs, job descriptions and training records. Results: For the Phase I study, a thorough analysis of all existing relevant SOPs and Work Instructions, forms and other quality elements was performed, uncovered trial-related processes were identified and a work plan was established to fill the gaps with the smallest possible number of newly developed organisation-related SOPs/Work Instructions and by preparing trial-specific process manuals. Demonstration of the trial team members was ensured by completing documentation concerning CVs, job description and training records. Among the sub-teams, the “GCLP” environment of the bioanalytical laboratory was started from scratch and could adequately support the LENA Phase I study by “GCLP” compliant quality work and sample logistics. Conclusion: The consortium’s approach enabled the preparation of a comprehensive, reliable GxP compliant quality system within a short timeframe and with the limited resources of a publicly funded project. Acknowledgments: The research leading to these results has received funding from the European Union Seventh Framework Programme (FP7/2007-2013) under grant agreement n°602295 (LENA). References: 1. GCP Inspectors Working Group, Reflection paper for laboratories that perform the analysis or evaluation of clinical trial samples (EMA/INS/GCP/532137/2010), 2012. These results have been presented at the ESDPPP Congress 2015. POS.208 State exam in pharmacy according to the German regulations for pharmacist training: Performance profiles in written (MCQ) exams and frequency of highest scores in part 1/3 POS.209 Pharmaceutical innovation in the early 20th century – Salvarsan and its derivatives Odenweller, S.1, Helmstädter, A.1 1 Institute of Pharmaceutical Chemistry, Goethe Universität Frankfurt, Max-von-Laue-Straße 9, 60438 Frankfurt am Main, Germany Arsphenamine (Salvarsan), developed by Paul Ehrlich and launched in 1910 on the german market, was the first drug with significant effect against the syphilitic disease [1]. It was the first drug of the so called “arsenophenamines” and the breakthrough innovation in this class. It was, however, not easy in handling, showed several side effects and faced severe criticism in public. This is why Paul Ehrlich and the Hoechster Farben AG continued to develop additional compounds of the same class. Between 1912 and 1931 nine derivatives reached the market: - Neosalvarsan (Neoarsphenamine, 1912) - Kupfer-Salvarsan (Copper Arsphenamine, 1914) - Salvarsan-Natrium (Arspenamine Sodium, 1915) - Sulfoxylsalvarsan (1919) - Silbersalvarsan (Silver Arsphenamine, 1920) - Neosilbersalvarsan (Neosilver Arsphenamine, 1920/21) - Myosalvarsan (Sulpharsphenamine, 1926) - Solusalvarsan (1931) - Isosalvarsan As a first step in a research project about pharmaceutical innovation in the 20th century, it was investigated which Salvarsan derivative, and why, eventually earned greatest success. As could be shown, Neoarsphenamine was the most successful compound preparation with a market share of 94% between 1927 and 1942 [2], although being neither the most potent nor the easiest to handle derivative in retrospective. It represented the first major improvement after Salvarsan had prepared the ground for chemotherapy of syphilis. References: 1. Helmstädter, A.: Pharmazeutische Zeitung 2010, 155: 4844-4851. 2. Rothermundt, M.: Zeitschrift für Hygiene und Infektionskrankheiten 1942, 124: 366-400. Shahla, H. 1; Spahn-Langguth, H.2 1 Institut für Medizinische und Pharmazeutische Prüfungsfragen – IMPP, Departments of Data Processing, Statistics, and Documentation Große Langgasse 8, D-55116 Mainz 2 Institut für Medizinische und Pharmazeutische Prüfungsfragen – IMPP, Departments of Pharmacy, Große Langgasse 8, D-55116 Mainz According to the regulations in Germany (AAppO), the pharmaceutical examination consists of 3 different segments, P1, P2, and P3. During the past 40 years, the regular 1st pharmacy state exam (P1) has been carried out as a cluster of four separate written MCQ tests in 4 different topics or subject groups (P1-I to -IV), including e.g., pharmacy-relevant basic natural sciences (physical science and life science), introductory drug formulation, and drug analysis*. Between 2400 and 2850 candidates per year (= numbers throughout the recent 15 years) are taking the written tests. Grades for the tests range from 1 (very good) to 5 (failed). Failure rates have been ranging from 3 to 26%. It is well-known that performance in these tests largely correlates with school grades of the students. The purpose of the current investigation was to evaluate the overall performance profiles over the past four decades and the predictive value of the MCQ tests’ outcome for the subsequent oral exams (= 2nd state exam P2 and 3rd exam P3). In order to generate an overview on the detailed results, the four P1 exams were summarized, and numbers given below include the arithmetical mean of all candidates in P1-I, -II, III, and -IV. The overall mean of the candidates’ P1 grades amounts to 3.13. Approximately five percent of the grades (5.11%) are above or at 2.0, and only 0.33% of all candidates reach the best possible grade in all four P1 tests (mean = 1.0). For P2 vs. P3, the respective numbers are as follows: arithmetical means of the overall grades, 2.43 vs. 2.53; fraction of candidates with grades >1.0 and <2.0, 32.8 vs. 31.5%; fraction with highest possible grade of 1.0, 2.51 vs. 7.2%. From grades obtained in P1, P2, and P3, an average grade is generally calculated according to the weighting scheme given in the AAppO (weighting factors P1 – P2 – P3: 2 – 3 – 2). The overall analysis yielded a very small group of students/candidates with an overall grade of 1.0 in P1-3 (i.e. grade/score of 1.0 in all single state exam parts), which is represented by a candidate fraction as small as approximately 0.07% on average. (*AAppO, Anlage 13) POS.210 Understanding Plasmodium falciparum’s exploitation of the innate immune system aids the identification of potential novel intervention strategies Schmidt, C. Q.1; Kennedy, A. T.2; Harder, M. J.1; Lim, N. Y. T.2; Tham, W. H.2 1 Institute of Pharmacology of Natural Products & Clinical Pharmacology, Ulm University, Ulm, Germany 2 Department of Medical Biology and The Walter and Eliza Hall Institute of Medical Research, The University of Melbourne, Parkville, Australia. For abstract see Short Poster Lectures SPL.012 page 109. POS.211 Ethnopharmacological information from the botanical correspondence of Berthold Seemann (1825-1871) – a pilot study Helmstädter, A.1 1 Institute of Pharmaceutical Chemistry, Goethe University, Max-von-Laue-Str. 9, 60438 Frankfurt, Germany Historical research may be able to contribute to the exploration of traditional knowledge about medicinal plants and promising attempts have been made investigating byzantine texts, early modern herbals, and writings of christian missionaries. In this pilot study it should be explored if publications, travel reports, diaries or correspondence of the botanical explorers of the 19th and early 20th centuries may serve a source of ethnopharmacological information as well and may be able to guide modern phytopharmacological research. Writings of Berthold Seemann (1825-1871), a german investigator exploring the botany of Middle DPhG Annual Meeting 2015 Conference Book • 193 POSTERS America, the Fiji islands and other regions, are investigated as a first example. It could be shown that Seemann’s heritage mainly kept at Kew Garden Archives, does contain ethnopharmacological information which in part has already been confirmed by recent study results indicating some reliability of his observations. However, there are also reports about traditional medicinal plants scarcely investigated so far, including Schultesia stenophylla Mart. (syn. S. guainensis (Aubl.) Malme), Trixis inula Crantz, Waltheria glomerata Presl., Gonophlebium attenuatum (Humb. & Bonpl. Es Willd) C. Presl., or Pseudoelephantopus spicatus (Juss ex Aubl.) C.F. Baker. It is suggested to further explore their potential as medicinal plants. In general, as Seemann’s example has shown, publications and correspondence of botanical explorers of the past seem to be a valuable and hitherto almost neglected source of information to be considered in further historical and ethnopharmacological research. POS.212 Displaced German pharmacist after World War II – The pharmacist Joseph Krause from East Prussia Schröder, J.1; Friedrich, C.1 Institute for the History of Pharmacy, Philipps University Marburg,Roter Graben 10, 35032 Marburg, Germany 1 The fate of the expulsion struck about 14 million Germans, who had to leave their region of origin in the former German territories and settlement areas in Central, Southeast and Eastern Europe. In the general refugee literature pharmacists are not addressed. Because of the existing system of concessions at that time no larger number of pharmacies could initially arise, despite the massively increased population due to the influx of millions of displaced persons. Displaced pharmacists were not substituted for their lost operating rights. The poster shows the professional positions in the curriculum vitae of the pharmacist Joseph Krause, who was born in 1906 in East Prussia and graduated in pharmacy at Wroclaw University in 1933. Since 1936, he was manager and later owner of a pharmacy in Olsztyn / East Prussia. Sent to the front in Italy as a soldier, he met his family again in April 1946 in Flensburg, after his captivity. Until 1952 he worked as a senior pharmacist in army hospitals, which served as hospitals for returnees from Russia. He got a leading position in a public pharmacy from 1952 to 1958. After the Federal Constitutional Court established the unrestricted freedom of pharmacy establishment as a legal principle on the 11th June 1958, Joseph Krause founded a pharmacy in Göttingen in 1958. POS.213 Professor Erika Cremer (1900–1996) – A pioneer in gas chromatography Michler, V.1; Friedrich, C.1 Institut für Geschichte der Pharmazie, Institute for the History of Pharmacy, Universität Marburg, University of Marburg, Roter Graben 10, 35037 Marburg, Germany 1 Professor Cremer is one of the real pioneers in gas chromatography: in fact, one can surely say that she is the real pioneer of this discipline. Despite all troubles during World War II, she conceived gas chromatography in 1944, when this technique did not yet exist. One may say that this work occurred after the 1941 publication of A.J.P. Martin (1910–2002) [2] and R.L.M. Synge (1914–1994) [3] on liquid-liquid partition chromatography, in which the possibility of using a gas as the mobile phase was predicted [4]. But however, it is important to note that during the war the issues of the Biochemical Journal, where Martin and Synge had published their results, were not available in Germany [4]. We would like to emphasize here that all activities from Erika Cremer happened years before James and Martin published the first report dealing with gas chromatography in 1952. Thus, the priority of professor Erika Cremer’s work in carrying out the first actual gas chromatographic experiments and doing it on her own intuition, definitely without any outside suggestions, is beyond any dispute. In this poster, we will present life and work of Erika Cremer. Looking finally back over more than sixty years to this exciting period when all these pioneering activities of chromatography were undertaken, it is 194 • DPhG Annual Meeting 2015 Conference Book amazing to see under what primitive conditions Professor Erika Cremer and all her students had to work. Scientists belonging to the post-war generation cannot imagine the hardships they had to face at that time. This fact still underlines more the importance of her work: even under these circumstances and difficulties she could achieve great findings and was one the first to publish gas-chromatographic results [5]. References: 1. Beneke, K.: Beiträge zur Geschichte der Kolloidwissenschaften 1999, 8: 311–334. 2. Adlard, E.: LC-GC Europe 2002, 15(9): 610–611. 3. Gordon, H.: Biographical Memoirs of the Fellows of the Royal Society 1996, 42: 455–479. 4. Martin, A.; Synge, R.: Biochemical Journal 1941, 35(12): 1359. 5. Bobleter, O.: Chromatographia 1996, 43(11/12): 581–582. 7. Ettre, L.S.: Chromatographia 1990, 29(9/10): 413–414. 8. Bobleter, O.: Chromatographia 1990, 30(9/10): 471–476. POS.214 Heinrich Hörlein – A life between science and economy Zummersch, M.1; Friedrich, C.1 Institut für Geschichte der Pharmazie der Philipps-Universität Marburg / Institute for the History of Pharmacy, University of Marburg, Roter Graben 10, 35032 Marburg, Germany 1 Heinrich Hörlein (1882–1954) belonged to the kind of chemists who were not contented with their research work in the industrial area, but also successful in the economic field. As chemist, physicist and economist he combined his natural-scientist expertise with specialist knowledge in economy which was imperative for his work in the industry, making him much better informed than he would have been if he had been restricted to the area of specialised chemistry in his long-term employment with the paint manufacturers, formerly Friedrich Bayer & Co. Furthermore, he supported the development of science by being active over decades in numerous scientific societies and advisory boards, serving the chemical-medical research, the support of German high schools, care for the young generation and scientific literature. Thanks to personal contacts and friendships with former university chemists and significant physicians along with insights into human nature, Hörlein was able to employ suitable staff for pharmaceutical research. Hörlein was active in creating the development of modern pharmaceutical chemistry and medication synthesis. The profiling of pharmaceutical research, especially the conversion of the Elberfeld laboratories to the biggest pharmaceutical research facility has to be listed as Hörlein’s life’s work. Acknowledgments: References and picture credits: Bayer AG Corporate History and Archives OTHER TOPICS AUTHOR INDEX A Abbas, Z. .................................................................................. 185 Abdel-Aziz, H. .................................................................. 62, 119 Abebe, D. ................................................................................. 178 Abrahamsson, B. .................................................................... 113 Abu Jhaisha, S. .......................................................................... 61 Acher F. C. .................................................................................. 33 Adami, M. ................................................................................ 148 Adams R. H.............................................................................. 104 Adamski, J................................................................................ 171 Affini, A. ................................................................................... 149 Ahrem, H.................................................................................. 124 Aigner, A. ................................................................................. 133 Akone, H. S. ................................................................................ 53 Alban, S. ..........................................................................153, 179 Alhazmi, H. A. ........................................................ 63, 138, 140 Alkhatib, Y............................................................................... 123 Alves Avelar, L. A. .................................................................. 103 Andermark, V. ........................................................................ 157 Andreas, J. O. .......................................................................... 187 Andrews, K. ............................................................................. 164 Ansideri, F. ................................................................................. 67 Apel, A. K. ................................................................................... 92 Arencibia, J. M. ....................................................................... 152 Aristotelous, T. .......................................................................... 35 Armbruster, M........................................................................ 127 Arntz, P..................................................................................... 129 Asare-Nkansah, S. .................................................................. 162 Asmari, M. ............................................................................... 138 Assaf, J...................................................................................... 141 Astigiano, S. ............................................................................... 69 B Bachmeier, B. E. ........................................................................ 69 Baghdan, E............................................................................... 126 Bakowsky, U. ..................................................................122, 126 Balzus, B. .................................................................................. 125 Bange, F.-C. .............................................................................. 174 Banoub, M. .............................................................................. 176 Barbieri, O. ................................................................................. 69 Barnscheid, L. ......................................................................... 115 Baron, R. ................................................................................... 115 Bartels, I. K. ............................................................................. 184 Bauer, S. M. ................................................ 155, 156, 161, 163 Baumann, K. ................................................................ 30, 61, 63 Bautista, O. .............................................................................. 147 Bayer, T..................................................................................... 153 Becher, K. ................................................................................. 142 Becker, A. ................................................................................... 61 Becker, W. .................................................................................. 61 Bednarski, P. J. .............................................................. 156, 157 Beheiri, S. N. ............................................................................ 183 Behnisch, S. ................................................................... 156, 157 Beißner, N. ............................................................................... 137 Bekeschus, S. ............................................................................. 90 Bekiesch, P. ................................................................................ 92 Bendas, G. ................................................................................ 102 Bering, L. .................................................................................. 174 Bermudez, M. .......................................................................... 147 Bertoletti, N. ............................................................................ 171 Bertrand, H.-O. .......................................................................... 33 Bertsche, T. ................................................................................ 37 Bibb, J. A. .................................................................................. 104 Biel, M. ........................................................................................ 14 Bierwisch, A. ............................................................................ 140 Binder, A. .................................................................................. 115 Biondi, R. M. ............................................................................ 152 Bischoff, F. ............................................................................... 155 Bischoff, I. ................................................................................ 178 Bitter, K. .............................................................................89, 186 Blankenbach, K. ...................................................................... 164 Blättermann, S. ....................................................................... 146 Bock, C. ..................................................................................... 167 Böckler, F. M. ............................................................................. 63 Bödefeld, T. ............................................................................. 144 Bodem, J. .................................................................................. 160 Bodmeier, R. ............................................................................ 125 Boeckler, F. M............................................................................ 67 Boesecke, P. ............................................................................... 99 Böger, R. H. .............................................................................. 116 Bollacke, A................................................................................ 142 Bonus, M. ................................................................................. 176 Bopp, B. ....................................................................................... 71 Börger, C................................................................................... 173 Borsig, L. ................................................................................... 102 Bouchut, A. .............................................................................. 164 Bouron, A. ................................................................................ 144 Brabet, I. ..................................................................................... 33 Bracher, F. .................................................. 138, 154, 173, 178 Braig, S...................................................................................... 120 Braun, A. ......................................................................... 123, 189 Braun, F..................................................................................... 171 Bräuner-Osborne, H. ................................................................ 34 Bravin, A. .................................................................................... 99 Breit, B. ....................................................................................... 74 DPhG Annual Meeting 2015 Conference Book • 195 AUTHOR INDEX Breitinger, H.-G....................................................................... 176 Breitinger, U. ........................................................................... 176 Breitkreutz, J. ................................................................... 13, 192 Brengel, C. ............................................................................... 173 Briel, D. ............................................................................145, 170 Brinkjost, T. ............................................................................. 162 Broich, K...................................................................................... 17 Bros, M. .................................................................................... 132 Brueck, S. ................................................................................. 115 Brümmer, A. ............................................................................ 190 Brüning, D................................................................................... 30 Bruno, A. ..................................................................................... 69 Brunschweiger, A...................................... 167, 168, 169, 190 Brust, P. .................................................................................... 170 Budde, P. ..................................................................................... 79 Buenemann, M. ...................................................................... 145 Bugain, O. ............................................................. 167, 168, 169 Büllesbach, K. ......................................................................... 147 Bunjes, H. .......................................................................... 56, 157 Burau, D. .........................................................................139, 186 Burckhardt, B. B. ................................................. 138, 183, 192 Busch, D. .................................................................................. 115 Buschauer, A. .................................................................120, 149 Busserolles, J. ............................................................................. 33 C Calderón, M............................................................................. 125 Calin, M. ................................................................................... 102 Cardinaux, J.-R. ....................................................................... 145 Carrillo García, C....................................... 117, 121, 175, 176 Cascorbi, I................................................................................ 115 Castro Diez, C. ........................................................................ 188 Cawello, W. ............................................................................. 187 Chaikuad, A. ............................................................................... 61 Chakrabarti, A. ..............................................................151, 153 Chakrabarti, I. ......................................................................... 153 Chamrad, D. ............................................................................... 79 Charbaji, N. ............................................................................. 125 Cheung, S.-Y. .......................................................................... 180 Chevillard, F. .............................................................................. 35 Ciglia, E. ............................................................................ 71, 151 Ciplea, A. M. ............................................................................ 192 Clement, B. ................................................. 140, 155, 170, 172 Csordás, K................................................................................ 133 Czarnecki, K. .............................................................................. 38 Czupalla, C. J. ............................................................................. 27 D Dai, B......................................................................................... 178 Daletos, G.................................................................................... 54 Dallanoce, C. ........................................................................... 144 De Amici, M. ........................................................................... 144 196 • DPhG Annual Meeting 2015 Conference Book de Mello Martins, A. G. ......................................................... 173 de Souza Carvalho-Wodarz, C. ........................................... 135 Decker, C. ................................................................................. 159 Decker, H. ..........................................................................99, 134 Decker, S. ................................................................................... 74 Derix, S. ...................................................................................... 39 Dersch, P................................................................................... 163 Devraj, K. .................................................................................... 27 Dewaldt, M............................................................................... 123 Dey, S. ....................................................................................... 144 Diederich, W. .......................................................................... 160 Diedrich, D. ............................................................. 76, 103, 154 Dierks, C. .................................................................................... 74 Dietzel, A. ................................................................................. 137 Dietzel, D. ................................................................................. 132 Dimde, M.................................................................................. 125 Dobner, B. ...................................................................... 165, 166 Döring, E. .................................................................................. 156 Dörje, F. .................................................................................... 185 Dormann, H. ............................................................................ 185 Doroz-Płonka, A. ........................................................... 174, 175 Drabczyńska, A. ...................................................................... 174 Drews, G. .................................................................................... 29 Drückes, P. ............................................................................... 156 Du, W. J. ................................................................................... 130 Duburs, G. ................................................................................ 168 Düfer, M. .................................................................................... 31 Duque Escobar, J. ................................................................... 116 Dzikowski, R. ................................................................. 160, 169 E Eberle, J..................................................................................... 144 Ebert, R. .................................................................................... 189 Efferth, T. ................................................................................... 70 Ehmann, L. ............................................................................... 186 Ehrig, K. .................................................................................... 153 Ehrt, C. ...................................................................................... 162 Eichner, A. ...................................................................... 165, 166 Einsle, O...................................................................................... 74 Eisend, S. .................................................................................. 188 El Deeb, S. ........................................... 63, 138, 139, 140, 141 Elgaher, W. A. ......................................................................... 161 Elsinghorst, P. W. ................................................................... 140 Elz, S. ......................................................................................... 149 Empting, M. ................................................................... 163, 173 Engel, M.................................................................................... 152 Engelhardt, B. .......................................................................... 164 Engelhardt, K. H. .................................................................... 126 Engelke, L. H. ........................................................................... 152 Erdmann, F. ............................................................................. 159 Erdogmus, S. ............................................................................. 46 Eriksson, P. O. ......................................................................... 115 Eschalier, A. ............................................................................... 33 Eschenhagen, T....................................................................... 145 AUTHOR INDEX Esteban, G. ............................................................................ 149 Exner, T. ....................................................................................63 F Falke, H. ....................................................................................61 Fallegger, D. .......................................................................... 164 Fedorov, O. ............................................................................ 154 Felsch, M. .................................................................................38 Fenoy, C. G. ........................................................................... 190 Ferger, B. ............................................................................... 117 Fiene, A. ................................................................................. 146 Fiori, M. E. ................................................................................69 Fischer, D. .....................................................................123, 124 Fischer, K. .............................................................................. 180 Fischer, S. .............................................................................. 156 Flath, T. .................................................................................. 135 Fleischmann, B. .................................................................... 117 Flögel, U. ................................................................................ 125 Flötgen, D. ....................................................................168, 175 Forchhammer, K. ....................................................................92 Forster, M. ............................................................................. 163 Franz, L. ................................................................................. 170 Fricker, G. .................................................................................28 Friedland, K. .................................................................144, 185 Friedrich, C...................................................................192, 194 Frieg, B. .........................................................................166, 177 Fritz, N. .................................................................................. 144 Fröhner, W. ........................................................................... 152 Fromm, M.............................................................................. 185 Fruth, M. ................................................................................ 163 Fujiwara, R. ..............................................................................44 Fujiwara, T. ........................................................................... 178 Funke, M. ............................................................................... 146 Fürst, R. ............................................................... 119, 120, 178 Fürtig, M.-A. .................................................................139, 186 G Garidel, P. .............................................................................. 122 Garscha, U. ......................................................... 107, 178, 179 Gawron, S. ................................................................................35 Geertz, B. ............................................................................... 145 Gehringer, M...................................................... 155, 161, 163 Geisslinger, G...........................................................................80 Gellert, A................................................................................ 160 Gerber, A. P. .......................................................................... 190 Gerhardt, H. .............................................................................27 Gerhardt, S. ..............................................................................74 Gershkovich, M. ................................................................... 159 Gersonde, F. .......................................................................... 188 Gerstmeier, J. ...............................................................107, 178 Gertzen, C. G. W. ........................................................ 103, 147 Giaisi, M. ...................................................................................68 Gilbert, I. H. ............................................................................. 35 Girreser, U. ......................................................... 140, 170, 172 Giurg, M. ................................................................................ 172 Glatzel, A. ............................................................................... 116 Glatzel, D................................................................................ 119 Glen, R. C. ................................................................................ 85 Gloriam, D. E. .......................................................................... 34 Goebgen, E. B. ............................................................. 184, 192 Goerigk, G. ............................................................................... 99 Gohlke, H. ...... 49, 71, 103, 147, 151, 165, 166, 167, 173, 176, 177 Gohr, K. .................................................................................. 152 Göke, K. .................................................................................. 157 Gollos, S. ................................................................................ 146 Gomeza, J. .............................................................................. 146 Gopireddy, S. R........................................................... 132, 136 Görg, B.................................................................................... 166 Goudet, C. ................................................................................ 33 Grabow, N. ............................................................................ 100 Grapentin, C. ......................................................................... 125 Greber, B. ............................................................................... 176 Gressenbuch, M. ................................................................... 135 Griebel, L................................................................................ 185 Grohganz, H. ......................................................................... 130 Groll, A. H. ............................................................................. 187 Gross, J. .................................................................................. 122 Grote, S. ................................................................................. 131 Groth, G.................................................................................... 71 Grundmann, M. ...................................................................... 65 Grünefeld, J. ................................................................ 160, 169 Gudd, J.................................................................................... 172 Gudermann, T. ........................................................................ 46 Guennewig, B. ....................................................................... 190 Guerit, S. .................................................................................. 27 Günter, S. ................................................................................. 74 Günther, M. ........................................................................... 156 Gutknecht, S. ......................................................................... 113 Gutmann, M. ......................................................................... 189 Gütschow, M. ........................................... 140, 147, 170, 172 H Haag, R. .................................................................................. 125 Haake, N................................................................................. 188 Hacker, M................................................................................. 57 Hacker, M............................................................................... 135 Haenisch, S. ........................................................................... 115 Hahne, T................................................................................. 112 Hall, J. ..................................................................................... 190 Halver, J. ................................................................................. 121 Hamacher, A.................................... 103, 151, 152, 156, 169 Hamacher, S. ........................................................................... 38 Hanke, F. .................................................................................. 40 Hanke, T. ................................................................................ 180 Hansen, F. K. .................. 76, 103, 151, 152, 154, 156, 169 DPhG Annual Meeting 2015 Conference Book • 197 AUTHOR INDEX Harbaum, L.............................................................................. 116 Harder, M. J. ............................................................................ 109 Hartmann, R. W. ....................................... 161, 163, 171, 173 Hasenpusch, D. ....................................................................... 116 Hasse, S. ...................................................................................... 90 Haupenthal, J. ......................................................................... 161 Hauser, A.-T. ........................................................................... 153 Häussinger, D. ............................................. 16, 147, 166, 176 He, D. ........................................................................................ 112 Hedtrich, S......................................................................125, 181 Heidrich, J. .................................................................................. 67 Heimburg, T. ........................................................................... 153 Heine, A.................................................................................... 171 Heinemann, S. ........................................................................... 42 Heinrich, M.............................................................................. 144 Heisig, F. .................................................................................. 146 Helfert, S. M............................................................................ 115 Hellmann, N. .................................................................... 99, 134 Hellmich, U. ............................................................................. 160 Hellwig, M. .............................................................................. 101 Helmer, R. ................................................................................ 160 Helmstädter, A. .............................................................119, 193 Hempel, G. ............................................................ 142, 143, 187 Hendrikx, S. ............................................................................. 135 Hennen, S. ............................................................................... 146 Hensel, A. ................................................................................. 120 Heppner, H. J........................................................................... 184 Herdewijn, P............................................................................... 18 Herich, L. ..................................................................................... 38 Hermans, S. M. A. ..................................................................... 49 Herrmann, J. ............................................................................ 163 Hess, C. ..................................................................................... 117 Hesse, M................................................................................... 117 Heuer, G. .................................................................................. 186 Hildebrand, J. ............................................................................. 40 Hildebrandt, C. ....................................................................... 132 Hilger, R. A. ............................................................................. 101 Hillenbrand, M. ......................................................................... 36 Hinz, S. ............................................................................145, 174 Hirt, M. ..................................................................................... 168 Hobernik, D. ............................................................................ 132 Hoeppner, A. ........................................................................... 177 Höfer, H. H. ............................................................................. 130 Hoffmann, C. ............................................................................. 46 Hofmann, B. ...................................................................180, 181 Holloway, S. ............................................................................ 160 Hölscher, C. ............................................................................. 174 Holzgrabe, U. .................................................................144, 176 Homeyer, N. ...................................................................165, 166 Hongwiset, D. ......................................................................... 174 Hönzke, S. ................................................................................ 181 Hopkins, A. L....................................................................... 35, 83 Hopkins-Navratilova, I. ............................................................ 35 Hudson, B. D. ............................................................................. 65 Hügle, M...................................................................................... 74 Humbeck, L. ............................................................................ 163 198 • DPhG Annual Meeting 2015 Conference Book Huschmann, F. U. ................................................................... 171 Huwiler, A. ............................................................................... 164 I Imeri, F...................................................................................... 164 Imig, J. ....................................................................................... 190 Immer, M. ................................................................................ 162 Ingebrandt, S. ......................................................................... 123 Itoh, T.......................................................................................... 44 J Jaehde, U. ........................................................ 40, 89, 101, 186 Jakob, F. .................................................................................... 189 Jensen, K. T. ............................................................................. 130 Jin, N.......................................................................................... 130 John, C......................................................................................... 38 Johnson, R. ........................................................................99, 134 Jones, P. G. ................................................................................. 61 Jose, J ......................................................................................... 120 Jose, J. ....................................................................... 71, 142, 190 Jung, K....................................................................................... 168 Jung, M. ................................................................ 151, 153, 160 Juntke, J. ................................................................................... 135 K Kalayda, G. V. .......................................................................... 101 Kaleta, M. ................................................................................. 175 Kalliokoski, T. .......................................................................... 167 Kalscheuer, R............................................................................. 53 Kamińska, K. ............................................................................ 148 Kandil, R. .................................................................................. 178 Kanitz, M. ................................................................................. 160 Kankowski, S. .......................................................................... 157 Karcz, T. ................................................................ 146, 174, 175 Karkossa, F. .............................................................................. 113 Karow, A. R. ............................................................................. 122 Kascholke, C. ........................................................................... 135 Kassack, M. U. .......76, 103, 151, 152, 154, 156, 169, 174 Kassel, S.................................................................................... 150 Keck, C. M. ........................................................... 123, 129, 130 Keiser, M. ................................................................................. 115 Keitel, V. ......................................................................... 147, 166 Kelber, O. ..........................................................................62, 119 Kenakin, T. ................................................................................. 65 Kennedy, A. T.......................................................................... 109 Kessel, E.................................................................................... 112 Keßler, S. .................................................................................... 93 Khalife, J. .................................................................................. 164 Khalil, F. .......................................................................... 183, 188 Khan, N. .................................................................................... 148 AUTHOR INDEX Khosa, S. .................................................................................. 177 Khoshakhlagh, P. ................................................................... 134 Kieć-Kononowicz, K. ...................... 146, 148, 164, 174, 175 Kiefer, W. ................................................................................ 160 Killian, P. H. ................................................................................ 69 Kindgen, S. .............................................................................. 113 Kirchhefer, U........................................................................... 187 Kirchmair, J. ............................................................................... 85 Kishore, S................................................................................. 190 Klebe, G. ..........................................................................171, 172 Klein, C. D. .................................................................................. 51 Klein, P. .................................................................................... 112 Klein, S. .................................................................................... 113 Kleine, K. .................................................................................. 192 Kleinebudde, P. ............................... 127, 130, 131, 133, 134 Kleinschrodt, D. ...................................................................... 177 Klemm, D. ................................................................................ 124 Klika Škopić, M. ..................................................................... 169 Klingler, F. ............................................................................... 159 Klingmann, I. .......................................................................... 192 Kloft, C. ............................................. 139, 141, 184, 186, 192 Klose, H. ................................................................................... 116 Knapp, S..................................................................... 59, 61, 154 Knop, K. ...........................................................................127, 130 Köberlein-Neu, J. ....................................................................... 38 Koburg, M................................................................................ 155 Koch, M. ...................................................................................... 66 Koch, O........................................................ 162, 163, 168, 174 Koch, P............................................................................... 67, 156 Koeberle, A. ............................................................................. 119 Köhler, R. .................................................................................... 68 Kolb, P. ........................................................................................ 35 Kolter, M. .................................................................................... 28 König, S. ...................................................................................... 98 Köppler, J. ................................................................................ 156 Korp, J. ......................................................................................... 98 Köse, M. ..........................................................................146, 174 Kostenis, E. ............................................................. 65, 146, 147 Kottke, T. ................................................................................. 150 Kotz, S. ..................................................................................... 101 Krafczyk, K. ............................................................................. 192 Krainitzki, L. ............................................................................ 132 Kralisch, D. .....................................................................123, 124 Kramer, D. .................................................................................. 94 Kramer, F. ................................................................................... 78 Krämer, W. .............................................................................. 125 Krammer, H. P. .......................................................................... 68 Krebs, L. ...................................................................................... 99 Kremer, M................................................................................ 127 Kretschmer, S. B. M. ....................................................180, 181 Kronski, E. ................................................................................... 69 Krüger, A.................................................................................. 113 Krüger, M.......................................................................... 39, 186 Kubas, B. .................................................................................. 174 Kubbutat, M. H. G.........................................................161, 162 Kubicova, L. ............................................................................. 145 Kuder, K. ................................................................................... 164 Kulick, M. ................................................................................... 89 Kullmann, M. ........................................................................... 101 Kunick, C. ............................................ 61, 160, 161, 162, 169 Kunschke, N............................................................................. 124 Kunze, T.................................................................................... 188 Küppers, J. ................................................................................ 172 Kurz, T. ............................ 76, 103, 151, 152, 154, 156, 169 L Lächelt, U. ................................................................................ 112 Läer, S. ................................................. 88, 138, 183, 188, 192 Lalk, M. ....................................................................................... 90 Lancelot, J................................................................................. 153 Lande, D. H. ............................................................................. 169 Lang, M. .......................................................................... 145, 170 Lange, A. ..................................................................................... 67 Lange, S. ......................................................................... 165, 166 Langer, K. ................................ 122, 126, 127, 128, 129, 190 Langguth, P. .................................................. 99, 113, 132, 134 Längle, D......................................................................... 168, 175 Langner, A. ..................................................................... 165, 166 Lappe, S. ................................................................................... 122 Laufer, S. ........................................................................ 155, 161 Laufer, S. A. ......................................................... 156, 163, 165 Lautscham, G............................................................................. 79 Law, J. K. Y. .............................................................................. 123 Łażewska, D. ............................................................................ 164 Le Borgne, M. .......................................................................... 142 Le Duc, G. ................................................................................... 99 Lee, D. J. .................................................................................... 112 Lehmann, W. D. ...................................................................... 140 Lehr, C. M....................................................................... 124, 135 Lehr, T. ...................................................................................... 104 Lehrnbecher, T........................................................................ 187 Lehto, T..................................................................................... 112 Lemcke, T. ...................................................................... 116, 159 Lemmerhirt, C. ........................................................................ 167 Lemmerhirt, H............................................................... 156, 157 Lengers, I. ................................................................................. 120 Lenz, E. ...................................................................................... 130 Li, T. ........................................................................................... 140 Li, Y............................................................................................ 178 Liebich, M. ............................................................................... 143 Liebl, J. ...................................................................................... 104 Liebner, S. .................................................................................. 27 Liening, S. ................................................................................ 179 Liewert, I. ................................................................................. 153 Lillig, C. H. ................................................................................ 156 Lim, N. Y. T.............................................................................. 109 Link, A. ...................................................................................... 167 Listing, M. ................................................................................ 180 Litty, F.-A. ................................................................................. 172 Li-Weber, M............................................................................... 68 DPhG Annual Meeting 2015 Conference Book • 199 AUTHOR INDEX Loaëc, N. ........................................................................... 61, 160 Löbmann, K. ............................................................................ 130 Loretz, B. .................................................................................. 124 Lozach, O. ................................................................................... 61 Lucas, X. ...................................................................................... 74 Lüdeke, S. ......................................................................... 76, 154 Ludwig, S .................................................................................... 24 Lühmann, T. ...................................................................108, 189 Lüneburg, N. ........................................................................... 116 Lunter, D. .................................................................................... 58 Luong, B. .................................................................................. 120 Lutz, S....................................................................................... 145 M Maas, R. .................................................................................... 185 Maček, B...................................................................................... 92 Madea, B. ................................................................................. 117 Maier, J. .................................................................................... 156 Makowski, N. .......................................................................... 188 Marchais-Oberwinkler, S. .................................................... 171 Marek, M.................................................................................. 153 Martin, P. ................................................................................. 115 Masur, K. ..................................................................................... 90 Mattern, K................................................................................ 137 Matz, M. ...................................................................................... 30 Maul, K. J. ................................................................................ 112 Maurer, C. K. ........................................................................... 163 Mayer-Wrangowski, S. C. .................................................... 165 Mederos y Schnitzler, M......................................................... 46 Meier, C. ................................................................................... 155 Meier, R. ................................................................................... 134 Meijer, L. ................................................................. 61, 160, 173 Meinel, L. .............................................................. 108, 128, 189 Melesina, J. .............................................................................. 153 Mély, Y. .................................................................................... 161 Melzig, M. F............................................................................. 120 Memmel, E. ............................................................................. 189 Menche, D................................................................................ 120 Mennemann, H. S. .................................................................... 38 Merk, D. ...........................................................................106, 159 Merk, H..................................................................................... 104 Merkel, O. M. .......................................................................... 178 Merten, N................................................................................. 146 Mertens, M. D. ........................................................................ 140 Mertens-Keller, D. ..................................................................... 38 Mesken, J. ................................................................................ 129 Messerer, R.............................................................................. 144 Metz, A. .................................................................................... 171 Metzger, S. .............................................................................. 101 Meyer zu Heringdorf, D. ...................................................... 164 Meyer, A..........................................................................146, 157 Miceli, E.................................................................................... 125 Milligan, G. ................................................................................. 65 Minichmayr, I. K. ................................................................... 186 200 • DPhG Annual Meeting 2015 Conference Book Mirisola, V.................................................................................. 69 Mittal, N. .................................................................................. 190 Mohammadi, M. ..................................................................... 178 Mohr, K. .............................................................. 48, 64, 65, 144 Möller, G. .................................................................................. 171 Monsuur, F............................................................................... 130 Mootz, H. D. ............................................................................. 142 Morhenn, K. ............................................................................. 145 Moritz, S. .................................................................................. 123 Moskovits, J. ............................................................................ 187 Mozafari, M. ........................................................ 139, 140, 141 Mueller, U................................................................................. 171 Müller, C. ........................................................................ 138, 187 Müller, C. E. ................ 12, 62, 81, 117, 145, 146, 174, 175 Müller, D................................................................................... 174 Müller, R. .................................................... 119, 120, 129, 163 Müller, R. H. ............................................................................. 130 Müller, W. W. ........................................................................... 68 Müller-Fahrnow, A. .................................................................. 60 Müller-Goymann, C. C........................................................... 122 Mulnaes, D. .................................................................... 167, 177 München, S. E. ........................................................................ 187 Muth, F. .................................................................................... 156 N Nachbar, M. ............................................................ 63, 140, 141 Nachtigall, A. ........................................................................... 184 Nägele, E. ................................................................................... 45 Nasereddin, A. .............................................................. 160, 169 Nassut, R. ................................................................................. 129 Nawroth, T.............................................................. 99, 113, 134 Neimanis, S. ............................................................................ 152 Nerlich, A. G. ............................................................................. 69 Nett, M. ....................................................................................... 98 Neubert, R. H. H. .......................................................... 165, 166 Newcomer, M.......................................................................... 178 Neyts, J........................................................................................ 25 Nieber, K. ..........................................................................62, 119 Niedermeyer, T......................................................................... 94 Nienberg, C.................................................................... 142, 190 Noonan, D. M. ........................................................................... 69 Nowak, G. ................................................................................. 144 O Obst, K. ..................................................................................... 125 Odadzic, D................................................................................ 152 Odenweller, S.......................................................................... 193 Oetjen, E. ................................................................. 32, 116, 145 Ohta, S. ....................................................................................... 43 Okpanyi, S................................................................................ 119 Olejarz, A.................................................................................. 174 Olsen, C. A. ................................................................................ 75 AUTHOR INDEX Oltmann-Norden, I. ........................................................ 63, 112 Orlando, Z. .............................................................................. 120 Ortmann, R................................................................................. 77 Ostrovsky, D............................................................................... 74 Oswald, S. ................................................................................ 115 Ott, I. ......................................................................................... 157 Otter, M.................................................................................... 115 Ouald-Chaib, A. ......................................................................... 71 P Pagani, A. .................................................................................... 69 Pagani, G. ................................................................................. 165 Page, S. ........................................................................................ 55 Palus, J. ..................................................................................... 172 Parnham, M................................................................................ 80 Parr, M. K. ............................................................... 45, 138, 141 Paulsen, D. .................................................................................. 21 Pehe, C...................................................................................... 186 Pérez Gago, A. ........................................................................ 131 Peter, K. .................................................................................... 125 Peters, O................................................................................... 138 Pfaffenrot, E. ........................................................................... 155 Pfeffer, U. ................................................................................... 69 Pfeifer, A. .................................................................................... 81 Pfeilschifter, J. ........................................................................ 164 Pfleger, C. ......................................................................... 49, 173 Pieper, S. .........................................................................126, 187 Pierce, R. .................................................................................. 153 Pierrot, C. ................................................................................. 164 Pietschmann, T.......................................................................... 20 Pin, J.-P......................................................................................... 33 Pinnapireddy, S. ..................................................................... 126 Plate, K. H. .................................................................................. 27 Platzer, C. ................................................................................. 159 Plaza, A. .................................................................................... 173 Plückthun, A. .............................................................................. 36 Pockes, S. ................................................................................. 149 Polier, G. ...................................................................................... 68 Polosek, P. ................................................................................ 170 Popp, A. T. ............................................................................... 154 Porazik, C. ................................................................................ 117 Pötzinger, Y............................................................................. 124 Praefke, B. A. ........................................................................... 155 Preis, M. ................................................................................... 132 Pretzel, J. .................................................................................. 174 Preu, L......................................... 61, 140, 160, 161, 162, 169 Probst, M. ................................................................................ 136 Prochnicka, A. ......................................................................... 157 Prokopets, O. S. ...................................................................... 145 Prokosch, H. U. ....................................................................... 185 Proksch, P. .................................................................................. 53 Proschak, E. .......................................................... 105, 152, 159 Punčochová, K. ....................................................................... 134 Pyo, S. M. ................................................................................. 129 Q Quinke, R. ................................................................................. 186 R Rabel, M.................................................................................... 124 Rach, R. ..................................................................................... 113 Rades, T. ................................................................................... 130 Rahnfeld, L. .............................................................................. 124 Rasool, M. F. ............................................................................ 183 Rathmer, B. .............................................................................. 176 Raudszus, B.................................................................... 128, 190 Rauh, D. .............................................................................72, 165 Raulf, A. .................................................................................... 117 Redweik, S. .............................................................................. 140 Rehberg, N. ................................................................................ 53 Reichl, S. ............................................................... 135, 137, 157 Reimer, M................................................................................. 115 Reiss, G. J.................................................................................. 103 Rempel, V. ................................................................................ 175 Rengelshausen, J. ................................................................... 115 Richling, I. .................................................................................. 38 Rickmeyer, T. .......................................................................... 172 Ridder, S. .................................................................................. 127 Rigault, D.................................................................................... 33 Ritschel, T. ................................................................................. 84 Ritzer, J. .................................................................................... 189 Robaa, D. ........................................................................ 160, 164 Roblek, M. ................................................................................ 102 Rödl, C. B. ....................................................................... 180, 181 Rodrigues Moita, A. J. .......................................... 76, 151, 154 Romier, C. ................................................................................ 153 Rose, O........................................................................................ 38 Rothbauer, U. .......................................................................... 156 Rötzer, K. M. ............................................................................ 136 Rovera, F..................................................................................... 69 Rubbiani, R. ............................................................................. 157 Rübsamen-Schaeff, H.............................................................. 23 Rudo, A. .................................................................................... 174 Rustenbeck, I. ............................................................................ 30 Rüther, A. ..........................................................................76, 154 S Saaber, D. ................................................................................. 135 Sadek, B. ................................................................................... 148 Sanoh, S. .................................................................................... 43 Sauer, B........................................................................... 160, 164 Saur, O. ..................................................................................... 150 Sayle, S. .................................................................................... 122 Schäberle, T. F. ......................................................................... 95 Schächtele, C. ................................................................ 161, 162 Schächtele, S. .......................................................................... 185 DPhG Annual Meeting 2015 Conference Book • 201 AUTHOR INDEX Schade, D. ............................... 117, 121, 168, 172, 175, 176 Schaefer, C. ............................................................................. 187 Schaefer, J. .............................................................................. 138 Schaefer, K. H. ........................................................................ 123 Schäfer-Korting, M. ............................................................... 181 Schaffert, C. ............................................................................... 38 Schäke, F. ................................................................................. 145 Schaller, D. .............................................................................. 148 Scharf, R. E. ............................................................................. 165 Scherließ, R. ...................................................................129, 132 Scherneck, S. ............................................................................. 87 Scheunemann, M. .................................................................. 170 Schieback, P. ........................................................................... 159 Schiebel, J. ............................................................................... 171 Schieferdecker, S. ..................................................................... 98 Schipke, C. ............................................................................... 138 Schirmeister, T. ............................................................... 50, 160 Schlesinger, M. ....................................................................... 102 Schlosser, E. ............................................................................ 136 Schlütke, L. .............................................................................. 161 Schmidberger, H....................................................................... 99 Schmidt, A. .......................................................................... 45, 90 Schmidt, C. .............................................................................. 157 Schmidt, C. Q. ........................................................................ 109 Schmidt, J. ............................................................................... 159 Schmidt, M. ................................................ 136, 153, 159, 164 Schmidtkunz, K. ..................................................................... 153 Schmueser, L. ......................................................................... 134 Schneefeld, M......................................................................... 174 Schneider, G............................................................................... 86 Schneider, J. ............................................................................ 140 Schneider, M.............................................................................. 79 Schneider, T. ........................................................................... 153 Schneider-Daum, N. .............................................................. 135 Schoeder, C. ...................................................................117, 175 Schoenfeld, A.-K. ................................................................... 179 Scholz, M. S. ........................................................................... 180 Scholz, P................................................................................... 123 Schönherr, R. ............................................................................. 42 Schöppe, J................................................................................... 36 Schori, C...................................................................................... 36 Schrader, J. .............................................................................. 125 Schrage, R......................................................................... 64, 144 Schröder, J. .............................................................................. 194 Schröder, R.............................................................................. 147 Schröpf, S. ............................................................................... 186 Schubert, R.............................................................................. 125 Schubert-Zsilavecz, M. ................................................159, 180 Schulte, M. .............................................................................. 152 Schulz, D. ................................................................................. 183 Schulze, J. .......................................................................133, 152 Schulze, P. ............................................................................... 135 Schulze, T. ............................................................................... 141 Schulz-Fincke, A. C. ............................................................... 170 Schulzke, C. ............................................................................. 157 Schulz-Knappe, P. ..................................................................... 79 202 • DPhG Annual Meeting 2015 Conference Book Schulz-Siegmund, M. ............................................................ 135 Schumacher, K. ......................................................................... 30 Schumann, D. .......................................................................... 135 Schuster, D............................................................................... 180 Schwab M. ................................................................................. 82 Schwalm, S. ............................................................................. 164 Schwed, J. S. .................................................................. 148, 149 Schweins, R. .............................................................................. 99 Scriba, G. ...........................................................................42, 179 Seemann, W. K. ........................................................................ 48 Seibel, J. .................................................................................... 189 Seidlitz, A. ...................................................................... 100, 136 Sendker, J. .................................................................................. 91 Senz, V. ..................................................................................... 100 Shahla, H. ................................................................................. 193 Sharma, K................................................................................. 161 Siegmund, W. ......................................................................... 115 Simionescu, M. ....................................................................... 102 Simon, K. .................................................................................. 146 Sippl, W.............................................151, 153, 159, 160, 164 Skarżewski, J. .......................................................................... 172 Škopić, K. M. ........................................................................... 167 Smith, N. J.................................................................................. 65 Smits, H. J. S............................................................................ 177 Sommerfeld, A........................................................................ 176 Sommerhoff, C. P..................................................................... 69 Sonnenberger, S. .......................................................... 165, 166 Spahn-Langguth, H. ............................................................... 193 Spanier, C................................................................................. 146 Spanier, L. ................................................................................ 151 Špatenková, L. ......................................................................... 192 Spieler, V. ................................................................................. 108 Spomer, L. ................................................................................ 147 Stan, D. ..................................................................................... 102 Stanic, M. ................................................................................... 45 Stark, H. ......................... 148, 149, 150, 152, 164, 180, 181 Staufenbiel, S.......................................................................... 130 Steckel, H. ................................................................................ 129 Steiger, C. ................................................................................. 128 Steinhilber, D. .............................................. 80, 159, 180, 181 Steinicke, F. ......................................................................63, 112 Stenzel, K. ................................................................................ 169 Štěpánek, F. ............................................................................. 134 Stephan, C.................................................................................. 22 Steuber, H. ............................................................................... 160 Stoiber, K. ................................................................................ 120 Stoldt, V. R............................................................................... 165 Storch, U. ................................................................................... 46 Strödke, B. ................................................................................ 178 Strohalm, M............................................................................... 94 Strohmann, C. ......................................................................... 168 Stühler, K. .................................................................................. 41 Subramanian, D...................................................................... 148 Syntschewsk, V. ........................................................... 103, 156 Szekely, N. K. .......................................................................... 134 AUTHOR INDEX T Tallant, C.................................................................................. 154 Tateno, C. ................................................................................... 43 Täuber, A. ................................................................................ 122 Telaar, A. ..................................................................................... 79 Temme, S. ............................................................................... 125 Temml, V. ................................................................................ 180 Teubel, J. .................................................................................. 138 Thabet, Y. ................................................................................ 132 Tham, W. H............................................................................. 109 Thimm, D. ................................................................................ 146 Thoma, F. ................................................................................. 127 Thomann, A. ........................................................................... 173 Thürmann, P. A. .............................................................. 40, 184 Thurmond, R. L. ........................................................................ 15 Tietz, K. .................................................................................... 113 Tikhomirov, A. ........................................................................ 170 Tikhonova, I. G. ......................................................................... 65 Tins, J. ..............................................................................138, 183 Titz, A. ......................................................................................... 52 Totzke, F. ........................................................................161, 162 Trappe, J. ................................................................................. 156 Tremmel, R................................................................................. 26 Tremmel, T. ............................................................................. 173 Tschammer, N. .......................................................................... 47 Tsikrika, P. ............................................................................... 190 Tukey, R. H. ................................................................................ 44 Türeli, N. G. ............................................................................. 135 Tyzack, J. D................................................................................. 85 U Ulven, T. ...................................................................................... 65 Unzeta, M. ............................................................................... 149 Urbanetz, N. A. ..............................................................132, 136 V Ventura Pereira, J. P. ............................................................. 165 Villmann, C. ............................................................................ 176 Vogt, D. ...........................................................................180, 181 Völler, S.................................................................................... 183 Vollmar, A. M. ..................................................... 104, 120, 155 von Hammerstein, F.............................................................. 160 von Woedtke, T. ....................................................................... 90 Vordenbäumen, S..................................................................... 79 Voß, U. .............................................................................. 62, 119 Vukosavljevic, B. .................................................................... 134 W Wagner, E. ............................................................................... 112 Wagner, S. ............................................................................... 170 Wagner, T. ............................................................................... 160 Wahl, M. A. .............................................................................. 127 Wähnert, D. ............................................................................. 183 Wallmeyer, L. .......................................................................... 181 Walter, A. ....................................................................... 153, 160 Walter, M. ...................................................................... 148, 149 Walter, N.................................................................................. 165 Waltering, I................................................................................ 38 Wang, X.................................................................................... 125 Wätzig, H. ..................................................... 63, 112, 139, 140 Weber, J.................................................................................... 142 Weidel, E. ................................................................................. 173 Weidner, T. .............................................................................. 160 Weiser, C. ................................................................................. 141 Weiss, M. S. ............................................................................. 171 Weitschies, W............................................................... 100, 136 Weizel, L................................................................................... 148 Wende, K. .................................................................................. 90 Wentsch, H. K. ........................................................................ 165 Wentzlaff, M. .......................................................................... 100 Werner, T. ................................................................................ 168 Werner, V. ............................................................................... 108 Werz, O. .................................... 98, 107, 119, 178, 179, 180 Wesselborg, S. .......................................................................... 73 Wicha, S. G. ............................................................................. 192 Więcek, M. ............................................................................... 148 Wille, T. .................................................................................... 140 Willems, E. ............................................................................... 121 Windbergs, M. ........................................................................ 134 Witting, A. ............................................................................... 117 Wohlwend, D. ........................................................................... 74 Wolber, G. ...................................................................... 147, 148 Wölfel, S................................................................................... 162 Wölker, J................................................................................... 138 Woltersdorf, S. ............................................................. 180, 181 Wu, H. ....................................................................................... 160 Wuest, B. .................................................................................. 141 Wünsch, B. ..................................................................... 144, 162 Wurzel, J................................................................................... 189 Wüst, B. .............................................................................45, 138 X Xie, Y. ........................................................................................ 178 Z Zahler, S. .................................................................................. 104 Zander, J. .................................................................................. 186 Zavolan, M. .............................................................................. 190 Zeisig, R. ................................................................................... 102 Zeitlinger, M. ........................................................................... 141 Zeuzem, S. ............................................................................... 152 DPhG Annual Meeting 2015 Conference Book • 203 AUTHOR INDEX Zhang, S. .................................................................................. 104 Zhao, Q........................................................................................ 70 Zhu, Q.......................................................................................... 42 Ziegler, N. ................................................................................... 27 Ziller, A. .................................................................................... 132 Zivkovic, A. .............................................................................. 164 204 • DPhG Annual Meeting 2015 Conference Book Zlotos, D. P............................................................................... 176 Zoller, M. .................................................................................. 186 Zubair, M.................................................................................. 157 Zucht, H. ..................................................................................... 79 Zulkiewicz Gomes, D. ............................................................ 141 Zummersch, M........................................................................ 194 Geschäftsführer und Leiter der Geschäftsstelle Apotheker Dr. Michael Stein DPhG Geschäftsstelle Varrentrappstr. 40 – 42 60486 Frankfurt Tel.: 069-7191596-0 Fax: 069-7191596-29 Email: info@dphg.de http://www.dphg.de Univ.-Prof. Dr. Holger Stark Heinrich-Heine-Universität Düsseldorf Institut für Pharmazeutische and Medizinische Chemie Universitätsstr. 1 40225 Düsseldorf, GERMANY Phone: +49-(0)211–81 1 0478 Fax: +49-(0)211–81 1 3359 E-mail: stark@hhu.de 15.09.2015 DPhG Annual Meeting 2015 Conference Book • 205 NOTES 206 • DPhG Annual Meeting 2015 Conference Book NOTES DPhG Annual Meeting 2015 Conference Book • 207 NOTES 208 • DPhG Annual Meeting 2015 Conference Book SHORT PROGRAM Annual Meeting of the German Pharmaceutical Society 2015- DPhG Tuesday, September 22nd 14:00 – 17:45 Bürgersymposium: Geschichte der Pharmazie in Düsseldorf (Haus der Universität, Düsseldorf Schadowplatz 14) 19:00 – 21:00 Treffen/Vortrag Arbeitsgemeinschaft Katastrophenpharmazie (Haus der Universität, D, Schadowplatz 14) Wednesday, September 23rd 9:00 – 11:00 Sitzung VdPPHI e.V. Hochschullehrersitzung, B. Clement 11:30 – 13:00 DPhG Fachgruppen Meetings 13:00 – 13:30 (HS 3A) Opening of the Annual DPhG Meeting 2015 (HS 3A) I3 – Interactions, Integrations and Innovations 13:30 – 14:15 P.1 – C. E. Müller: I3 in Medicinal Chemistry of purinergic signaling (HS 3A) 14:15 – 15:00 P.2 – J. Breitkreutz: New concepts and products for individual drug dosing (HS 3A) 15:00 – 15:30 Coffee break - Poster viewing (even numbers) 15:30 – 17:00 SL1 - Antiviral drugs 17:00 – 18:00 Short poster lectures 17:00 – 18:00 Freunde der DPhG-Stiftung 18:00 – 22:00 Poster viewing (even numbers) and Welcome Reception SL2 - Blood/brain barrier SL3 - Regulation of beta-cell function – Implications for (HS 3E) (HS 3D) diabetes (HS 3A) (HS 3A) (building 23.02, room U1.23) Thursday, September 24th 8:30 – 9:15 P.3 – M. Biel: Ebola and more: Endolysosomal cation channels as novel drug targets (HS 3A) 9:15 – 10:00 P.4 – R. Thurmond: The pre-clinical and clinical development of H4R antagonists (HS 3A) 10:00 – 10:30 Coffee break - Poster viewing (odd numbers) 10:30 – 12:00 SL4 - GLISTEN - GPCR Medicinal Chemistry SL5 - Evidence based medication management SL6 - (Bio)Analytics (HS 3D) (HS 3A) (HS 3E) 12:00 – 13:30 Break for lunch time - Poster viewing (odd numbers) 13:30 – 15:00 SL7 - Allosteric regulation SL8 - Anti-infective compounds SL9 - Poorly solubles (HS 3A) (HS 3D) 15:00 – 15:30 Coffee break - Poster viewing (odd numbers) 15:30 – 17:00 SL10 - PPP in drug development SL11 - Screening techniques in SL12 - Signaling in cell death pharmacology & drug (HS 3A) development (HS 3D) (HS 3E) 17:00 – 18:30 SL13 - Anticancer and epigenetic drugs 17:00 – 18:30 Workshop, T. Hotopp, Fördermaßnahmen der DFG – Tipps und Hinweise 19:30 Conference dinner (HS 3E) SL14 - Personalized Medicine – SL15 - Focused pharmaceutical Biomarker and diagnostics research (HS 3A) (HS 3D) (HS 3E) (HS 3B) Friday, September 25th 8:30 – 9:15 P.5 – D. Häussinger: Liver research in Düsseldorf: Bridging basic and clinical science (HS 3A) 9:15 – 10:00 P.6 – K. Broich: Drug approval and regulatory science – Where to go? (HS 3A) 10:30 – 12:00 SL16 - Future molecular design SL17 - Medicaton safety in special patient groups SL18 - Hot topics in Pharmaceutical Biology – Young investigators in the spotlight (HS 3D) (HS 3E) (HS 3A) 12:00 – 13:00 Short lunch break 13:00 – 13:45 P.7 – P. Herdewijn: From synthetic nucleic acids to artificial genes and genomes: … (HS 3A) 14:00 – 15:00 Closing ceremony (HS 3A) 15:15 – 16:30 DPhG Jahreshauptversammlung (HS 3A) Saturday, September 26 th 15:00 – 18:30 Tag der Offizinpharmazie – Schmerzschulung (1. Teil) (HS 3D)