11.11.2012 Views

DPhG Jahrestagung 2012 - Ernst-Moritz-Arndt-Universität Greifswald

DPhG Jahrestagung 2012 - Ernst-Moritz-Arndt-Universität Greifswald

DPhG Jahrestagung 2012 - Ernst-Moritz-Arndt-Universität Greifswald

SHOW MORE
SHOW LESS

Create successful ePaper yourself

Turn your PDF publications into a flip-book with our unique Google optimized e-Paper software.

<strong>DPhG</strong>-<strong>Jahrestagung</strong> <strong>2012</strong><br />

Moleküle, Targets und Tabletten -<br />

Translationale Forschung für Arzneimittel der Zukunft<br />

11. – 13. Oktober in <strong>Greifswald</strong><br />

Tagungsband<br />

www.dphg.de<br />

ISBN: 978-3-00-039318-1<br />

Verlag: <strong>Universität</strong>sbibliothek <strong>Greifswald</strong>


Sponsoren der <strong>DPhG</strong>-<strong>Jahrestagung</strong> <strong>2012</strong>


Förderer der <strong>DPhG</strong>-<strong>Jahrestagung</strong> <strong>2012</strong>


<strong>DPhG</strong><br />

Deutsche Pharmazeutische Gesellschaft e. V.<br />

<strong>Jahrestagung</strong> <strong>2012</strong><br />

Programm<br />

&<br />

Tagungsband<br />

Institut für Pharmazie<br />

der<br />

Mathematisch-Naturwissenschaftlichen Fakultät<br />

<strong>Ernst</strong>-<strong>Moritz</strong>-<strong>Arndt</strong>-<strong>Universität</strong><br />

<strong>Greifswald</strong><br />

11.–13. Oktober


4<br />

Moleküle, Targets und Tabletten –<br />

Translationale Forschung für Arzneimittel der Zukunft<br />

Wissenschaftliches Komitee Organisationskomitee<br />

Prof. Dr. Patrick J. Bednarski Prof. Dr. Werner Weitschies<br />

Prof. Dr. Thomas Jira Prof. Dr. Christoph Ritter<br />

Prof. Dr. Heyo K. Kroemer Prof. Dr. Andreas Link<br />

Prof. Dr. Ulrike Lindequist PD Dr. Gregor Radau<br />

Prof. Dr. Sandra Klein Dr. Winfried Stahlkopf<br />

Prof. Dr. Andreas Link Dr. Thomas Emmrich<br />

Prof. Dr. Bernhard H. Rauch Dipl.-Pharm. Lisa Wilde<br />

Prof. Dr. Christoph Ritter Dipl.-Pharm. Felix Wilde<br />

Prof. Dr. Thomas Schweder<br />

Prof. Dr. Werner Siegmund<br />

Prof. Dr. Werner Weitschies<br />

Dipl.-Pharm. Heidi Lemmerhirt


Inhaltsverzeichnis<br />

Allgemeine Informationen 6<br />

Programm 7<br />

Vorprogramm – Mittwoch, 10. Oktober <strong>2012</strong> 8<br />

Pharmaziehistorische Veranstaltung Pharmazie in <strong>Greifswald</strong> 8<br />

Hauptsymposium Arzneimittelkontrolle 8<br />

Hauptprogramm – Donnerstag, 11. Oktober <strong>2012</strong> 9<br />

Diskussionsveranstaltungen Pharmazie 2020 – Perspektiven in Forschung und Lehre 9<br />

Eröffnung der <strong>Jahrestagung</strong> <strong>2012</strong> 9<br />

Hauptprogramm – Freitag, 12. Oktober <strong>2012</strong> 11<br />

Hauptprogramm – Sonnabend, 13. Oktober <strong>2012</strong> 15<br />

Vorträge 17<br />

Plenarvortrag Moleküle 18<br />

Plenarvortrag Targets 19<br />

Plenarvortrag Tabletten 20<br />

Sessionvorträge Moleküle 21<br />

Session Struktur-basiertes Design 21<br />

Session Molekulare Werkzeuge 25<br />

Session IVIVC: Modelle und Modellierungen 29<br />

Session Moderne Trenntechniken 33<br />

Session Naturstoffe 36<br />

Session Klinische Wirkstoffentwicklung 40<br />

Sessionvorträge Targets 44<br />

Session Proteomics und Metabolomics 44<br />

Session Transporter und Ionenkanäle 48<br />

Session Nukleäre Rezeptoren und Transkriptionsfaktoren als Wirkstofftargets 52<br />

Session Thrombozytenfunktion und Gerinnung 58<br />

Session Neue Targets und Testsysteme 61<br />

Session Tumortherapie und Resistenzmechanismen 65<br />

Sessionvorträge Tabletten 69<br />

Session Nanomedizin 69<br />

Session Pharmaverfahrenstechnik 75<br />

Session Bioanalytik: Testsysteme und Datenqualität 79<br />

Session Spezielle Darreichungsformen 83<br />

Session Optimierte Patientenversorgung 87<br />

Session Arzneimittelüberwachung/Regulatory Affairs 90<br />

Poster 93<br />

Autorenverzeichnis 141<br />

5


Allgemeine Informationen<br />

Lagepläne der Innenstadt <strong>Greifswald</strong>s sowie vom Campus finden sich am Ende des Tagungsbandes.<br />

Internetzugang<br />

Die <strong>Universität</strong> <strong>Greifswald</strong> nimmt am eduroam-Projekt (education roaming) teil. eduroam ermöglicht es Ihnen, sich<br />

mit Ihrer persönlichen Nutzerkennung Ihres Rechenzentrums in das Internet einzuloggen und einen gesicherten<br />

WLAN-Zugang an allen teilnehmenden Forschungs- und Ausbildungseinrichtungen, also auch hier während der<br />

Tagung in <strong>Greifswald</strong> nutzen zu können. Die allgemeinen, von Ihrer Hardware (Laptop, PC, Mobiltelefone)<br />

unabhängigen Einstellungen zur Nutzung dieses Dienstes sind im Folgenden aufgeführt:<br />

SSID eduroam<br />

Sicherheitstyp Firmenweiter WPA2 (WPA2 Enterprise)<br />

äußeres Identifizierungsverfahren TTLS<br />

inneres Identifizierungsverfahren PAP<br />

externe Identität anonymous@uni-greifswald.de<br />

innere Identität Ihre persönliche Nutzerkennung am URZ<br />

Vertrauenswürdige Zertifikate Deutsche telekom Root CA 2<br />

Weitere Informationen unter: http://www.rz.uni-greifswald.de/dienste/zugang-zum-uni-netz/eduroam.html<br />

Alternativ dazu kann man sich über den Tagungsaccount in das Netzwerk der Uni <strong>Greifswald</strong> einloggen:<br />

Benutzer<br />

Passwort<br />

<strong>DPhG</strong>_<strong>2012</strong><br />

Die <strong>Universität</strong> übernimmt für Schäden, die aus der Anwendung des Funknetzes (Wireless-LAN) entstehen, keinerlei<br />

Haftung.<br />

Auf Seite 151 findet sich ein Linienplan des öffentlichen Nahverkehrs <strong>Greifswald</strong> (Bus).<br />

6


Programm


Vorprogramm – Mittwoch, 10. Oktober <strong>2012</strong><br />

► Die Veranstaltungen des Vorprogramms finden im Hörsaal des C_DAT statt.<br />

Pharmaziehistorische Veranstaltung<br />

Pharmazie in <strong>Greifswald</strong><br />

14:00–14:15 Begrüßung und Einführung<br />

Prof. Dr. Christoph Friedrich (Marburg)<br />

14:15–15:00<br />

15:00–15:45<br />

15:30–15:45 Kaffeepause<br />

15:45–16:30<br />

16:30–17:15<br />

Zur Entwicklung des Pharmaziestudiums an der <strong>Universität</strong> <strong>Greifswald</strong> vor 1945<br />

Prof. Dr. Christoph Friedrich (Marburg)<br />

Zur Entwicklung der Pharmazie an der <strong>Universität</strong> <strong>Greifswald</strong> nach 1945<br />

Prof. Dr. Thorsten Beyrich (<strong>Greifswald</strong>)<br />

Das Apothekenwesen der Stadt <strong>Greifswald</strong> von den Anfängen bis zum 20.<br />

Jahrhundert<br />

Dr. Hartmut Bettin (<strong>Greifswald</strong>)<br />

Die Homöopathie an der <strong>Universität</strong> <strong>Greifswald</strong><br />

Dr. Ulrich Meyer (Berlin)<br />

Vorprogramm<br />

Abend für Offizinapotheker<br />

18:00–18:45 Chair: Prof. Dr. Andreas Link<br />

Michael Jackson – DieSehnSUCHT nach Schlaf<br />

Prof. Dr. Dieter Steinhilber/Prof. Dr. Theo Dingermann (Frankfurt am Main)<br />

19:00–20:00<br />

Individualisierte Medizin<br />

Podiumsgespräch Dr. Hildegard Kaulen (FAZ), Prof. Dr. Theo Dingermann (Frankfurt am<br />

Main), Prof. Dr. Wolfgang Lieb (<strong>Greifswald</strong>), Dr. Christian Ude (Darmstadt), u.a.<br />

Ganztägiges<br />

Hauptsymposium Arzneimittelkontrolle<br />

Eigenverantwortlich organisiert und selbständig durchgeführt von der Fachgruppe Arzneimittelkontrolle –<br />

Pharmazeutische Analytik.<br />

► Ort: Biotechnikum, Raum 0.09<br />

Das Programm publiziert die Fachgruppe unter:<br />

http://www.pharmchem.tu-bs.de/forschung/waetzig/#veranstaltungen<br />

ab 17:00 Uhr<br />

Hörsaal<br />

Pharmazie<br />

Sitzung des Verbandes der Professoren an Pharmazeutischen Hochschulen der<br />

Bundesrepublik Deutschland e. V. (VdPPHI)<br />

8 Programm – 10.10.


Hauptprogramm – Donnerstag, 11. Oktober <strong>2012</strong><br />

Diskussionsveranstaltungen<br />

Pharmazie 2020 – Perspektiven in Forschung und Lehre<br />

08:30–10:00 5 Parallelveranstaltungen der einzelnen Prüfungsfächer<br />

Audimax HS 1 Audimax HS 2 Audimax HS 3 Audimax HS 4 Audimax HS 5<br />

09:45<br />

Uni Hauptgebäude<br />

Pressekonferenz<br />

im Beratungsraum des Rektorats, Domstr. 11, Eingang 2<br />

10:00–10:30 Pause (Fußweg in die Stadthalle)<br />

10:30–12:00<br />

Stadthalle, Kaisersaal<br />

13:00–13:30<br />

Stadthalle, Kaisersaal<br />

13:30–14:15<br />

Stadthalle, Kaisersaal<br />

14:15–15:00<br />

Stadthalle, Kaisersaal<br />

Abschlussdiskussion Plenarveranstaltung<br />

Chair: Prof. Dr. Angelika M. Vollmar /Prof. Dr. Dieter Steinhilber<br />

Eröffnung der <strong>Jahrestagung</strong> <strong>2012</strong><br />

Moleküle, Targets und Tabletten –<br />

Translationale Forschung für Arzneimittel der Zukunft<br />

Dipl.-Ing. Jörg Hochheim, Leiter des Dezernats für Bauwesen und Umwelt und<br />

1. Stellvertreter des Oberbürgermeisters der Hansestadt <strong>Greifswald</strong><br />

Prof. Dr. Rainer Westermann, Rektor der <strong>Ernst</strong>-<strong>Moritz</strong>-<strong>Arndt</strong>-<strong>Universität</strong> <strong>Greifswald</strong><br />

Plenarvortrag Moleküle<br />

Prof. Dr. Rolf Müller (Saarbrücken)<br />

Genom-basierte mikrobielle Naturstoff-Forschung für die<br />

Entwicklung von anti-Infektiva<br />

Plenarvortrag Targets<br />

Prof. Dr. Kathrin Mädler (Bremen)<br />

Anti-entzündliche Therapien für Diabetes – Rettung für die<br />

Beta-Zelle<br />

15:00–15:30 Fußweg zum neuen Campus<br />

15:30–16:00 Kaffeepause<br />

Programm – 11.10. 9


Moleküle ► Hörsaal Biochemie<br />

16:00–17:30 Struktur-basiertes Design<br />

Chair: Prof. Dr. Wolfgang Sippl & Prof. Dr. Christoph Sotriffer<br />

Prof. Dr. Gerhard Wolber (Berlin): „New algorithms for 3D pharmacophore-based virtual screening“<br />

Prof. Dr. Holger Gohlke (Düsseldorf): „From Determinants of RUNX1/ETO Tetramerization to Small-Molecule<br />

Protein-Protein Interaction Inhibitors Targeting Acute Myeloid Leukemia“<br />

Dr. Peter Kolb (Marburg): „Docking to GPCRs: ligand diversity, efficacy and selectivity“<br />

Dr. Andrea Straßer (Regensburg): „Molecular dynamic simulation of a symmetric active state hβ2R-Gαβγhomodimer<br />

complex - structural and energetical analysis“<br />

Targets ► Hörsaal Biotechnikum<br />

16:00–17:30 Proteomics und Metabolomics<br />

Chair: Prof. Dr. Irmgard Merfort & Prof. Dr. Thomas Schweder<br />

Prof. Dr. Uwe Völker (<strong>Greifswald</strong>): „OMICs-Analysen von Populations-basierten Kohorten – Mögliche Ansätze<br />

für das Biomarkerscreening“<br />

Prof. Dr. Thomas Efferth (Mainz): „Drug target fishing in silico and in vitro“<br />

Dr. Manuel Liebeke (London): „From whole-organism to sub-cellular localization of metabolites by mass<br />

spectrometry imaging“<br />

Rico Schwarz (Halle): „Incorporation of unnatural amino acids to monitor conformational changes in peroxisome<br />

proliferator-activated receptor alpha (PPARα)“<br />

Tabletten ► Hörsaal Physik<br />

16:00–17:30 Nanomedizin<br />

Chair: Prof. Dr. Udo Bakowsky & Prof. Dr. Claus-Michael Lehr<br />

Prof. Dr. Claus-Michael Lehr (Saarbrücken): „Nanomedicines for targeted drug delivery to the inflamed intestinal<br />

mucosa“<br />

Prof. Dr. Udo Bakowsky (Marburg): „Selective gene vehicles for enhanced DNA and siRNA delivery“<br />

Dr. Christian Wischke (Teltow): „Modulating immune responses of human cells by adjuvant loaded particulate<br />

carriers“<br />

Jun.-Prof. Dr. Peter R. Wich (Mainz): „Acid-Degradable Dextran Particles for the Delivery of Biotherapeutics“<br />

Astrid Müller (Jena): „Characterization of Bacterial Nanocellulose as Drug Delivery System for Low and High<br />

Molecular Weight Drugs“<br />

Dr. Mathias Könczöl (Freiburg): „Biomedical applications of magnetite in the respiratory system - Toxicological<br />

considerations and mechanistic studies in A549 lung cells“<br />

17:30–19:00 Posterpräsentationen (Institut für Biochemie)<br />

19:00–22:00 Begrüßungsabend, Posterpräsentationen (Institut für Biochemie)<br />

10 Programm – 11.10.


Hauptprogramm – Freitag, 12. Oktober <strong>2012</strong><br />

Moleküle ► Hörsaal Biochemie<br />

09:00–10:30 Molekulare Werkzeuge<br />

Chair: Prof. Dr. Ulrike Holzgrabe & Prof. Dr. Andrea Sinz<br />

Dr. Marcus Bantscheff (Cellzome AG): „Chemoproteomic target deconvolution of bioactive molecules“<br />

Christopher Lang (Erlangen): „Molecular tools for positron emission tomography (PET): Development of a highly<br />

selective radiotracer for the detection of neurotensin receptor 1 positive tumors“<br />

Prof. Dr. Wolfgang Maison (Hamburg): „Analogs of siderophores and mussel adhesion proteins for applications<br />

in clinical hygiene and implant medicine“<br />

Dr. Eberhard Heller (Würzburg): „Microwave-Enhanced Synthesis of Fluorescent Dyes for Pharmacological<br />

Purposes Using Real-Time Observation by UV/Vis Spectroscopy“<br />

Prof. Dr. Armin Buschauer (Regensburg): „Fluorescent and radiolabelled ligands as molecular tools for NPY<br />

receptors“<br />

10:30–11:00 Kaffeepause<br />

11:00–12:30 IVIVC: Modelle und Modellierungen<br />

Chair: Prof. Dr. Charlotte Kloft & Prof. Dr. Werner Weitschies<br />

Dr. Thorsten Lehr (Saarbrücken): „From Mice to Men: Application and Impact of in vivo Modeling and Simulation<br />

in Drug Research and Development“<br />

Dr. Grzegorz Garbacz (<strong>Greifswald</strong>): „Estimating the In Vivo Release Behaviour of Extended- and Immediate-<br />

Release Products using biorelevant bissolution stress tests“<br />

Dr. Steffi Hansen (Saarbrücken): „Improved input parameters for diffusion models of skin absorption“<br />

Arun Jain (<strong>Greifswald</strong>): „Interindividual gastrointestinal erosion and inter- as well as intraindividual<br />

gastrointestinal transit time variability of hydrophilic matrix tablets“<br />

12:30–13:30 Mittagspause (C_DAT Seminarraum)<br />

13:30–15:00 Moderne Trenntechniken<br />

Chair: Prof. Dr. Thomas Jira & Prof. Dr. Michael Lämmerhofer<br />

Prof. Dr. Hermann Wätzig (Braunschweig): „Continuous Improvement in Pharmazeutischer Analytik: schneller,<br />

präziser, selektiver“<br />

Prof. Dr. Andrea Sinz (Halle): „Untersuchung von Proteinen und Metaboliten -- Was können moderne<br />

Massenspektrometer leisten?<br />

Evamaria Falck (Münster): „Metabolism studies of Ifenprodil, a potent NR2B-selective NMDA receptor<br />

antagonist“<br />

Dr. Anette Thiessen (Anton Paar GmbH): „Macroscopic Questions and Molecular Answers: Measuring Optical<br />

Rotation in Latest Chiral Separation Techniques“<br />

15:00–15:30 Kaffeepause<br />

15:30–17:00 Naturstoffe<br />

Chair: Prof. Dr. Ulrike Lindequist & Prof. Dr. Angelika M. Vollmar<br />

Dr. Birgit Kraus (Regensburg): „Hepatic bioactivity of phenolic natural compounds“<br />

Dr. Karin von Schwarzenberg (München): „Mode of cell death induction by pharmacological V-ATPase<br />

inhibition“<br />

Dr. Timo Niedermeyer (Berlin/<strong>Greifswald</strong>): „Cyanobacteria in Anticancer Natural Products Research“<br />

Prof. Dr. Florian C. Stintzing (WALA Heilmittel GmbH): „Applied phytochemical research – an industry’s<br />

perspective“<br />

Programm – 12.10. 11


17:15–18:45 Klinische Wirkstoffentwicklung<br />

Chair: Prof. Dr. Klaus Mohr & Prof. Dr. Werner Siegmund<br />

Prof. Dr. Jörg König (Erlangen): „Single- and multiple-transfected cell lines as tools for the analysis of drug<br />

transport and of the interplay of drug transport and drug metabolism“<br />

Dr. Stefan Oswald (<strong>Greifswald</strong>): „Methods to investigate the clinical relevance of drug transporters“<br />

Dr. Dieter Runge (PRIMACYT GmbH): „The functionality of uptake transporters in primary hepatocytes of<br />

different species can be verified by estrone-3-sulfate”<br />

Anett Engel (<strong>Greifswald</strong>): „Solubilizing agents influence the pharmacokinetics of probe drugs by transporter<br />

interaction“<br />

20:00–23:00 Gesellschaftsabend (Stadthalle, Theatercafé)<br />

Targets ► Hörsaal Biotechnikum<br />

09:00–10:30 Transporter und Ionenkanäle<br />

Chair: Prof. Dr. Bernhard H. Rauch & Prof. Dr. Peter Ruth<br />

Prof. Dr. Marc Freichel (Heidelberg): „TRP channels as regulators of blood pressure and fertility“<br />

Dr. Robert Lukowski (Tübingen): „Role of Ca2+ -activated K + Channels in Breast Cancer Proliferation“<br />

PD Dr. Gabriele Jedlitschky (<strong>Greifswald</strong>): „Transporters in human platelets: physiological function and impact for<br />

pharmacotherapy“<br />

PD Dr. Stephan Reichl (Braunschweig): „Expression analysis of drug transporter proteins in RPMI 2650 cell line<br />

and excised human nasal mucosa“<br />

10:30–11:00 Kaffeepause<br />

11:00–12:30 Nukleäre Rezeptoren und Transkriptionsfaktoren als<br />

Wirkstofftargets<br />

Chair: Prof. Dr. Manfred Schubert-Zsilavecz & Prof. Dr. Dieter Steinhilber<br />

Prof. Dr. Ronald Gust (Innsbruck): „Investigation on the estrogen receptor alpha selectivity of 2,3,5-triaryl-1Hpyrroles“<br />

Prof. Dr. Manfred Jung (Freiburg): „Chemical epigenetics: histone modifying enzymes as targets to regulate<br />

transcription“<br />

Prof. Dr. Irmgard Merfort (Freiburg): „Helenalin derivatives and their impact on transcriptional control of<br />

mediators involved in rheumatic diseases“<br />

Prof. Dr. Frank Böckler (Tübingen): „Rescuing mutant p53 using halogen-enriched fragment libraries (HEFLibs)“<br />

Jun. Prof. Dr. Eugen Proschak (Frankfurt): „Novel leads for PPARs and FXR: a computer-aided drug design<br />

approach“<br />

Prof. Dr. Dieter Steinhilber (Frankfurt): „Inhibitors of class I histone deacetylases induce 5-lipoxygenase<br />

expression“<br />

12:30–13:30 Mittagspause (C_DAT Seminarraum)<br />

13:30–15:00 Thrombozytenfunktion und Gerinnung<br />

Chair: Prof. Dr. Susanne Alban & Prof. Dr. Andreas Greinacher<br />

Prof. Dr. Susanne Alban (Kiel): „Novel oral anticoagulants – State of the art and aspects of their application“<br />

Prof. Dr. Andreas Greinacher (<strong>Greifswald</strong>): „Novel platelet inhibitors – State of the art and aspects of their<br />

application”<br />

Dr. Krystin Krauel (<strong>Greifswald</strong>): „Naturally occurring PF4/polyanion complexes prime for heparin-induced<br />

thrombocytopenia as a misdirected host defense mechanism“<br />

12 Programm – 12.10.


15:00–15:30 Kaffeepause<br />

15:30–17:00 Neue Targets und Testsysteme<br />

Chair: Prof. Dr. Manfred Jung & Prof. Dr. Stefan Laufer<br />

Prof. Dr. Conrad Kunick (Braunschweig): „PfGSK-3: A target for new antimalarial drugs?“<br />

Prof. Dr. Daniel Rauh (Dortmund): „Chemical Oncology – Converging Cancer Genetics, Structural Biology and<br />

Medicinal Chemistry“<br />

Dr. Dante Rotili (Rom): „Development of lysine mimic quinazoline-based selective inhibitors of a Jmj-C histone<br />

demethylase family“<br />

Prof. Dr. Mike Schutkowski (Halle): „Profiling of epigenetic targets using peptide microarrays“<br />

17:15–18:45 Tumortherapie und Resistenzmechanismen<br />

Chair: Prof. Dr. Patrick J. Bednarski & Prof. Dr. Conrad Kunick<br />

Prof. Dr. Rolf W. Hartmann (Saarbrücken): „Anti-infectives research to combat drug resistant tumors?“<br />

Prof. Dr. Matthias Kassack (Düsseldorf): „Chemoresistance of ovarian cancers against platinum complexes“<br />

Prof. Dr. Ingo Ott (Braunschweig): „Medicinal Chemistry with Organometallics - From Catalysts to Anticancer<br />

Drugs?“<br />

Dr. Matthias Tacke (Dublin): „NHC-Silver(I) and NHC-gold(I) complexes as bioorganometallic anticancer and<br />

antibacterial drugs“<br />

Riad Schulz (<strong>Greifswald</strong>): „A new class of glutathione peroxidase inhibitors is able to reverse resistance to<br />

chemotherapeutics in human B-cell lymphoma cell lines“<br />

20:00–23:00 Gesellschaftsabend (Stadthalle, Theatercafé)<br />

Tabletten ► Hörsaal Physik<br />

09:00–10:30 Pharmaverfahrenstechnik<br />

Chair: Prof. Dr. Peter Kleinebudde & Prof. Dr. Christel Müller-Goymann<br />

Prof. Dr. Heike Bunjes (Braunschweig): „Formulierungs-Screening mit kolloidalen Wirkstoffträgersystemen“<br />

Dr. Markus Thommes (Düsseldorf): „Pharmaceutical Extrusion technology – Recent developments“<br />

PD Dr. Hubert Rein (Bonn): „Hot melt extruded dosage forms“<br />

André Bitterlich (Braunschweig): „Nanogrinding of naproxen: influence of process parameters on product<br />

quality“<br />

10:30–11:00 Kaffeepause<br />

11:00–12:30 Bioanalytik: Testsysteme und Datenqualität<br />

Chair: Prof. Dr. Hermann Wätzig & Prof. Dr. Gerhard Winter<br />

Prof. Dr. Knut Baumann (Braunschweig): „Bildverarbeitung für biomedizinisch eingesetzte Mikroskopie“<br />

Dr. Achmed Besheer (München): „The use of microscale thermophoresis (MST) for protein characterization and<br />

formulation“<br />

Tim Menzen (München): „High-Throughput Protein Melting Temperature Analysis by Differential Scanning<br />

Fluorimetry“<br />

Prof. Dr. Christiane Ritter (Braunschweig): „NMR Spektroskopie von hochmolekularen Proteinkomplexen“<br />

12:30–13:30 Mittagspause (C_DAT Seminarraum)<br />

Programm – 12.10. 13


13:30–15:00 Spezielle Darreichungsformen<br />

Chair: Prof. Dr. Jörg Breitkreutz & Prof. Dr. Sandra Klein<br />

Prof. Dr. Jörg Breitkreutz (Düsseldorf): „Orodispersible dosage forms – novel concepts, innovative products,<br />

new therapeutic options“<br />

Marco Neumann (<strong>Greifswald</strong>): „Challenges in developing gastroretentive dosage forms“<br />

Matthias Rischer (Losan Pharma GmbH): „Innovative nanoparticular dosage forms – advantages in<br />

pharmacokinetics and administration“<br />

Susanne Kirchhof (Regensburg): „Development and pharmaceutical technological applications of Diels-Alder<br />

hydrogels”<br />

15:00–15:30 Kaffeepause<br />

15:30–17:00 Optimierte Patientenversorgung<br />

Chair: Prof. Dr. Ulrich Jaehde & Prof. Dr. Christoph Ritter<br />

Prof. Dr. Ulrich Jaehde (Bonn): „Medication safety research as challenge for pharmaceutical scientists“<br />

Prof. Dr. Thilo Bertsche (Leipzig): „Klinische Pharmazeuten zur Verbesserung der Patientensicherheit in der<br />

Arzneimitteltherapie“<br />

Prof. Dr. Petra A. Thürmann (Witten/Herdecke): „Arzneimitteltherapiesicherheit in Alten- und Pflegeheimen“<br />

Dr. Thomas Fiß (<strong>Greifswald</strong>): „Identifikation von Risikofaktoren für das Auftreten von Arzneimittelbezogenen<br />

Problemen im Rahmen von häuslichen Medikationsreviews“<br />

17:15–18:45 Arzneimittelüberwachung/Regulatory Affairs<br />

Chair: Dr. Olaf Queckenberg & PD Dr. Klaus Raith<br />

Dr. Andreas Sutter (Bayer AG): „Analyzing the use of (Q)SAR models for the evaluation of potential genotoxic<br />

impurities: A cookbook in support of ICH M7“<br />

Dr. Adrian Funke (Bayer AG): „Control strategy for an active coating process based on systematic process<br />

development (DoE) and in-line Raman spectroscopy (PAT)“<br />

Dr. Isabel Astner (Braunschweig): „Gewebe und Zellen – Arzneimittel für neuartige Therapien“<br />

Dr. Nicholas Schramek (Oberschleißheim): „Analytik von illegalen Arzneimitteln“<br />

20:00–23:00 Gesellschaftsabend (Stadthalle, Theatercafé)<br />

14 Programm – 12.10.


09:00–09:45<br />

HS Nord Med.<br />

10:00–11:30<br />

HS Nord Med.<br />

11:30–11:45<br />

HS Nord Med.<br />

Hauptprogramm – Sonnabend, 13. Oktober <strong>2012</strong><br />

11:45–12:15 Kaffeepause<br />

12:15–13:00<br />

HS Nord Med.<br />

Plenarvortrag Tabletten<br />

Prof. Dr. Wolfgang Frieß (München)<br />

Analyse von Aggregaten und Partikeln in Proteinarzneimitteln<br />

– was ist eigentlich das Problem?<br />

Kurzvorstellung von Leuchtturmprojekten<br />

• Helmholtz Institute for Pharmaceutical Research (HIPS)<br />

Saarland<br />

• Center of Drug Absorption and Transport (C_DAT)<br />

<strong>Greifswald</strong><br />

• Zentrum für Pharmaverfahrenstechnik (PVZ)<br />

Braunschweig<br />

• Fraunhofer Projektgruppe – Translationale Medizin und Pharmakologie (TMP) Frankfurt<br />

am Main<br />

Pharmazie 2020 – Perspektiven in Forschung und Lehre<br />

Chair: Prof. Dr. Dieter Steinhilber<br />

Abschlussveranstaltung<br />

Verleihung von Ehrungen und Preisen<br />

Programm – 13.10. 15


Vorträge


Plenarvortrag Moleküle<br />

Genom-basierte mikrobielle Naturstoff-Forschung für die Entwicklung von anti-Infektiva<br />

Müller R<br />

Helmholtz-Institute for Pharmaceutical Research Saarland, Helmholtz Centre for Infection Research and Pharmaceutical Biotechnology,<br />

Saarland University, Campus, Building C2 3, 66123 Saarbrücken, Germany<br />

Microorganisms continue to be one of the most important resources of bioactive molecules exhibiting potential for<br />

pharmaceutical application (1). This is especially true for urgently needed antibacterials which almost exclusively are<br />

derived from bacteria or fungi. Aiming at the identification of novel hit and lead structures ideally addressing new<br />

targets in pathogens, genome analysis of microbial producer organisms has become very useful for the in silico<br />

prediction of the hypothetical potential of a given organism for the production of secondary metabolites (2,3).<br />

Although revealing tremendous potential in terms of the presence of typical natural product biosynthetic genes, the<br />

translation of this knowledge into "real compounds in the flask" clearly represents the bottleneck in this approach. In<br />

the presentation I will discuss our recent efforts to develop and employ genome mining to a) identify (4,5), b)<br />

heterologously express (6,7) and c) modify secondary metabolite biosynthesis (8,9). In addition, I will present two<br />

success stories where novel natural product scaffolds were isolated and have been found to indeed address new<br />

targets in pseudomonads and staphylococci (10-12).<br />

Reference List:<br />

1. Newman, D. J., Cragg, G. M.: J.Nat.Prod. <strong>2012</strong>, 75(3): 311–335.<br />

2. Bode, H. B., Müller, R.: Angew.Chem.Int.Ed. 2005, 44(42): 6828–6846.<br />

3. Wenzel, S. C., Müller, R.: Natural Product Reports 2011,26(11):1385-407.<br />

4. Schneiker, S. et al.: Nat.Biotechnol. 2007, 25(11): 1281–1289.<br />

5. Cortina, N. S. et al.: Angew.Chem.Int.Ed. <strong>2012</strong>, 51(3): 811–816.<br />

6. Wenzel, S. C. et al.: Chem.Biol. 2005, 12(3): 349–356.<br />

7. Fu, J. et al.: Nat.Biotechnol. <strong>2012</strong>, 30 440–446.<br />

8. Rachid, S. et al.: ChemBioChem 2011, 12(6): 922–931.<br />

9. Quade, N. et al.: Nat Chem Biol Nat Chem Biol <strong>2012</strong>, 8(1): 117–124.<br />

10. Bielecki, P. et al.: ChemBioChem <strong>2012</strong>, in press.<br />

11. Steinmetz, H. et al.: Angew.Chem.Int.Ed. 2011, 50(2): 532–536.<br />

12. Dehn, R. et al.: Angew.Chem.Int.Ed. 2011, 50(17): 3882–3887.<br />

18 Plenarvorträge


Plenarvortrag Targets<br />

Anti-inflammatory compounds as novel tools to rescue the pancreatic β-cell in diabetes<br />

Maedler K<br />

Islet Biology Laboratory, Centre for Biomolecular Interactions Bremen, University of Bremen, Germany<br />

In both, type 1 and type 2 diabetes mellitus the main processes leading to β-cell failure are apoptosis and loss of<br />

function. Although the β-cell has an enormous capacity to adapt to conditions of higher insulin demand, e.g. in<br />

obesity and pregnancy, its continuous overstimulation leads to a β-cell overwork, subsequent loss of function and<br />

apoptosis and diabetes progression. This is a result from a complex interplay of environmental factors together with<br />

genetic predisposition.<br />

Many studies demonstrate how cytokines and chemokines have an active role in triggering the immune-response<br />

against the β-cell population. In a recent study we have identified 2 classical inflammatory mediators, the cytokine IL-<br />

1β and the chemokine CXCL10, which both play an active role in triggering β-cell destruction.<br />

We have identified the Toll like receptor 4 as the receptor for CXCL10 and as new pathway for the induction of β-cell<br />

apoptosis.<br />

In the lecture, I will give an overview on mechanisms of destruction of the β-cell in diabetes, the influence of obesity,<br />

the crosstalk between fat and pancreas and resulting new therapeutic approaches to fight onset and progression of<br />

diabetes.<br />

Key words: diabetes, β-cells, apoptosis, proliferation, inflammation, IL-1β, CXCL10<br />

Plenarvorträge 19


Plenarvortrag Tabletten<br />

Analysis of aggregates and particles in protein drug products – what is the challenge?<br />

Frieß W<br />

Department of Pharmacy; Pharmaceutical Technology and Biopharmaceutics; Ludwig-Maximilians Universitaet Muenchen, Butenandtstr. 5,<br />

D-81377 Muenchen<br />

Protein aggregates and particles are currently a hot topic in pharmaceutical industry as well as for the regulatory<br />

agencies. FDA sees a strong scientific basis for aggregates, including sub-visible particulates, to induce immune<br />

responses but insufficient data to determine the likelihood of immune response induction given the levels/types of<br />

aggregates in any given product. Therefore, aggregates and sub-visible particulates should be assumed critical to<br />

the observed safety and efficacy profile and controlled commensurate with risk [1]. In order to adequately define the<br />

actual risk, on the one hand more detailed studies on the causes and correlation of protein aggregates and particles<br />

with immunogenicity are deemed necessary. On the other hand the detailed characterization of protein particles is<br />

mandatory. The latter includes both quantity and quality such as size, shape, morphology, bond (covalent or<br />

noncovalent) or protein conformation. This implies that no single method can cover the entire range but instead a<br />

multitude of orthogonal methods is applied for characterization [2,3]. All methods come with limitations which need to<br />

be considered and new methods are developed. For example classical size exclusion chromatography (SEC) may<br />

provide inaccurate results in the analysis of protein aggregates due to protein adsorption to column media as well as<br />

dissociation or formation of aggregate species at the higher salt concentration typically applied in the eluent.<br />

Orthogonal methods like analytical ultracentrifugation or asymmetrical flow field flow fractionation should always be<br />

used in the development of SEC methods and to corroborate the accuracy of SEC results [4]. In order to obtain<br />

additional information, methods can be combined, for example a combination of SEC with addition of fluorescent dye<br />

enables to trace unfolding of the individual separate protein species [5]. Another option is to combine SEC with the<br />

determination of individual second virial coefficients of monomer and oligomers in protein samples [6]. For<br />

characterization of subvisible particles several new methods have been established. Nanoparticle tracking analysis<br />

(NTA) is an innovative system for sizing particles from about 30 to 1,000 nm based on laser light scattering<br />

microscopy to track individual nanoparticles moving under Brownian motion [7]. Another system called Archimedes is<br />

based on a fluidic microchannel embedded inside a resonator through which the sample is transported and as<br />

particles flow through the resonators the change in resonant frequency gives the individual particle mass and size<br />

with high resolution [8]. But also established methods like the classic Coulter Counter experience a renaissance as<br />

the results of the standard optic based methods to analyse subvisible particles like light obscuration of micro flow<br />

imaging depend on refractive index differences between particle and formulation [9]. Ultimately these developments<br />

also give rise to new guidelines to direct the industry and USP has just published a chapter “Subvisible Particulate<br />

Matter in Therapeutic Protein Injections” for comment in the Pharmacopia Forum. The critical evaluation of<br />

protein aggregates and particles is not only challenging for standard formulations but also presents a major<br />

roadblock for drug delivery systems [10].<br />

References:<br />

1. Rosenberg A.: Workshop on protein aggregation and immunogenicity, July <strong>2012</strong> Breckenridge, CO.<br />

2. Mahler H.-E. et al.: J. Pharm. Sci. 2009, 98(9): 2909–2934.<br />

3. Zölls S. et al.: J. Pharm. Sci. <strong>2012</strong>, 101(3): 914-935.<br />

4. Carpenter J. et al.: J. Pharm. Sci. 2010, 99(5), 2200–2208.<br />

5. Printz M., Friess W.: J. Pharm. Sci. <strong>2012</strong>, 101(2): 826-837.<br />

6. Printz M., Kalonia D., Friess W.: J. Pharm. Sci. <strong>2012</strong>, 101(1): 363-372.<br />

7. Filipe V., Hawe A., Jiskoot W.: Pharm. Res. 2010, 27(5): 796-810.<br />

8. Burg T.P. et al.: Nature 2007, 446(7139), 1066-1069.<br />

9. Hawe A.: Workshop on protein aggregation and immunogenicity, July <strong>2012</strong> Breckenridge, CO.<br />

10. Jiskoot W. et al.: J. Pharm. Sci. <strong>2012</strong>, 101(3): 946-954.<br />

20 Plenarvorträge


Sessionvorträge Moleküle<br />

Session Struktur-basiertes Design<br />

New algorithms for 3D pharmacophore-based virtual screening<br />

Spitzer G1 , Liedl K1 , Wolber G2 1Institute of General, Inorganic and Theoretical Chemistry, University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria<br />

2Institute of Pharmacy/Pharmaceutical Chemistry, Computer-Aided Drug Design, Freie <strong>Universität</strong> Berlin, Königin-Luisestr. 2+4, 14195 Berlin,<br />

Germany<br />

Virtual screening using three-dimensional arrangements of chemical features (3D pharmacophores) has become a<br />

highly relevant method for virtual hit identification and lead optimization. Although frequently used, considerable<br />

differences exist in the interpretation of chemical features and the implementation of their corresponding 3D overlay<br />

algorithms [1].<br />

We have recently continued the development of our 3D alignment algorithm relying on pattern recognition. Based on<br />

this method, a workflow for high-performance multi-conformational database screening has been created using new<br />

fingerprint filtering techniques that delivers a higher degree of geometric accuracy than previously published or<br />

commercially available approaches. Despite the considerable gain in geometric subsampling and the resulting<br />

increased alignment accuracy, this technique allows for computationally highly efficient virtual pharmacophore<br />

screening allowing to process millions of molecules in hours on a desktop computer. The impact of increased<br />

geometric accuracy on virtual screening will be presented and discussed by means of benchmarks and prospective<br />

screening studies.<br />

References:<br />

1. Spitzer G et al., J. Chem Inf. Model 2010, 50(7): 1241-1247.<br />

Sessionvorträge Moleküle 21


From Determinants of RUNX1/ETO Tetramerization to Small-Molecule Protein-Protein Interaction<br />

Inhibitors Targeting Acute Myeloid Leukemia<br />

Metz A1 ; Schanda J2 ; Wichmann C2 ; Grez M2 ; Gohlke H1 1 Institute of Pharmaceutical and Medicinal Chemistry, Heinrich-Heine-University, <strong>Universität</strong>sstr. 1, 40591 Düsseldorf, Germany<br />

2 Georg-Speyer-Haus, Institute for Biomedical Research, Paul-Ehrlich-Str. 42-44, 60596 Frankfurt, Germany<br />

A promising way to interfere with biological processes is through the control of protein-protein interactions by means<br />

of small molecules. Many of these small-molecule modulators known to date have not been found by rational design<br />

approaches. In large part this is due to the challenges that one faces in dealing with protein binding epitopes.<br />

However, recent advances in the understanding of the energetics and dynamics of protein binding interfaces open up<br />

a way to apply rational design approaches also for finding protein-protein interaction modulators. Here, we target the<br />

homotetramerization of the NHR2 domain within the RUNX1/ETO fusion protein using computational methods<br />

developed in our group [1, 2, 3]. Homotetramerization of the NHR2 domain leads to the onset of acute myeloid<br />

leukemia.<br />

Initially, energetic contributions of individual amino acids within the NHR2 homotetramer were analyzed. Ala<br />

substitution of amino acids with strong energetic contributions abolished tetramer formation. The resulting NHR2<br />

dimers fail to induce leukemia in mice. These studies revealed the existence of a hot spot at the NHR2 dimertetramer<br />

interface, suitable for a molecular intervention.[4] Based on the hot spot information, an 18-mer peptide was<br />

designed that inhibits NHR2 tetramerization. This peptide was used as a model compound to establish in vitro<br />

assays. Using the hot spot and peptide information, small-molecule inhibitors of the NHR2 tetramerization were<br />

identified by virtual screening and validated by several in vitro assays. Some of these compounds led to the induction<br />

of differentiation markers and apoptosis in NHR2-dependent cell lines. These compounds may thus be potential<br />

chemotherapeutics in the case of acute myeloid leukemia.<br />

References:<br />

1. Kruger DM, Gohlke H, Nucl. Acids Res. 2010, 38: W480-W486.<br />

2. Gohlke H, Kiel C, Case DA, J. Mol. Biol. 2003, 330: 891-913.<br />

3. Metz A et al., J. Chem. Inf. Model. <strong>2012</strong>, 52, 120-133.<br />

4. Wichmann C et al., Blood 2010, 116: 603-613.<br />

22 Sessionvorträge Moleküle


Docking to GPCRs: ligand diversity, efficacy and selectivity<br />

Kolb P 1 ; Rosenbaum D M 2 ; Schmidt D 1 ; Bernat V 3 ; Tschammer N 3 ; Kobilka B K 2 ; Shoichet B K 4<br />

1 Pharmaceutical Chemistry, Philipps-University Marburg, Marbacher Weg 6, 35039 Marburg, Germany<br />

2 Molecular and Cellular Physiology, Stanford University School of Medicine, 279 Campus Drive, Stanford, CA 94305, USA<br />

3 Chemistry and Pharmacy, Friedrich Alexander University Erlangen, Schuhstraße 19, 91052 Erlangen, Germany<br />

4 Pharmaceutical Chemistry, University of California, 1700 4th Street, Box 2550, San Francisco, CA 94158, USA<br />

With GPCRs becoming amenable to crystallization in the recent past [1], structure-based computational approaches<br />

to ligand discovery have become possible. As only about 1% of the GPCRome has been solved, however, receptor<br />

structure prediction is equally important. I will discuss the usage of x-ray structures and homology models for docking<br />

and highlight some of the conclusions that can be drawn from three recent docking campaigns.<br />

Campaign 1 used the recently determined X-ray structure of the β2-adrenergic receptor [2] to investigate the<br />

advantages and limitations inherent in a structure-based approach to ligand discovery against this and related GPCR<br />

targets. Approximately 1 million commercially available, “lead-like” molecules were docked against the β2-adrenergic receptor structure. On testing of 25 high-ranking molecules, 6 were active with binding affinities


Molecular dynamic simulation of a symmetric active state hβ2R-Gαβγ-homodimer complex –<br />

structural and energetical analysis<br />

Straßer A1 ; Wittmann H-J2 1 Department of Pharmaceutical/Medicinal Chemistry II, University of Regensburg, 93040 Regensburg, Germany<br />

2 Faculty of Chemistry and Pharmacy, University of Regensburg, 93040 Regensburg, Germany<br />

Homo- and heterodimers of several GPCRs are suggested by a large number of experimental studies [1]. Most<br />

experimental studies give hint for asymmetric dimers, consisting of two receptor protomers, coupling to only one<br />

heterotrimeric G protein [1]. However, there is only hint, but no clear experimental evidence for a symmetric<br />

homodimer, consisting of two receptor protomers, each coupling to one heterotrimeric G protein [1].<br />

Based on the crystal structure of the κ-opiod receptor [2] and bovine rhodopsin [3], we manually aligned two active<br />

hβ2R-Gαβγ-complexes [4] in a symmetric manner. For refinement of the resulting hβ2R-Gαβγ-homodimer, we<br />

performed an extensive scan of the potential energy surface in an analogous manner, as already described [5].<br />

Thereby, one hβ2R-Gαβγ-complex was translated along and rotated around the x-, y- and z-axis. Subsequently, the<br />

resulting hβ2R-Gαβγ-homodimers were energetically minimized and the complex with smallest potential energy was<br />

embedded into a POPC lipid bilayer and molecular dynamic simulation of the hβ2R-Gαβγ-homodimer (Fig. 1A) was<br />

performed, including lipid, sodium and chloride ions, extra- and intracellular water.<br />

The MD simulation revealed strong van-der-Waals interactions between both hβ2 receptors. POPC molecules placed<br />

between both hβ2 receptors stabilize the hβ2R-hβ2R-interaction (Fig. 1B). Additionally, the symmetric active state<br />

hβ2R-Gαβγ-homodimer is strongly stabilized by electrostatic and hydrogen bond interaction between the Gα- and<br />

Gβ-subunits of complex 1 and 2 (Table 1). Furthermore, hydrogen bond and electrostatic interactions were detected<br />

between the Gβ subunits of different monomers (Table 1).<br />

Table 1: Interactions within the hβ2R-Gαβγ-homodimer<br />

hβ2R (1) –<br />

hβ2R (2)<br />

Gα (1) –<br />

Gβ (2)<br />

Gβ (1) –<br />

Gβ (2)<br />

hβ2R (1)–<br />

Gαβγ (2)<br />

no.<br />

of H<br />

bonds<br />

dist.<br />

don-acc<br />

< 0.35<br />

nm<br />

Eelec.(SR)<br />

[kJ/mol]<br />

EvdW<br />

[kJ/mol]<br />

3 3 -288 ± 36 -329 ± 15<br />

6 7 -543 ± 46 -142 ± 15<br />

3 2 -135 ± 44 -105 ± 12<br />

0 0 -5 ± 4 -4 ±1<br />

The molecular dynamic simulation revealed a stable symmetric active state hβ2R-Gαβγ-homodimer. Thus, the<br />

suggested homodimerization between two hβ2 receptors, based on experimental studies [6], could be also found in<br />

silico. The amino acids, determined in silico to be responsible for the interaction between both hβ2R-Gαβγcomplexes<br />

represent a good starting point for further experimental studies.<br />

References:<br />

1. Kamal, M.; Maurice, P.; Jockers, R.: Pharmaceuticals 2011, 4 273-284.<br />

2. Wu, H. et al.: Nature <strong>2012</strong>, doi:10.1038/nature10939.<br />

3. Park, J.H. et al.: Nature 2008, 454 183-187.<br />

4. Rasmussen, S.G.F. et al.: Nature 2011, doi:10.1038/nature10361.<br />

5. Strasser, A.; Wittmann, H.-J.: J Comput Aided Mol Des 2010, 24 759-769.<br />

6. Angers, S. et al.: PNAS 2000, 97(7)3684-3689.<br />

24 Sessionvorträge Moleküle


Session Molekulare Werkzeuge<br />

Chemoproteomic target deconvolution of bioactive molecules<br />

Bantscheff M<br />

Cellzome AG, Meyerhofstrasse 1, 69117 Heidelberg, Germany<br />

Chemoproteomics represents an emerging research discipline at the interface of medicinal chemistry, biochemistry,<br />

and cell biology focused on studying the molecular mechanisms of action of drugs and other bioactive small<br />

molecules. In this presentation, experimental strategies in current chemical proteomics research will be described<br />

and illustrated using recent examples of successful applications (e.g. [1,2]). Finally, areas in drug discovery where<br />

chemical proteomics-based assays using native endogenous proteins are expected to have substantial impact will be<br />

highlighted.<br />

References:<br />

1. Bantscheff et al. Nat Biotechnol. 2011, 29(3): 255-65.<br />

2. Dawson et al., Nature 2011, 478(7370): 529-33.<br />

Microwave-Enhanced Synthesis of Fluorescent Dyes for Pharmacological Purposes Using Real-<br />

Time Observation by UV/Vis Spectroscopy<br />

Heller E1 ; Lohse M J2 ; Hoffmann C2 ; Holzgrabe U1 1 Institute for Pharmacy and Food Chemistry 1, Am Hubland, D-97074 Würzburg, Germany<br />

2 Institute for Pharmacology and Toxicology 2, Versbacher Straße 9, D-97078 Würzburg, Germany<br />

Fluorescent dyes play an important role as markers to investigate biological signalling pathways or to study binding<br />

of active agents to receptors. It is possible to label proteins with 2 dyes (FRET technology, e.g. FlAsH and ReAsH<br />

[1,2]) or to mark the active agent with one dye [3]. Tagging of active agents with fluorescent dyes is an alternative<br />

method for radioactive marking.<br />

Here we aimed to synthesize several dyes of different structural classes, e.g. substituted fluoresceins, pyrilium and<br />

pyridine dyes, by means of microwave irradiation. Microwave-enhanced reactions are rather fast in comparison to<br />

conventional heating and hence, the reaction time can be shortened from hours or days to minutes. Real time<br />

monitoring of the reaction progress is often difficult, because classical monitoring (e.g. HPLC, TLC and NMR) can<br />

take up to 30 minutes analysis time. This can cause overreaction or decomposition of the product. We created a<br />

novel method which allows a real-time observation of the reaction progress in the microwave cavity. This allows<br />

detecting the exact end-point of the reaction [4] and avoids the formation of side products. The knowledge was used<br />

to speed up the synthesis of FlAsH and analogues. Additionally the dyes were obtained with excellent purity and high<br />

yields.<br />

References:<br />

1. Hoffmann, C. Lohse M. J.: BIOspektrum 2006, 12(5): 495-497.<br />

2. Lohse, M. J. et al.: Curr. Opin. Pharm. 2007, 7, 547-553.<br />

3. Schneider, E. et al.: Chembiochem. 2007, 8(16), 1981-1988.<br />

4. Heller, E. et al.: Eur. J. Org. Chem. 2010, 19, 3569-3573.<br />

Sessionvorträge Moleküle 25


Molecular tools for positron emission tomography (PET): Development of a highly selective<br />

radiotracer for the detection of neurotensin receptor 1 positive tumors<br />

Lang C<br />

Department of Chemistry and Pharmacy, Friedrich Alexander University Erlangen-Nürnberg (FAU), Schuhstr.19, D-91052 Erlangen, Germany.<br />

Neurotensin (NT) is an endogenous, 13 amino-acid peptide that mediates a number of pharmacological effects<br />

involving dopamine transmission, analgesia, and hypothermia [1]. In addition to these attributes NT acts as a key<br />

player in multiple steps of cancer progression in numerous types of cancer cells.[2] Those carcinogenic effects are<br />

presumably evoked by an abnormal expression of the neurotensin receptor 1 (NTS 1). Based on this overexpression<br />

we aimed for a specific labeling of tumors by molecular imaging techniques such as positron emission tomography<br />

(PET).<br />

In collaboration with the Clinic of Nuclear Medicine of FAU, we have reported on the design and synthesis of a<br />

radiolabeled derivative of the hexapeptide NT 8-13, the active fragment of the endogenous agonist neurotensin [3]. As<br />

an extension of these investigations, we planned to construct a 18F-labeled non-peptidic NTS 1 selective ligand based<br />

on the potent NTS antagonist SR142948A [4]. We herein report on the novel PET-Tracer 18F-LC40, which could be<br />

synthesized by taking advantage of our click chemistry based ligation of 2-deoxy-2-[ 18F]fluoroglucosyl azide (azido-<br />

[ 18F]FDG) to the alkyne functionalized analog of the lead structure. Receptor binding experiments using NTS 1<br />

expressing CHO cells indicated a Ki value for F-LC40 of 0.5 nM with 70 fold selectivity towards NTS 2. The precursor for<br />

18F-labeling was obtained by a palladium-catalyzed aminocarbonylation [5] of the respective bromoarene derivative with<br />

an alkynylamine using Mo(CO)6 as carbon monoxide source. The CuAAC using azido-[ 18F]FDG afforded the glycosyl<br />

ligand 18F-LC40 in a radiochemical yield of 20% in a total synthesis time of 75 min.<br />

The µPET studies using HT29 (human colon carcinoma cell line) xenografted nude mice demonstrated specific binding<br />

of 18F-LC40 in vivo. Furthermore, the tracer displayed a good tumor to blood ratio and excellent metabolic stability both<br />

in vitro and in vivo.<br />

O<br />

O<br />

O<br />

N N<br />

O<br />

NH COOH<br />

N<br />

18 F-LC40<br />

N<br />

N<br />

N<br />

N<br />

O OH<br />

OH<br />

References:<br />

1. for review, see: Myers, R et al., ACS Chem. Biol. 2009, 4(7): 503–525<br />

2. for review, see: Dupouy, S et al., Biochimie. 2011, 93: 1369–1378<br />

3. Maschauer, S et al., Angew. Chem. Int. Ed., 2010, 49(5): 976–979<br />

4. Labeeuw, B et. Al, 1996, Patent WO 96/3282.<br />

5. Appukkuttan, P et al., Tetrahedron Lett., 2008, 49 : 5625–5628<br />

18 F<br />

OH<br />

26 Sessionvorträge Moleküle


Analogs of siderophores and mussel adhesion proteins for applications in clinical hygiene and<br />

implant medicine<br />

Maison W1 ; Franzmann E2 ; Khalil F1 1 <strong>Universität</strong> Hamburg, Pharmaceutical and Medicinal Chemistry, Bundesstr. 45, 20146 Hamburg, Germany<br />

2 Justus-Liebig-<strong>Universität</strong> Giessen, Organic Chemistry, Heinrich-Buff-Ring 58, 35390 Giessen, Germany<br />

The chemical modification of material surfaces by molecular self-assembly of functionalized anchor molecules is an<br />

attractive concept for various applications in implant medicine, bone engineering and clinical hygiene. The major<br />

advantages of this principle for the generation of functional surfaces are a) the availability of efficient and scalable<br />

coating techniques (compatible with large surface areas and three dimensional objects), b) the generation of<br />

chemically well-defined surfaces and c) the need of relatively low amounts of anchor molecules.<br />

Recently, catecholates have received considerable interest as reagents for the mild modification of metal and metal<br />

oxide surfaces by convenient dip-and-rinse protocols.[1-3] This approach may be considered biomimetic, because<br />

catecholate moieties are key components of natural metal-binders such as mussel adhesion proteins. In addition,<br />

catecholates are abundant motifs in many siderophores like enterobactin.<br />

Inspired by the intriguing molecular symmetry of siderophores such as enterobactin, we present the design and<br />

synthesis of biomimetic triscatecholates with a tripodal binding motif based on adamantane scaffolds. These<br />

triscatecholates form extremely stable molecular monolayers on metal surfaces and may be covalently conjugated to<br />

any effector molecule of choice. We describe the immobilization properties of these multimeric catecholates on<br />

implant materials such as TiO2 and demonstrate antifouling properties and toxicity data for PEG (effector =<br />

polyethylene glycol) conjugates.<br />

References:<br />

1. J. H. Waite, J. H.; Tanzer, M. L.: Science 1981, 212(4498): 1038-1040.<br />

2. Brubaker, C. E., Messersmith, P. B.: Langmuir <strong>2012</strong>, 28(4): 2200-2205.<br />

3. E. Franzmann, F. et al.: Chem.-- Eur. J. 2011, 17(31): 8596-8603.<br />

Sessionvorträge Moleküle 27


Fluorescent and radiolabelled ligands as molecular tools for NPY receptors<br />

Buschauer A1 ; Keller M1 , Pluym N1 ; Kaske M1 , Erdmann D1 , Bernhardt G1 1 Lehrstuhl für Pharmazeutische/Medizinische Chemie II, <strong>Universität</strong> Regensburg, 93040 Regensburg, Germany<br />

In search for pharmacological tools for various G-protein coupled receptors (GPCRs) we explored the synthesis of<br />

fluorescent ligands and radioligands as and the applicability of the labelled compounds to cellular studies of<br />

neuropeptide Y (NPY) receptors as examples of peptidergic GPCRs. Four NPY receptor subtypes (Y1R, Y2R, Y4R,<br />

Y5R) are functionally expressed in humans. Whereas Y1R, Y2R and Y5R are more sensitive to NPY and peptide YY<br />

(PYY), the Y4R prefers the pancreatic polypeptide (PP) as the endogenous agonist.<br />

Two complementary approaches to labelled NPY receptor ligands were considered:<br />

(1) Preparation of “universal” ligands, addressing more than one receptor subtype. For instance, fluorescent<br />

derivatives of the endogenous peptides are useful molecular tools for confocal microscopy or flow cytometry [1]. The<br />

latter enables the simultaneous multiparametric determination of receptor subtype selectivities of investigational<br />

compounds under equilibrium conditions, using mixtures of cells expressing the respective receptors [1].<br />

(2) Synthesis of selective radiolabelled or fluorescent tracers, preferably nonpeptides, for the identification of NPY<br />

receptor subtypes on cells and in tissues, and for the determination of binding data. Special attention was paid to<br />

labelling strategies based on side chain modifications at the ε-amino group of lysine in peptides such as NPY (Y1,2,5R<br />

agonist) or [K4 ]hPP (Y4R agonist), and, in particular, at the guanidino group of arginine, both in peptides and<br />

nonpeptides [1-7].<br />

So far, the functionalisation of arginine at NG has been largely neglected in the design of GPCR ligands. However,<br />

previous studies from our laboratory demonstrated that the acylation of strongly basic guanidine residues, which are<br />

characteristic of numerous biologically active compounds, is a general, very effective biosisosteric approach to retain<br />

(or even increase) the pharmacological activities and to substantially improve the drug-like properties by lowering the<br />

basicity of GPCR ligands. This also paved the way to radioactive ( 3H, 18F) tracers and fluorescent ligands.<br />

Tritiated high affinity Y1R [5, 6] and Y2R antagonists were accessible by coupling of amino-functionalized guanidine<br />

derivatives with succinimidyl [2,3-3H]propanoate. By analogy, prototypical [ 18F]-PET ligands for the detection of Y1Rs,<br />

e.g. on breast cancer cells, were obtained. Interestingly, space-filling fluorophores were tolerated as well. To improve<br />

the signal-to-noise ratio, red-emitting fluorophores, e. g. cyanine or pyrylium dyes, or dyes emitting in the near<br />

infrared were preferred for coupling with argininamide-type Y1R [2, 7] and Y2R antagonist core structure by linkers<br />

attached to the guanidine group. This approach resulted in receptor affinities in the one- to two-digit nanomolar<br />

range. Labelling of short C-terminal peptides, modified, e.g., with cyclic β-amino acids, gave selective fluorescent tool<br />

for the Y4R. Green emitting ligands were designed with respect to colocalization studies.<br />

The presented strategies resulted in valuable pharmacological tools for the detection of the receptor of interest in<br />

vitro and/or in vivo as well as for the characterisation of receptors and ligands in radiochemical and fluorescencebased<br />

assays, including flow cytometry and confocal microscopy.<br />

References:<br />

1. Schneider, E. et al.: Chembiochem 2006, 7: 1400-1409.<br />

2. Schneider, E. et al.: Chembiochem 2007, 8: 1981-1988.<br />

3. Pluym, N. et al.: ChemMedChem 2011, 6: 1727–1738.<br />

4. Ziemek et al.: J. Recept. Signal Transduct. 2007, 27: 217.<br />

5. Keller, M. et al.: J. Med. Chem. 2008, 51: 8168-8172.<br />

6. Keller, M. et al.: ChemMedChem 2011, 6: 1566-1571.<br />

7. Keller, M. et al.: Bioorg. Med. Chem. 2011, 19: 2859-2878.<br />

28 Sessionvorträge Moleküle


Session IVIVC: Modelle und Modellierungen<br />

From Mice to Men: Application and Impact of in vivo Modeling and Simulation in Drug Research and<br />

Development<br />

Lehr T<br />

1 Clinical Pharmacy, Saarland University, Campus Building C2 3, Room 2.33, 66123 Saarbrücken, Germany<br />

The application and implementation of modeling and simulation in drug research and development has significantly<br />

changed in the past decades. The enhancements were and are mainly triggered by improved computational<br />

resources, the increased implementation of conceptual biological understanding, and the need to optimize the drug<br />

research and development process. Nowadays, modeling and simulation techniques are frequently applied, whereas<br />

the level of detail of these models can differentiate between empirical and mechanistic. However, the level of detail<br />

and the model type applied is highly variable and depending on the stage of the drug research/development process,<br />

the a priori knowledge available (i.e. in vitro or preclinical data) and the question that needs to be answered. Overall,<br />

there is still an urgent need for a more efficient drug research and development process. The application of modeling<br />

and simulation might be a cornerstone to meet this need.<br />

The objective of this presentation is to provide a brief overview about the application of modeling and simulation in<br />

the drug research and development process. The concept and impact of modeling and simulation shall be illustrated<br />

by selected examples.<br />

Sessionvorträge Moleküle 29


Estimating the In Vivo Release Behaviour of Extended- and Immediate-Release Products using the<br />

Biorelevant Dissolution Stress Testers<br />

Garbacz G 1,2 , Koziolek M 1 , Klein S 1 , Weitschies W 1<br />

1 University of <strong>Greifswald</strong>, Department of Pharmaceutics and Biopharmaceutics, Felix-Hausdorf-Straße 3, 17489 <strong>Greifswald</strong>, Germany<br />

2 Physiolution GmbH, Walther-Rathenau-Strasse 49a, 17489 <strong>Greifswald</strong>, Germany<br />

Introduction<br />

The estimation of the in vivo drug delivery behavior oral dosage forms by in vitro dissolution tests is a prerequisite for<br />

successful development of immediate release (IR) and modified release (MR) formulations. Unfortunately, the<br />

relevant aspects of the physiology of the gastro-intestinal (GI) tract cannot be simulated realistically using<br />

compendial dissolution tests. Thus, the development of predictive in vitro test systems capable of biorelevant<br />

simulation of GI transit conditions appears mandatory for rational dosage form development and characterization.<br />

Materials and methods<br />

In order to improve the predictive power of dissolution testing, we developed a novel dissolution test apparatuses<br />

mimicking the physical conditions of the fasting gastrointestinal passage of IR and MR dosage forms which is<br />

enabled by application of different dissolution vessels [1]. Parameters that can be depicted by our devices include GI<br />

pressure exerted by gastrointestinal (GI) motility, shear stresses generated during phases of GI-transport,<br />

temperature changes, media flow and emptying patterns and intermittent contact with intestinal fluids.<br />

Results<br />

The test results obtained for MR formulations indicate the susceptibility of modified release dosage forms towards<br />

mechanical stresses of biorelevant intensity [2, 3]. Moreover, the relationship between the mechanical resistance of<br />

MR tablets and the physico-chemical properties of the dissolution media was demonstrated. Our test results for IR<br />

formulations clearly indicated that both the dosage form disintegration and the dissolution are impacted by<br />

mechanical stress events of biorelevant intensity, temperature changes and different media flow conditions as they<br />

may occur in the fasting stomach.<br />

Conclusion<br />

The study results confirmed the usability of this novel method and the corresponding test protocols for the estimation<br />

of the in vivo drug delivery characteristics of solid oral dosage forms. Moreover, the study demonstrated the<br />

applicability of the novel methods for the identification of undesired drug release behavior in vivo.<br />

Acknowledgments: Federal Ministry of Education and Research (GASTROMAX, FKZ 13 N11368-370) is kindly acknowledged for the financial<br />

support.<br />

References:<br />

1. Garbacz, G., et al., Eur. J. Pharm. Biopharm. 2008, 70(2): 421-482.<br />

2. Garbacz, G., et al., Expert Opin. Drug. Deliv. 2010, 7(11): 1251-1261.<br />

3. Garbacz, G., et al., J. Pharm. Pharmacol. <strong>2012</strong>, 64(7): 944-968.<br />

30 Sessionvorträge Moleküle


Improved input parameters for diffusion models of skin absorption<br />

Hansen S1 ; Schaefer U2 1 Helmholtz Institute for Pharmaceutical Research Saarland, Saarland University, 66123 Saarbruecken, Germany<br />

2 Biopharmaceutics and Pharmaceutical Technology, Saarland University, 66123 Saarbruecken, Germany<br />

To use a diffusion model for predicting skin absorption requires accurate estimates of input parameters on model<br />

geometry, affinity and transport characteristics [1]. We determined input parameters for diffusion models of skin<br />

absorption focusing on partition and diffusion coefficients. These include experimental methods, extrapolation<br />

approaches, and correlations that relate partition and diffusion coefficients to tabulated physico-chemical solute<br />

properties.<br />

Exhaustive databases on lipid-water and corneocyte protein-water partition coefficients were collected and analyzed.<br />

Based on these improved equations were deduced which allow the prediction of solute partitioning into corneocytes<br />

and lipids from the octanol water partition coefficient logKow and improve predictions of the macroscopic SC-water<br />

partition coefficient.<br />

With diffusion coefficients the situation is more complex. Due to the still ongoing discussion of lipophilic and polar<br />

absorption routes across the SC the representation of these pathways varies greatly between individual models<br />

which therefore require very different diffusion coefficients or mass transfer coefficients. A plethora of theories has<br />

been used to obtain estimates. As a common principle all diffusion and mass transfer coefficients are inversely<br />

related to a measure of molecular size. The strength of this size dependence may be different (less pronounced in an<br />

aqueous environment than in the highly organized SC lipid bilayers, more pronounced in bilayer crossing than in<br />

lateral diffusion).<br />

In order to improve modeling of skin absorption from realistic (i.e. finite) doses it is essential to consider slow<br />

equilibrating processes such as reversible protein binding [2, 3]. High keratin binding and slow desorption kinetics are<br />

responsible (among other factors) for the formation of the stratum corneum reservoir reported especially in the<br />

context of corticosteroid absorption. We can demonstrate that equilibrium binding and the rate of desorption are both<br />

functions of solute lipophilicity. Our results prove that slow desorption from keratin may be a major contributor to the<br />

stratum corneum reservoir. Also they prove that reservoir formation is relevant for lipophilic solutes independent of<br />

drug class, thus allowing new options for topical pharmacotherapy.<br />

From these studies we can conclude that in order to improve modeling of skin absorption in the future diffusion<br />

models should include the vertical stratum corneum heterogeneity, slow equilibration processes, the absorption from<br />

complex non-aqueous formulations, and an improved representation of dermal absorption processes. This will<br />

require input parameters for which no suitable estimates are yet available.<br />

References:<br />

[1] S. Hansen, C.M. Lehr, U.F. Schaefer, Improved input parameters for diffusion models of skin absorption, Advanced Drug Delivery<br />

Reviews, (<strong>2012</strong>) accepted.<br />

[2] S. Hansen, D. Selzer, U.F. Schaefer, G.B. Kasting, An extended database of keratin binding, Journal of Pharmaceutical Sciences, 100<br />

(2011) 1712-1726.<br />

[3] S. Seif, S. Hansen, Measuring the stratum corneum reservoir: desorption kinetics from keratin, Journal of Pharmaceutical Sciences,<br />

accepted (<strong>2012</strong>).<br />

Sessionvorträge Moleküle 31


Interindividual gastrointestinal erosion and inter- as well as intraindividual gastrointestinal transit<br />

time variability of hydrophilic matrix tablets<br />

Jain A K1 ; Abrahmsson B3 ; Abrahmsén-Alami S3 ; Anschütz M2 ; Blume H2 ; Donath F2 ; Knopke C2 ;Schug B3 ;<br />

Söderlind E3 ; Tajarobi F3 , Viridé A3 , Weitschies W1 ;Welinder A3 1 Department of Biopharmaceutics and Pharmaceutical Technology, University of <strong>Greifswald</strong>, <strong>Greifswald</strong>, Germany<br />

2SocraTec R&D GmbH, Oberursel, Germany<br />

3AstraZeneca R&D Mölndal, Sweden<br />

Purpose<br />

The aim of this study was to investigate the interindividual gastrointestinal erosion rate (%/hr.) as well as variability in<br />

gastric emptying and colon arrival time of magnetically marked hydroxpropylmethylcellulose based matrix tablets.<br />

Materials and methods<br />

Table 1 Formulation composition of the hydroxypropylmethylcellulose matrix tablets<br />

Formulation 1* Formulation 2* Formulation 3* Formulation 5*<br />

Methocel K4M % (w/w) 23 10.1 - -<br />

Methocel K100 % (w/w) 17 30 40 20<br />

Dicalcium phosphate % (w/w) 57.6 57.5 57.6 77.6<br />

* Additionally all formulations contain 1.4% (w/w) Iron oxide (E172) and 1 % (w/w) Sodium Stearyl Fumarate<br />

Circular biconvex tablets of 350 mg with a diameter of 10 mm were prepared by direct compression at AstraZeneca<br />

R&D Lund, Sweden. In vivo gastrointestinal performance of tablets was investigated by magnetic marker monitoring<br />

(MMM) [1, 2] in a single centre, open label, cross over clinical study which was carried out in 5 healthy male<br />

volunteers. Formulation 1 and Formulation 2 were administered in fasted state while Formulation 3 and Formulation<br />

5 were administered under fasting as well as fed (30 min. after standard FDA breakfast) conditions. Mean, standard<br />

deviation (SD) and percentage relative standard deviation (%RSD) for gastrointestinal erosion rate, gastric emptying<br />

time and colon arrival time were calculated for each formulation as well as in each volunteer.<br />

Results<br />

Low variability in between-subject gastrointestinal erosion rate (9-29 %RSD) was observed between the subjects.<br />

However, high between-subject variability was observed for gastric emptying time (56-110% RSD) while<br />

interindividual variability was low for colon arrival time (0-39 %RSD). In only one volunteer, the Formulation 3, when<br />

administered under fed condition reached the colon at 260 min. Formulation 5 when administered under fed condition<br />

did not reach the colon in any volunteer. High intraindividual variability in gastric emptying time (59-110% RSD) was<br />

observed for all the formulations when administered in fasted state. Moderate intraindividual variability in colon arrival<br />

time (19-52 %RSD) was observed for all the formulations when administered in fasted state.<br />

Conclusion<br />

Erosion rate was found to be dependent on the formulation composition and exhibits low interindividual variability.<br />

Gastric emptying time didn’t exhibit dependency on the formulation composition and/or on the volunteers and shows<br />

high inter- as well as intra individual variability.<br />

References:<br />

1. Weitschies, W. et al.: J. Control Release. 2005, 108(2-3): 375–385<br />

2. Weitschies, W. et al.: Adv. Drug Deliv. Rev. 2005, 57(8): 1210–1222<br />

32 Sessionvorträge Moleküle


Session Moderne Trenntechniken<br />

Continuous Improvement in pharmaceutical analytics: faster, more precise and more selective<br />

Cianciulli C1 ; Redweik S1 ; Hahne T1 ; Alhazmi H1 , Xu Y2 ; Wätzig H1 1 TU Braunschweig, Institut für Medizinische und Pharmazeutische Chemie, Beethovenstr. 55, D-38106 Braunschweig, Germany<br />

2 Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, Jilin 130022, China<br />

The increasing need to properly answer to pharmaceutical and bioanalytical challenges induced tremendous<br />

progresses in electrophoresis and chromatography. Meanwhile Capillary Gel Electrophoresis (CGE), also known as<br />

Capillary Electrophoresis-Sodium Dodecyl Sulfate (CE-SDS), is established in the pharmaceutical industry replacing<br />

SDS-PAGE gel electrophoresis, for example for the purity control of monoclonal antibodies. In order to quantify these<br />

components with sufficient precision using the same quality control method, a relative standard deviation (RSD [%])<br />

for the quantitative analysis under 2% is required. A reliable and highly precise CGE method could be obtained after<br />

thorough optimisation. It was crucial to increase the sample concentration and the injection volume in order to<br />

achieve sufficiently high signal-to-noise ratios (>70). The application of hydrodynamic injection is beneficial for the<br />

precision of the method compared to the traditionally used electrokinetic one. Linearity was demonstrated and limits<br />

of detection and quantitation were estimated. Both injection modes were compared in long series runs (n=48).<br />

Furthermore, the use of an internal standard was investigated. Thus, the RSD [%] of the migration time was reduced<br />

from 0.9% to 0.2% and the RSD [%] of peak areas was greatly improved. However, the normalisation to the total<br />

area further reduced the influence of the injection error. RSDs [%] for the peak area ratios of typically between 1 and<br />

2% were provided.<br />

Affinity Capillary Electrophoresis (ACE) can nicely detect interactions of proteins by the mobility changes which occur<br />

in the presence of various ligands [1]. ACE is able to explore changes in the conformation as well as charge changes<br />

of proteins. Six repeats were done with a very good precision due to the use of a special rinsing procedure [2]. The<br />

RSDs [%] of the migration times and the mobility ratios were typically below 2%, very often below 0.2%. The<br />

influence of various charged species e.g. metal ions, such as Cu2+ , Mn2+ and Mg2+ , pharmaceutical cations as<br />

ephedrine hydrochloride and pirenzepine dihydrochloride or anions such as glutamic acid and succinic acid was<br />

tested on the migration behavior of BSA, ß-lactoglobulin and ovalbumin. Furthermore the influence of<br />

phosphorylation on protein interactions was investigated at the activation site of the extracellular signal-regulated<br />

kinase 1 (ERK1). Organic cations and metal ions showed clearly different interactions with the proteins. Together<br />

with the advancements in mass spectrometry, NMR, and in other techniques such as THz spectrometry, deeper<br />

understanding of the behaviour of biomolecules will become possible, which in turn will strongly support the<br />

discovery of new pharmaceuticals.<br />

Y. Xu greatly acknowledges the funding from the Alexander von Humboldt Foundation<br />

References:<br />

1. Busch, M. H. A. et al.: J. Chromatogr. A 1997, 777(2): 311-328.<br />

2. El-Hady, D. et al.: J. Pharm. Biomed. Anal. 2010, 52(2): 232–241.<br />

Sessionvorträge Moleküle 33


Untersuchung von Proteinen und Metaboliten - Was können moderne Massenspektrometer leisten?<br />

Sinz A1 1 Abteilung für Pharmazeutische Chemie & Bioanalytik, Institut für Pharmazie, Martin-Luther-<strong>Universität</strong> Halle-Wittenberg, Wolfgang-<br />

Langenbeck-Str.4, D-06120 Halle/Saale<br />

Die Massenspektrometrie hat sich in den vergangenen Jahren zu einer immer bedeutenderen Methode für die<br />

Analyse klinischer Proben im Hochdurchsatz entwickelt. So findet man Massenspektrometer vor allem in den<br />

Bereichen, in denen die klassischen Verfahren, wie Immunoassays, limitiert sind, wie beispielsweise in der<br />

toxikologischen Analytik oder im Therapeutischen Drug Monitoring. Ebenso besitzt die Massenspektrometrie ein<br />

großes Potential für die qualitative und quantitative Untersuchung des Proteoms und Metaboloms in<br />

Körperflüssigkeiten, worauf in diesem Beitrag näher eingegangen wird.<br />

Biotransformation of the NR2B-selective NMDA receptor antagonist ifenprodil<br />

Falck E; Wünsch B<br />

Institut für Pharmazeutische und Medizinische Chemie der Westfälischen Wilhelms-<strong>Universität</strong>, Hittorfstr. 58-62, 48153 Münster, Germany<br />

The NR2B-subunit of the NMDA receptor has a particular binding site which is named after ifenprodil (1) [1]. This<br />

compound shows a high affinity to the NMDA receptor. Its antagonistic affect can be used for the therapy of<br />

neurodegenerative diseases such as schizophrenia, epilepsy or parkinson’s disease.<br />

1<br />

The presentation will show the metabolism of ifenprodil, regarding the phase I and phase II metabolites resulting from<br />

hydroxylation and conjugation reactions.<br />

The metabolites were obtained by biotransformation with rat liver microsomes. Identification was possible using<br />

HPLC-ESI-MSn . The structures of the resulting fragments were confirmed by recording the exact mass yielding from<br />

time-of-flight detection.<br />

References:<br />

1. Williams, K.: Curr. Drug Targets 2001, 2: 285-298<br />

34 Sessionvorträge Moleküle


Macroscopic Questions and Molecular Answers: Measuring Optical Rotation in Latest Chiral<br />

Separation Techniques<br />

Thiessen A; Bertram N<br />

Anton Paar OptoTec GmbH, Albert-Einstein-Str. 5, 30926 Seelze-Letter, Germany, www.anton-paar.com<br />

Optical instruments are used in bio analytics to determine physical properties such as optical rotation or refractive<br />

index of chemical as well as biological compounds. The optical methods are among the oldest methods known, yet<br />

do the macroscopic optical properties tell a lot about the behaviour on the nano and molecular level. Technical<br />

innovations have dramatically improved the data quality of optical measurements in general and polarimeters<br />

especially within the last years, making the determination of optical rotation of chiral substances as accurate and<br />

traceable as never before.<br />

Pharmaceutical drugs often come as chiral substances. Their enantiomers may differ in their pharmacodynamic and<br />

pharmacokinetic interactions with receptors, enzymes and proteins in general. Therefore, one enantiomer – the<br />

eutomer – may have the desired pharmaceutical activity, whereas the other enantiomer – the distomer – may be<br />

inactive or even cause unwanted side effects. To ensure the desired efficacy and to avoid unwanted adverse effects<br />

of a drug, chiral substances may reliably be separated by the use of chiral HPLC columns.<br />

Polarimeters are used as chiral detectors in HPLC setups. But also after the separation process, a traceable and<br />

accurate polarimetric analysis is mandatory to verify the qualitative and quantitative separation of those enantiomers.<br />

Spectral polarimetry can be used to differentiate optically active substances by their rotational dispersion and may<br />

shed light onto molecular structures.<br />

We will present the possibilities of using polarimeters as rotation sensitive HPLC detectors and discuss principles<br />

and instrumentation for spectral polarimetry. We will then introduce novel developments in polarimetry to minimise<br />

the risk of measuring errors. This includes a live and recordable visualisation of the sample conditions inside the<br />

sample cell, avoiding errors from bubbles or particles. Saving a picture of the filling condition greatly improves the<br />

data traceability for pharmaceutical applications. The technology also indicates proper temperature distributions<br />

within the sample, raising the bar for high quality data generation in polarimetric measurements like never before.<br />

Another innovation automatically identifies the sample cell and checks its suitability for the selected method in order<br />

to avoid human error.<br />

We will briefly discuss how the enhanced traceability and data security improves compliance with pharmaceutical<br />

industry standards.<br />

Sessionvorträge Moleküle 35


Session Naturstoffe<br />

Hepatic bioactivity of phenolic natural compounds<br />

Kraus B 1 ; Decker M 1 ; Wolff H2 1 Lehrstuhl Pharmazeutische Biologie, <strong>Universität</strong> Regensburg, <strong>Universität</strong>sstrasse 31, 93053 Regensburg, Germany<br />

2 Carl Zeiss Microscopy, Kistlerhofstrasse 75, 81379 München, Germany<br />

The liver is the largest organ in the human body and manages a great number of functions. Its role as clearance<br />

organ bears the risk that substances that should be degraded or eliminated are hepatotoxic and lead to tissue<br />

damage [1]. The capacity of a substance to produce liver damage often results from the interaction of a series of<br />

complex cellular processes, involving uptake, biotransformation and elimination of the substance [2].<br />

There are a number of medicinal plants and natural compounds, e.g. silymarin/silibinin from milk thistle (Silybum<br />

marianum (L.) GAERTN.), which posess hepatoprotective properties and are used for the therapy of liver diseases [3].<br />

The mechanism of hepatoprotection by these compounds is generally by exerting multiple effects, including e.g.<br />

antioxidant, immunomodulatory, and antiinflammatory activities. In recent years Xanthohumol (XAN), a prenylated<br />

chalkone found in hop (Humulus lupulus L.) cones, has been reported to show hepatoprotective activity and to inhibit<br />

hepatic inflammation and fibrosis [4].<br />

We are interested in the hepatic bioactivity of prenylated and not prenylated chalcones from Humulus lupulus and<br />

(the putative hepatotoxic) Piper methysticum G. FORST. as well as metabolites and structural derivatives thereof,<br />

looking for structure activity-relationships. For this, we turned our attention to cellular bioavailability of XAN,<br />

visualizing and tracing its uptake, intracellular accumulation and distribution [5].<br />

In addition, we studied the hepatic bioactivity of the chalcones and metabolites. The influence of the substances on<br />

cell viability and mitotoxicity was determined and antiinflammatory mechanisms were adressed.<br />

In a further project, we analysed the potential of the flavonolignan silibinin to act hepatoprotective against tacrineinduced<br />

liver cell damage in hepatic stellate cells. We determined the capability of silibinin to reduce tacrinemediated<br />

cyto- and mito-toxicity applying either an equimolar mixture of silibinin and tacrine or a stable hybrid<br />

structure with silibinin covalently connected to tacrine [6].<br />

Acknowledgments: We like to thank Katharina Zenger and Magdalena Motyl for carrying out the cellular assays, Xinyu Chen for synthesis of<br />

the silibinin-tacrin-hybrid, Petr Jirásek and Susanne Vogel for synthesis of chalcones and metabolites, and Jörg Heilmann for fruitful discussion<br />

and support (all from Lehrstuhl Pharmazeutische Biologie, University Regensburg).<br />

References:<br />

1. Ramadori, G. et al.: J. Physiol. Pharmacol. 2008, 59 Suppl 1, 107-117.<br />

2. Guillouzo, A.: Environ. Health. Perspect. 1998, 106 Suppl 2, 511-532.<br />

3. Muriel, P., Rivera-Espinoza, Y.: J. Appl. Toxicol. 2008, 28, 93-103.<br />

4. Dorn, C., et al.: Mol. Nutr. Food Res. 2009, 54 Suppl 2:S205-13.<br />

5. Wolff, H., et al.: J. Agric. Food Chem. 2011, 59(24):12893–12901.<br />

6. Chen, X., et al.: J. Med. Chem. <strong>2012</strong>, DOI: 10.1021/jm300246n.<br />

36 Sessionvorträge Moleküle


Mode of cell death induction by pharmacological V-ATPase inhibition<br />

von Schwarzenberg K1 ; Wiedmann R M1 ; Oak P2 ; Schulz S3 ; Zischka H3 ; Wanner G4 ; Efferth T5 ; Trauner D6 ;<br />

Vollmar A M1 1Department of Pharmacy, Pharmaceutical Biology, Ludwig-Maximilians-University, 81377 Munich, Germany<br />

2Department of Pharmacy, Pharmaceutical Biology-Biotechnology, Ludwig-Maximilians-University, 81377 Munich, Germany<br />

3Institute of Toxicology, Helmholtz Center Munich, German Research Center for Environmental Health, 85764 Neuherberg, Germany<br />

4Department of Biology I, Ludwig-Maximilians-University, 82152 Martinsried, Germany<br />

5Department of Pharmaceutical Biology, Institute of Pharmacy and Biochemistry, University of Mainz, 55099 Mainz, Germany<br />

6Department of Chemistry, Ludwig-Maximilians-University, 81377 Munich, Germany<br />

Recently V-ATPase, a multisubunit proton pump, has come into focus as an attractive target in cancer cell invasion.<br />

However little is known about the role of V-ATPase in cell death and especially the underlying mechanisms remain<br />

mostly unknown. We used archazolid B, a macrolide of myxobacterial origin being a novel very potent inhibitor of the<br />

V-ATPase as an experimental drug as well as a chemical tool to decipher V-ATPase related cell death signaling. We<br />

found that archazolid induced apoptosis in highly invasive tumor cells at nanomolar concentrations while showing<br />

almost no cytotoxicity in non tumorus cells. Apoptosis was caspase-dependant and executed by the mitochondrial<br />

pathway shown by breakdown of mitochondrial potential, leakage of cytochrom c from the mitochondria and<br />

activation of caspase-9. Prior to apoptosis induction archazolid lead to the activation of stress responses such as<br />

autophagy and glycolysis. Autophagy was induced at concentrations that do not alkalize lysosomes and was shown<br />

by degradation of p62 or fusion of autophagosomes with lysosomes. Autophagy was found to be a survival<br />

mechanism at low concentrations but could not be accomplished at higher concentrations due to lysosome<br />

alkalization by V-ATPase inhibition. Archazolid also lead to glycolysis induction which is mediated by the hypoxiainducible-factor-1<br />

alpha and induced due to energy stress. This was shown by a decline of the ATP level and arrest<br />

of energy consuming processes (AMPK, mTOR, eIF2alpha). As silencing HIF1alpha increases apoptosis, glycolysis<br />

was suggested to be an adaptive mechanism. We conclude that archazolid leads to energy stress which activates<br />

autophagy and glycolysis and finally leads to apoptosis. We propose V-ATPase as a promising drugable target in<br />

cancer therapy caught up at the interplay of apoptosis, autophagy and cellular/metabolic stress.<br />

Sessionvorträge Moleküle 37


Cyanobacteria in Anticancer Natural Products Research<br />

Niedermeyer T H J1 ; Daily A2 ; Swiatecka-Hagenbruch M1 , Moscow J A2 1 Cyano Biotech GmbH, Magnusstr. 11, 12355 Berlin, Germany<br />

2 Hematology-Oncology Department of Pediatrics of the University of Kentucky, 740 South Limestone, Lexington, Kentucky 40536, USA<br />

Cyanobacteria are a prolific source of natural products. Many secondary metabolites isolated from cyanobacteria<br />

show remarkable cytotoxicity. In the first part of this talk, cyanobacteria and some of the most interesting compounds<br />

that have potential for the exploitation as lead compounds or have already been advanced into clinical studies (e.g.<br />

the cryptophycins and dolastatins) are highlighted. In the second part, microcystins are presented as novel leads.<br />

Microcystins, rather known as hepatotoxins produced by cyanobacteria, are currently also studied for their potential<br />

as leads for anticancer drug development. They potently inhibit the eukaryotic protein phosphatase families PP1 and<br />

PP2A [1]. Microcystins display their toxicity after transporter-mediated uptake by the cell. Three human proteins are<br />

thought to be able to mediate this uptake, the organic anion transporting polypeptides OATP1B1, OATP1B3, and<br />

OATP1A2. Liver cells express OATP1B1 and OATP1B3 transporters and are thus able to absorb microcystins from<br />

the blood, resulting in a pronounced liver toxicity of microcystins. OATP1B3 is also expressed in hepatocellular<br />

carcinoma cells (HCC) as well as gastrointestinal and lung tumors. Recent data offer support that the OATP1B3<br />

protein is also expressed in a significant percent of breast and colon tumors [2, 3]. Interestingly, compared to<br />

OATP1B1, OATP1B3 is found only in low abundance in liver cells. Thus selectivity that favors OATP1B3 over<br />

OATP1B1 should lead to a decreased hepatic clearance / toxicity and an increased uptake in OATP1B3-expressing<br />

tumors, creating a therapeutic window for the respective compound.<br />

Since about 90 naturally occurring microcystins with variations in the seven constituting amino acids are known and<br />

many more structures are not described yet, there is high potential for isolating variants with unique properties. In<br />

fact, in our initial screening of 20 microcystins, we have found four microcystin structural variants with an IC50 in the<br />

low nanomolecular range and with transporter selectivity that favors OATP1B3 over OATP1B1 by a factor of more<br />

than ten. Some of these microcystin variants have not yet been described in the literature.<br />

Testing of further microcystin variants, the elucidation of their structures and the deduction of structure-activity<br />

relationships will enable us to find and create more selective and potent compounds.<br />

OH<br />

HN<br />

References:<br />

1. Watanabe, M. et al., Toxic Microcystis (CRC Press) 1995.<br />

2. Muto, M. et al., Cancer Sci. 2007, 98(10): 1570–1576.<br />

3. Lee, W. et al., Cancer Res. 2008, 68(24): 10315–10323.<br />

O<br />

NH 2<br />

NH<br />

N<br />

NH<br />

38 Sessionvorträge Moleküle<br />

COOH<br />

O<br />

H<br />

N<br />

O<br />

O<br />

N<br />

H<br />

N<br />

O<br />

O<br />

COOH<br />

Structure of a typical microcystin (Microcystin LR)<br />

NH<br />

HN<br />

O


Applied phytochemical research – an industry’s perspective<br />

Stintzing F C<br />

WALA Heilmittel GmbH, Dorfstrasse 1, 73087 Bad Boll/Eckwälden, Germany<br />

An overview is given on research conducted on selected European plants used for cosmetics and complementary<br />

medicine. A first key aspect will deal with the composition of a lipophilic extract from Cydonia oblonga and its<br />

ramifications for cosmetic purposes [1]. The impact of location and harvest date on the phenolic composition at the<br />

example of Salvia officinalis [2], Alchemilla mollis and A. vulgaris [3] will be a second focal point. A third topic will<br />

cover the biotransformation of hydrophilic compounds from Betula pendula [4], Hamamelis virginiana [5,6], Nicotiana<br />

tabacum [7] and Atropa belladonna [8] upon lactic acid fermentation. Fourth, the findings obtained by an in-depth<br />

screening including lipophilic and hydrophilic compounds of Mercurialis perennis will be presented [9,10,11]. Finally,<br />

most recent data on the stability evaluation of selected essential oils during exposure to light and elevated<br />

temperatures will be shown also covering new approaches for quality control measures [12,13,14,15].<br />

The data presented demonstrate that phytochemical research on the European flora is most rewarding and deserves<br />

a more attentive dedication even on plants that are seemingly studied well. The impact of processing resulting in<br />

specific compound patterns but also differing bioactivities should encourage both industry and academia to join<br />

forces in discovering and making use of the plethora of phytochemicals in local plants.<br />

Acknowledgments: The author is grateful to all co-authors and collaborators for their common interest and enthusiasm in conducting the<br />

investigations presented.<br />

References:<br />

1. Lorenz, P. et al.: Anal. Bioanal. Chem. 2008, 391(2): 633-646.<br />

2. Schnitzler, P. et al.: Phytomed 2008, 15(1/2): 62-70.<br />

3. Duckstein, S.M. et al., Ztschr. Naturforsch C/ J. Biosci. <strong>2012</strong>: submitted.<br />

4. Millet, A., Stintzing, F., Merfort, I.: J. Pharm. Biomed. Anal. 2009, 49(5): 1166-1171.<br />

5. Duckstein, S.M.., Stintzing, F.C.: Anal. Bioanal. Chem. 2011, 401(2): 677-688.<br />

6. Duckstein, S.M. et al.: Phytochem. Anal. <strong>2012</strong>: doi.10.1002/pca.2359.<br />

7. Millet, A., Stintzing, F., Merfort, I.: J. Pharm. Biomed. Anal. 2010, 53(2): 137-144.<br />

8. Schwarzenberger, M. et al.: Pharmazie <strong>2012</strong>, 67(5): 460-466.<br />

9. Lorenz, P. et al.:, Phytochem. Anal. 2009, 21(3): 234-245.<br />

10. Lorenz, P. et al., Ztschr. Naturforsch C/J. Biosci. 2010, 65(3/4): 174-179.<br />

11. Lorenz, P. et al., Phytochem. Anal. <strong>2012</strong>, 23(1): 60-71.<br />

12. Turek, C., Stintzing, F.C.: Anal. Bioanal. Chem. 2011, 400(9): 3109-3123.<br />

13. Turek, C., Stintzing, F.C.: J. Food Sci. 2011, 76(9): C1365-C1375.<br />

14. Turek, C., Stintzing, F.C.: Food Res. Int. <strong>2012</strong>, 46(1): 341-353.<br />

15. Turek, C., Kirschmann, N., Stintzing, F.C.: Ztschr. Arznei Gewürzpfla/J. Med. Spice Plants <strong>2012</strong>, 17(2): 73-79.<br />

Sessionvorträge Moleküle 39


Session Klinische Wirkstoffentwicklung<br />

Single- and multiple-transfected cell lines as tools for the analysis of drug transport and of the<br />

interplay of drug transport and drug metabolism<br />

König J, Fahrmayr C, Fromm M F<br />

Institute of Experimental and Clinical Pharmacology and Toxicology, Department of Clinical Pharmacology and Clinical Toxicology, Friedrich-<br />

Alexander-<strong>Universität</strong> Erlangen-Nürnberg, Fahrstrasse 17, 91054 Erlangen, Germany<br />

In liver drug disposition and effects are determined by the coordinated processes of drug uptake, drug metabolism<br />

and drug export. Following transporter-mediated drug uptake (e.g. by the organic anion transporting polypeptide<br />

OATP1B1, gene symbol SLCO1B1) and subsequent phase I (e.g. by the cytochrom P450 enzyme CYP3A4) and / or<br />

phase II metabolism (e.g. by UDP-glucuronosyltransferase 1A1) parent compounds or their metabolites are<br />

frequently exported by members of the ABC transporter superfamily (e.g. by the apically localized export pump<br />

MRP2, gene symbol ABCC2) into bile. So far, mostly single-transfected cells recombinantly overexpressing the<br />

protein of interest were used to investigate its role in drug disposition. This setup may be useful for determining the<br />

substrate spectrum of transporters or metabolizing enzymes or for the analysis of transporter-mediated drug-drug<br />

interactions but results in limited data regarding the interplay of drug transport and drug metabolism. Therefore,<br />

double- and multiple-transfected cell lines have been established expressing an uptake transporter together with an<br />

export pump [1, 2, 3] or expressing an uptake transporter simultaneously with the phase II enzyme UGT1A1 and the<br />

export pump MRP2 [4]. The latest development was the establishment of a quadruple-transfected cell line<br />

recombinantly overexpressing the uptake transporter OATP1B1, the phase I enzyme CYP3A4, the phase II enzyme<br />

UGT1A1 and the apically localized export pump MRP2. This presentation summarizes data regarding the usage of<br />

single- and multiple-transfected cell lines for the analysis of drug transport, transporter-mediated drug-drug<br />

interactions and the interplay of drug transport and drug metabolism.<br />

References:<br />

1. Cui et al.: Mol Pharmacol 2001, 60: 934-943.<br />

2. Letschert et al.: J Pharmacol Exp Ther 2005, 313: 549-556.<br />

3. Nies et al.: Naunyn Schmiedebergs Arch Pharmacol 2007, 376: 449-461.<br />

4. Fahrmayr et al.: Br J Pharmacol <strong>2012</strong>, 165: 400-406.<br />

40 Sessionvorträge Moleküle


Methods to investigate the clinical relevance of drug transporters<br />

Oswald S<br />

University Medicine of <strong>Greifswald</strong>, Center of Drug Absorption and Transport, Department of Clinical Pharmacology, Felix-Hausdorff-Str. 3,<br />

17487 <strong>Greifswald</strong>, Germany<br />

Transporter proteins are crucial determinates in the pharmacokinetics (absorption, distribution and excretion) of<br />

many drugs. Many of these transporters are highly expressed in tissues that are of outstanding relevance in drug<br />

disposition such as the intestine, the liver, the kidneys and at physiologically relevant blood-tissue barriers such as<br />

the blood-brain-barrier or the placenta.<br />

In general, one can distinguish between SLC (solute carrier) transporters which mostly facilitate the cellular uptake of<br />

their substrates, and ABC (ATP-binding cassette) transporters such as P-glycoprotein (P-gp, ABCB1) which pump<br />

their substrates out of the cell in an energy-dependent manner.<br />

So far hundreds of these transporters have been described, whereas the clinical impact of only few candidates (e.g.<br />

P-gp, OATP1B1) is well established. On the other side, there is an urgent need to know the distinct cellular transport<br />

properties of a certain drug to avoid unwanted transporter-based drug-drug interactions (by induction or inhibition),<br />

high inter-subject variability in drug disposition (e.g. by genetic variants) and safety issues.<br />

Consequently, current guidelines from regulatory agencies (e.g. FDA, EMA) recommend comprehensive in vitro<br />

characterization of new molecular entities with regards to their transporter affinity. After verification that a certain drug<br />

may be a substrate for a certain transporter protein clinical studies in humans are recommended to conclude on the<br />

potential clinical impact.<br />

The presentation will provide a brief overview about clinical state-of-the-art approaches as well as experimental<br />

studies in humans to conclude on the clinical relevance of the most important ABC and SLC transporters.<br />

Sessionvorträge Moleküle 41


The functionality of uptake transporters in primary hepatocytes of different species can be verified<br />

by estrone-3-sulfate<br />

Runge D1 ; Keiser M2 ; Ullrich A1 ; Pohl J2 ; Radebold J2 ; Damm G3 ; Nüssler A4 ; Siegmund W2 1PRIMACYT Cell Culture Technology GmbH, Schwerin, 19061, Federal Republic of Germany<br />

2Department of Clinical Pharmacology, University Medicine of <strong>Greifswald</strong>, <strong>Greifswald</strong>, 17487, Federal Republic of Germany<br />

3Charité Virchow Klinik, Berlin, 13353, Federal Republic of Germany<br />

4BG Trauma Hospital, Tübingen, 72076, Federal Republic of Germany<br />

Introduction: Primary mammalian hepatocytes are used for several in vitro applications like testing of drug<br />

metabolism, toxicity and transporter assays. However, little is known about species specific differences or similarities<br />

in the activity of uptake and efflux transporter. Therefore, we started a species specific characterization and<br />

compared the uptake of different substrates in hepatocytes from pharmacological relevant species which are<br />

commonly used for drug testing.<br />

Materials & Methods: Human, rat, dog and monkey hepatocytes were incubated in serum free media. 2-4 days after<br />

cell isolation a time and concentration dependent uptake of [3H]- estrone-3-sulfate (E3S), -bromosulfophthaleine<br />

(BSP), -digoxine (Dig) and -taurocholic acid (TA) at 4°C and 37°C was measured by liquid scintillation counting.<br />

Competition assays were performed using E3S as substrate and rifampicin, BSP, Dig or TA as competitors.<br />

Results: All hepatocytes showed a time-dependent and saturable increase in E3S and BSP uptake at 37°C<br />

compared to the uptake at 4°C. By contrast, Dig and TA showed a time-dependent and saturable transport only in<br />

hepatocytes of humanoids and rats, but not in dog. Moreover Dig uptake was about 10 fold higher in human than in<br />

rat and monkey hepatocytes. E3S revealed a high affinity to human (Km = 12.9 ± 10.1 µmol/l; Vmax = 84.2 ± 30.3<br />

pmol/mg × min), monkey (Km = 9.2 ± 2.2 µmol/l; Vmax = 24.8 ± 3.3 pmol/mg × min) and dog (Km = 3.3 ± 2.3 µmol/l;<br />

Vmax = 10.0 ± 2.1 pmol/mg × min). In rat hepatocytes E3S showed an increased uptake with increasing<br />

concentrations but no kinetic data could be calculated. For BSP and Dig no pharmacokinetic data could be calculated<br />

in any species and TA was transported only into hepatocytes of monkey (Km = 17.5 ± 12.5 µmol/l; Vmax = 5.8 ± 2.1<br />

pmol/mg × min). Rifampicin clearly inhibited the E3S uptake in monkey (IC50 = 7.9 (2.4; 25.9) µmol/l) and dog<br />

hepatocytes while uptake of E3S in rat hepatocytes was not influenced. E3S uptake was also inhibited by increasing<br />

concentration of BSP in dog (IC50 = 7.5 (5.1; 11.1) µmol/l), monkey (IC50 = 5.9 (3.1; 11.3) µmol/l) and rat (IC50 = 200<br />

(177; 226) µmol/l) hepatocytes. Dig and TA did not significantly influence the E3S uptake in primary hepatocytes in<br />

dog, rat and monkey hepatocytes.<br />

Discussion: Our data suggest that among all substrates tested E3S is the most suitable substrate to verify the<br />

functionality of uptake transporters in primary hepatocytes of humanoids, dogs, and rats. However, major differences<br />

were observed in the uptake of drugs within the species tested here. These species specific differences in<br />

hepatocellular uptake should receive attention when data obtained in drug transport, metabolism and toxicity assays<br />

are transferred from animal experiments to humans.<br />

This work was supported by the Ministry of Economy, Labour and Tourism of Mecklenburg-Vorpommern (Grant V630-F-108-2011/040 and<br />

V630-VB-196-2011/038).<br />

42 Sessionvorträge Moleküle


Solubilizing agents influence the pharmacokinetics of probe drugs by transporter interaction<br />

Engel A1 ; Keiser M1 ; Oswald S1 ; Scheuch E1 ; Berg S2 ; Freyse E-J2 ; Weitschies W3 ; Siegmund W1 1 Institute of Pharmacology, Dpt. of Clinical Pharmacology, Felix-Hausdorff-Str. 3, 17487 <strong>Greifswald</strong>, Germany<br />

2 Institute of Diabetes “Gerhardt Katsch” e.V., <strong>Greifswald</strong>er Str. 11e, 17495 Karlsburg, Germany<br />

3 Institute of Pharmacy, Dpt. of Biopharmaceutics and Pharmaceutical Technology, Felix-Hausdorff-Str. 3, 17487 <strong>Greifswald</strong>, Germany<br />

Background & Aims: Pharmaceutical excipients are supposed to be pharmacologically inactive by definition.<br />

However, there is ample evidence that solubilizing agents (e.g. Cremophor ® EL) might significantly influence CYP450<br />

dependent drug metabolism and efflux transport by ABCB1 and ABCC2 [1, 2, 3]. So far, little is known about the<br />

influence of pharmaceutical excipients on uptake transporters, which were shown to be involved in the disposition of<br />

many drugs. Therefore, we investigated the influence of polyethylene glycol 400 (PEG), hydroxypropyl-ß-cyclodextrin<br />

(HPCD), Solutol ® HS15 (SOL) and Cremophor ® EL (CrEL) on the organic anion-transporting polypeptides (OATP)<br />

1A2, 1B1, 1B3 and 2B1 and on the Na + /taurocholate cotransporting polypeptide (NTCP) in vitro. Furthermore, we<br />

studied the impact of PEG, HPCD and SOL on the pharmacokinetics of the probe drugs acetaminophen (AAP),<br />

talinolol (TAL) colchicine (COL) and cyclosporine A (CSA) in rats.<br />

Materials & Methods: In vitro inhibitory effects of the excipients were studied in competition assays using HEK293<br />

cells stably transfected with OATP1A2, 1B1, 1B3, 2B1 and NTCP, respectively, and in each case two established,<br />

radio-labelled reference substrates. Intracellular accumulation of taurocholic acid (TA; OATP1A2, NTCP), estrone-3sulfate<br />

(E3S; OATP1A2, 1B1, 2B1), bromosulfophthaleine (BSP; OATP1B1, 1B3, 2B1, NTCP) and estradiol-17ßglucuronide<br />

(E17ßGln; OATP1B3) was measured by liquid scintillation counting after cell lysis. For pharmacokinetic<br />

investigations, solutions of each probe drug with each excipient were administered intravenously as single or multiple<br />

doses to male Wistar rats (each N=6). Drug in saline served as control. Blood samples, urine, feces and organs were<br />

collected and drug concentrations determined by LC-MS/MS or HPLC-UV.<br />

Results: In vitro, PEG selectively inhibited the uptake of TA and E3S by OATP1A2 (IC50 0.05% and 0.14%) but not by<br />

1B1, 1B3, 2B1 and NTCP. HPCD strongly inhibited the TA, E3S and E17ßGln uptake by OATPs (IC50 0.001 –<br />

0.24%). SOL and CrEL were potent inhibitors of all investigated transporters with the strongest effect on the TA, BSP<br />

and E3S uptake by OATP1A2 and OATP2B1 (IC50 0.0003 – 0.01%). In vivo, all investigated excipients were able to<br />

significantly modulate the pharmacokinetics of at least one probe drug. Pharmacokinetics of CSA were most<br />

strikingly influenced by all three solubilizing agents, its single and multiple dose AUC was increased 2-5-fold and<br />

volume of distribution (VD) was reduced to a similar degree while the elimination half-life (t1/2) remained unchanged.<br />

SOL likewise affected AUC and VD of COL and AAP, but to a lesser extent. It also prolonged t1/2 of TAL and COL 2-3<br />

times. HPCD increased VD of TAL, supported by a dramatic accumulation of TAL in kidneys and duodenum.<br />

Furthermore t1/2 of TAL was doubled by HPCD. PEG only caused minor changes in the kinetics of TAL and COL but<br />

significantly increased renal clearance of unchanged AAP and reduced renal clearance of AAP conjugates.<br />

Conclusion: Frequently used solubilizing agents were shown to substantially influence intestinal and hepatic uptake<br />

transporters in vitro and to significantly modulate the pharmacokinetics of drugs in the rat. This might lead to<br />

unwanted drug interactions or adverse drug effects, which should be considered in drug development. However, the<br />

clinical relevance should be evaluated in further investigations.<br />

References:<br />

[1] Bravo González, R.C. et al.: Biopharm. Drug Dispos. 2004, 25(1): 37–49.<br />

[2] Hanke, U. et al.: Eur. J. Pharm. Biopharm. 2010, 76(2): 260–268.<br />

[3] ten Tije, A. J. et al.: Clin. Pharmacokinet. 2003, 42(7): 665–685.<br />

Sessionvorträge Moleküle 43


Sessionvorträge Targets<br />

Session Proteomics und Metabolomics<br />

OMICs approaches in the analysis of population-based cohorts – Potential avenues for biomarker<br />

screening<br />

Völker U, Ameling S1 , Endlich K2 , Felix S B3 , Gesell-Salazar M1 , Hammer E1 , Hoffmann W4 , Homuth G1 , Jehmlich N1 ,<br />

Kocher T5 , Lerch M M6 , Schurmann C1 , Nauck M7 , Schmidt F1 , Teumer A1 , Völzke H4 , Wallaschofski W7 1 Interfaculty Institute of Genetics and Functional Genomics<br />

2 Institute of Anatomy and Cell Biology<br />

3 Clinic for Internal Medicine B<br />

4 Institute for Community Medicine<br />

5 Department of Restorative Dentistry, Periodontology and Endodontology<br />

6 Clinic for Internal Medicine A<br />

7 Institute for Clinical Chemistry and Laboratory Medicine, University Medicine <strong>Greifswald</strong>, D-17475 <strong>Greifswald</strong>, Germany<br />

Different OMICs approaches are currently widely applied in the screening for diagnostic and prognostic biomarkers in<br />

case-control settings or for the exploration of the pathophysiology of different diseases. On the other side populationbased<br />

cohorts provide an extremely valuable resource particularly for the study of prevalent diseases. One such<br />

study, the Study of Health in Pomerania (SHIP), has been established in the last 15 years at the University of<br />

<strong>Greifswald</strong> and it is internationally acknowledged for its comprehensive high-quality phenotyping and quality control<br />

measures (1). The first wave (SHIP-0) was a cross-sectional epidemiological survey in West Pomerania, in the<br />

North-East of Germany, conducted from 1997-2001 and comprising 4310 participants. Two additional waves (SHIP-1<br />

and 2) with expanded phenotyping were conducted between 2001-2006 and 2008-<strong>2012</strong>. Finally, a separate stratified<br />

random sample of 8016 adults aged 20 to 79 years was drawn for SHIP-TREND for which examination started in<br />

2009 and until October <strong>2012</strong> more than 3700 subjects participated.<br />

Recently, we started to perform different OMICs analyzes with biomaterials of SHIP and SHIP-TREND, which started<br />

with genome-wide genotyping and metabolome analysis of urine. Joining epidemiological, clinical and functional<br />

genomics expertise we are now active partners of international consortia addressing the genetics of cardiovascular<br />

diseases (2, 3).<br />

The OMICs analyses are currently being expanded to include metabolomics measurements of plasma, expression<br />

profiling of whole blood and proteomics approaches for the characterization of biofluids such as plasma, urine and<br />

saliva. In the talk the progress in the analysis of the two population-based cohorts using OMICs technologies will be<br />

presented.<br />

The second part will be used to demonstrate, how such population-based cohorts can be used to provide reference<br />

values in case-control settings particularly in the field of cardiovascular research. The application of these<br />

technologies will be illustrated using dilated cardiomyopathy as an example (4).<br />

References:<br />

1. Völzke, H. et al.:Int J Epidemiol 2011, 40: 294-307.<br />

2. Newton-Cheh, C. et al.: Nat Genet 2009, 41: 666-676.<br />

3. Suhre, K. et al.: Nat Genet 2011, 43: 565-569.<br />

4. Ameling, S. et al.: Eur Heart J <strong>2012</strong>, in press.<br />

44 Sessionvorträge Targets


Drug target fishing in silico and in vitro<br />

Efferth T<br />

Department of Pharmaceutical Biology, Institute of Pharmacy and Biochemistry, Johannes Gutenberg University, Staudinger Weg 5, 55128<br />

Mainz, Germany<br />

Mechanisms of action of drugs are frequently difficult to unravel in a comprehensive fashion. Elegant techniques<br />

which appeared in recent years represent transcriptome-wide and proteome-wide expression profilings by<br />

microarrays to connect gene and protein expression signatures to drug, genes and diseases.<br />

Although clinical parameters are valuable for the determination of the prognosis of larger cohorts of patients, they are<br />

frequently less helpful to predict the treatment success for each individual patient due to inter-individual variability<br />

and heterogeneity. Therefore, personalized medicine aims to develop individualized treatment options adapted to<br />

factors relevant for the prognosis of each single patient. Molecular biomarkers identified by “-omics” technologies are<br />

expected to predict the likelihood of individual responsiveness (or toxicity) to treatment. This information can then be<br />

used to optimize treatment schedules in a custom-made fashion.<br />

However, microarray-based data do not allow conclusions on the protein targets of drugs. Considering that there is a<br />

huge amount of potentially drugable proteins, but that only a few hundred drug targets have been exploited so far,<br />

the development of drug target fishing techniques is urgently required. Identification of drug targets is mostly<br />

performed by fishing proteins with a modified drug from a protein solution isolated from cells or tissues. Activitybased<br />

probe profiling and compound-centric chemical proteomics are established methods for this purpose.<br />

Pointing to individualized cancer therapy, a current concept is to identify mutations in “driver” genes of cancer<br />

development and progression and to specifically develop small molecules or antibodies against patient-specific<br />

mutated proteins in cancer tissues. The hope is not only to improve individual response rates of tumors, but also to<br />

reduce side effects in normal tissues.<br />

In this presentation, we give an overview of current bioinformatical and wet-lab technologies to identify and verify<br />

drug targets and present selected examples.<br />

References:<br />

1. Volm, M.et al., Clin. Cancer Res. 2002, 8(6): 1843-8.<br />

2. Gillet, J.P./Efferth, T. et al., Cancer Res. 2004, 64(24): 8987-93.<br />

3. Efferth, T. et al., Mol. Cancer Ther. 2006, 5(8): 1986-94.<br />

4. Efferth, T. et al., Mol. Cancer Ther. 2008, 7(1): 152-61.<br />

5. Konkimalla, V.B., Efferth, T. Biochem. Pharmacol. 2010, 79(8): 1092-9.<br />

6. Youns, M. et al., Planta Med. 2010, 76(17): 2019-25.<br />

7. Sertel, S. et al., PLoS One <strong>2012</strong>, 7(5): e35584.<br />

8. Eichhorn, T. et al., Mol. Biosyst. <strong>2012</strong>, 8(4): 1311-8.<br />

Sessionvorträge Targets 45


From whole-organism to sub-cellular localization of metabolites by mass spectrometry imaging<br />

Liebeke M<br />

Department of Surgery and Cancer, Imperial College London, Sir Alexander Fleming Building, London, SW7 2AZ, UK<br />

(m.liebeke@imperial.ac.uk)<br />

Localization of metabolites in tissues or even within single cells is of emerging interest for comprehensive<br />

understanding of biological systems. Advances in mass spectrometry imaging (MSI) allow the mapping of individual<br />

molecules across a spatial distribution, but depending on the method used this distribution image is limited by the<br />

technical possibilities of the desired device. We report here the use of matrix assisted laser desorption ionization<br />

(MALDI) FTICR-MS to image tissue cross-sections (0.5cm 2 – 20µm resolution) and the application of secondary ion<br />

mass spectrometry (SIMS) to further elucidate the cellular distribution of selected small molecules (0.5mm 2 – 100nm<br />

resolution). Both MSI techniques gained comparable image results for the whole tissue cross-section but showed<br />

distinct differences for high spatial resolution imaging. The advantages of TOF-SIMS and MALDI MSI approaches<br />

will be pointed out, as well as problems and limitations of both techniques. Results of clinical interest (infected mouse<br />

lung) and environmental biology (earthworm) will be presented. We show the power of the whole-organism to subcellular<br />

metabolite localization, which may be able to reveal novel possible physiological implications in the analyzed<br />

organism.<br />

Figure 1 images from tissue sections generated by TOF-SIMS, observed ions indicated. Images for are presented in a gray scale from black to<br />

white. Higher brightness corresponds to higher signal intensities. Total counts (TC) correspond to the total ion signal from the specified ion.<br />

Maximum counts (MC) correspond to the maximal number of counts in a pixel. The field of view is 500 × 500 μm 2 .<br />

46 Sessionvorträge Targets


Incorporation of unnatural amino acids to monitor conformational changes in peroxisome<br />

proliferator-activated receptor alpha (PPARα)<br />

Schwarz R, Tänzler D, Ihling C H, Kölbel K, Sinz A<br />

Dept. Pharm.Chem./Bioanalytics, Institute of Pharmacy, Martin-Luther University Halle-Wittenberg, Halle/Saale, Germany<br />

Chemical cross-linking combined with an enzymatic digestion and mass spectrometric analysis of the reaction<br />

products has evolved into an alternative strategy to identify protein-protein and protein-ligand interactions. Here, we<br />

present the investigation of conformational changes in PPARα upon ligand binding by photo-affinity labeling<br />

combined with mass spectrometry. Peroxisome proliferator-activated receptors (PPARs) belong to the superfamily of<br />

nuclear receptors. PPARα plays a crucial role in lipid metabolism and poses an important target for designing<br />

antidyslipidemic drugs for the treatment of the metabolic syndrome.[1].<br />

Expression and purification of PPARα variants were optimized in respect to obtain high protein yields. The<br />

photoreactive amino acid para-benzoylphenylalanine (Bpa) was incorporated at position 258 of PPARα for<br />

conducting photo-affinity labeling studies (Figure 1). Bpa incorporation was confirmed by peptide mass fingerprint<br />

analyses. PPARα variant L258Bpa was employed for photo-cross-linking experiments in the absence and presence<br />

of the PPARα ligand GW6471. After the photo-cross-linking reaction, reaction mixtures were enzymatically digested<br />

and peptides were analyzed by high-resolution mass spectrometry (nano-HPLC/nano-ESI-LTQ-Orbitrap-MS/MS).<br />

The distance constraints imposed by the photochemical cross-links served to monitor conformational changes in<br />

PPARα upon ligand binding.<br />

Conformational changes in PPARα that had already been suggested in previous studies [2] were verified. In the<br />

presence of the PPARα antagonist GW6471 a high number of cross-linked products were identified, which were<br />

mainly localized in the region comprising amino acids 215 to 226. These cross-links can only be accomodated if the<br />

flexible omega loop in PPARα, harboring the photo-amino acid Bpa258, exerts a huge conformational change and<br />

thus closes the ligand binding pocket according to a “mouse trap” principle. Our strategy demonstrates that photoaffinity<br />

labeling followed by a mass spectrometric analysis of the reaction products allows monitoring conformational<br />

changes in target proteins in a highly efficient manner.<br />

Figure 1: Three dimensional structure (pdb: 1KKQ) of GW6471-bound PPARα; Leu-258 that was replaced by Bpa is shown in green, Ω-loop<br />

(yellow), activation function helix 2 (orange), and GW6471 (magenta).<br />

References:<br />

[1] Plutzky et al.: Am J Cardiol 2003.<br />

[2] Müller et al.: J Med Chem 2009.<br />

Sessionvorträge Targets 47


Session Transporter und Ionenkanäle<br />

TRP channels as regulators of blood pressure and fertility<br />

Freichel M<br />

48 Sessionvorträge Targets


Role of Ca 2+ -activated K + Channels in Breast Cancer Proliferation<br />

Lukowski R<br />

Sessionvorträge Targets 49


Transporters in human platelets: physiological function and impact for pharmacotherapy<br />

Jedlitschky G 1 , Rauch B 1 , Greinacher A 2 , Kroemer H K 1<br />

1Department of Pharmacology, Center of Drug Absorption and Transport (C_DAT), University Medicine <strong>Greifswald</strong>, Felix-Hausdorff-Str. 3,<br />

17487 <strong>Greifswald</strong>, Germany<br />

2Department of Immunology and Transfusion Medicine, University Medicine <strong>Greifswald</strong>, Sauerbruchstrasse, 17487 <strong>Greifswald</strong>, Germany<br />

Platelets contain many biologically active molecules, such as factors triggering platelet aggregation, growth factors<br />

and many other compounds, which are secreted upon platelet activation. Transporters mediate accumulation of<br />

these molecules in platelet granules and –upon platelet activation- their translocation across the plasma membrane.<br />

From a more pharmacologic point of view, platelets represent pharmacokinetic micro-compartments, in which the<br />

combination of uptake and elimination transport, possibly together with intracellular metabolism determines<br />

pharmacokinetics of drugs. As a result of transporter-mediated uptake, drugs can act on the target structures inside<br />

the platelets and provoke therapeutically positive effects or negative side effects. On the other hand, elimination of<br />

drugs from the platelet by export pumps can limit the effect of the drug and lead to drug resistance.<br />

Several members of the two major transporter families, ATP-binding cassette (ABC) transporters and solute carriers<br />

(SLCs), have been identified in platelets. 1 An example of an ABC transporter is MRP4 (ABCC4), which has been<br />

shown to facilitate ADP accumulation in dense granules. 2 Several additional functions of MRP4 in platelets have<br />

been proposed, since MRP4 is a very versatile transporter and its localization can be shifted from granules to the<br />

plasma membrane upon activation of the platelets and under certain pathophysiological conditions. These functions<br />

include the release of lipid mediators as well as a role in aspirin resistance. Several other ABC proteins have been<br />

detected in platelets with functions in glutathione and lipid homeostasis including ABCC1 and ABCC3 as well as<br />

several members of the ABCA subfamily. 3 The serotonin transporter (SERT, SLC6A4) in the platelet plasma<br />

membrane represents a well characterized example of the SLC family. Furthermore, expression of the organic anion<br />

transporter OATP2B1 (SLCO2B1), a high affinity transporter for certain statins, has been demonstrated in platelets.<br />

Changes in transporter localization and expression can affect platelet function and drug sensitivity. Thus, due to their<br />

various functions, platelet transporters can either serve as drug targets or as drug delivery systems and these<br />

functions can be greatly altered in the setting of inherited or acquired platelet defects. Therefore, a detailed<br />

understanding of the expression, function and regulation of transporters in platelets will add to a better understanding<br />

of platelet biology and in many ways may result in improved therapeutic options.<br />

References:<br />

1. Jedlitschky, G. et al. Blood. <strong>2012</strong>, 119(15):3394-3402.<br />

2. Jedlitschky, G. et al. Am.J. Pathol. 2010, 176(3):1097-1103.<br />

3. Niessen, J. et al. Pharmacogenet. Genomics. 2010, 20(6):396-400.<br />

50 Sessionvorträge Targets


Expression analysis of drug transporter proteins in RPMI 2650 cell line and excised human nasal<br />

mucosa<br />

Reichl S<br />

Institut für Pharmazeutische Technologie, Technische <strong>Universität</strong> Carolo-Wilhelmina zu Braunschweig, Mendelssohnstraße 1, 38106<br />

Braunschweig, Germany<br />

The nasal mucosa is an interesting site for application of drugs with systemic action, since it has a number of<br />

advantages as a delivery route, including easy accessibility, extensive vascular supply and avoidance of<br />

gastrointestinal degradation as well as first pass metabolism. Therefore more and more applications for nasal<br />

systemic drug delivery have been developed during the last decades, and their number is still increasing [1].<br />

In the development of drugs for nasal application, identification and characterization of the different uptake and efflux<br />

systems are necessary. P-glycoprotein (P-gp) and multidrug resistance-associated proteins (MRP) are classified as<br />

ATP binding cassette (ABC) transporters based on their sequences, organisation of the ATP-binding domains and<br />

efflux function. ABC proteins represent a large family of integral membrane transporters that utilize the energy of ATP<br />

hydrolysis to carry specific substrates across membranes [2]. The solute carrier gene (SLC) superfamily encodes<br />

another large family of membrane-bound transporters, located in almost every cellular and organelle membranes.<br />

Proteins of the SLC family include passive transporters, ion-coupled transporters and exchangers [3].<br />

A comparison was performed between excised human nasal mucosa from turbinectomy surgeries and our in vitro<br />

model based on immortalized human nasal epithelial cells (RPMI 2650) concerning the mRNA expression of different<br />

efflux and uptake transporter proteins. First investigations to prove functionality of different ABC transporters in RPMI<br />

2650 cells was carried out using permeation studies and uptake assays.<br />

The mRNA expression of eight ABC transporters as well as 13 SLC transporters of excised human tissue and RPMI<br />

2650 epithelial model were investigated and compared. The results showed promising similarity in the possible<br />

presence or absence of these efflux and influx transporters in human tissue and the in vitro model. A similarity<br />

between RPMI 2650 models and human nasal mucosa has already been shown for barrier properties to evaluate<br />

passive drug permeation [4].<br />

To verify the functionality of the first transporter protein, P-gp, bidirectional transport studies using rhodamine 123<br />

with RPMI 2650 cells grown on polyethylene terephthalate filter were performed. Indirect immunofluorescence<br />

staining for P-gp was also carried out. The findings confirmed the assumption that P-gp is not functionally expressed<br />

in these epithelial cells.<br />

An uptake assay using 5(6)-carboxy-2′,7′-dichlorofluorescein as a substrate for MRP1 and MRP5 was carried out.<br />

The result indicated functional expression of these efflux transporters in RPMI 2650 cells. Identification and<br />

localization of the responsible transporter proteins have to be proved by immunohistochemistry.<br />

Further investigations will be focused on western blot, immunohistochemical analysis and permeation as well as<br />

uptake studies using specific substrates for active transport to verify the protein expression and functionality of<br />

further drug transporters in human nasal mucosa and in vitro model based on RPMI 2650 cell line.<br />

Acknowledgments:<br />

We are grateful to the German Federal Institute for Risk Assessment (BfR), which funded this work under grant no. 3-1329-469. Furthermore,<br />

the authors thank Prof. Dr. Schroeder, Dr. Schmidt, Städtisches Klinikum Braunschweig and Dr. Reintjes, HNO-Praxis Schlosscarree<br />

Braunschweig for the supply of human nasal mucosa specimens.<br />

References:<br />

1. Dimova, S. et al.: Toxicology In Vitro 2005, 19: 107–122.<br />

2. Jones, P.M., George, A.M.: Cell. Mol. Life Sci. 2004, 61: 682–699.<br />

3. He, L., Vasiliou, K., Nebert, D.W.: Human Genomics 2009, 3: 195–206.<br />

4. Wengst, A., Reichl, S.: Eur. J. Pharm. Biopharm. 2010, 74: 290–297.<br />

Sessionvorträge Targets 51


Session Nukleäre Rezeptoren und Transkriptionsfaktoren als Wirkstofftargets<br />

Investigation on the estrogen receptor alpha selectivity of 2,3,5-triaryl-1H-pyrroles<br />

Gust R<br />

Institute of Pharmacy, University of Innsbruck, 6020 Innsbruck, Austria<br />

The estrogen receptors ERα and ERβ are ligand-inducible transcription factors which belong to the nuclear hormone<br />

receptor superfamily. ER activation or inhibition influences growth, differentiation and physiological function of<br />

several organs. Selective ER modulators (SERM) like raloxifene or tamoxifen are used as partial ERα antagonists in<br />

the therapy of osteoporosis and hormone dependent ERα-positive breast cancer, respectively. Despite their obvious<br />

benefits, SERMs may cause some serious side effects, including thromboembolisms and endometrial cancer, which<br />

urgently require the search for new derivatives.<br />

In this study we wanted to design molecules that bind exclusively to ERα, which is over-expressed in hormonedependent<br />

breast cancer cells. If the compounds displayed cytotoxic activity too, we would have an adequate drug<br />

for the therapy of the ER-positive mammary carcinoma.<br />

We started our investigations from the observation that triarylpyrazoles (e.g. PPT) but not related 2,4,5triarylimidazoles<br />

showed high agonistic/antagonistic potency and ER subtype selectivity. This finding documents that<br />

the position of the N-atoms of the heterocycle in relation to the aromatic rings determines the ER binding and<br />

subtype selectivity.<br />

In continuation of this study we synthesized 1H-pyrroles whose N-position in relation to the aryl rings at the<br />

heterocycle was selected from the position in PPT resulting in Type A pyrroles or Type B pyrroles.<br />

While Type A pyrroles showed strong degradation under physiological conditions, Type A pyrroles were stable and<br />

activated the ER only in U2-OS/ERα cells. For all pyrroles no transactivation could be observed in ERß containing<br />

U2-OS cells, which documents that they are selective ERα agonists in cellular systems. Additionally, the most<br />

interesting drug 2,3,5-tris(2-fluoro-4-hydroxyphenyl)-1-propyl-1H-pyrrole inhibited the growth of hormone-dependent<br />

tumor cells comparable to the established drug 5-fluorouracil and therefore fulfills the above mentioned<br />

pharmacological profile.<br />

52 Sessionvorträge Targets


Chemical epigenetics: histone modifying enzymes as targets to regulate transcription<br />

Jung M<br />

Institute of Pharmaceutical Sciences, Albert-Ludwigs-<strong>Universität</strong>, Albertstr. 25, 79104 Freiburg,<br />

Background<br />

Epigenetics is defined as inheritable phenotypic traits that determined by mechanisms beyond the DNA sequence.<br />

Histone modifications are an important component of the epigenetic machinery and we are working on the drug<br />

discovery of epigenetic inhibitors as well as the use of small molecule substrates to test for cellular enzyme activity.<br />

Material and Methods<br />

We have used focussed library and virtual screening in collaboration with Wolfgang Sippl (University of Halle) to<br />

identify potential inhibitors of epigenetic targets. The candidates were then screened in in-vitro assays in our lab and<br />

in collaborating labs (M. Lübbert, R. Schüle, University Freiburg Medical Centre) for phenotypic and functional<br />

assays. We have used a fluorogenic substrate to screen cell culture, animal tissue samples and blood cells for<br />

histone deacetylase activity and have validate this using western blotting of acetyl-histones.<br />

Results<br />

A combination of virtual and focussed library screening has identified new inhibitors of histone deacetylases as well<br />

as the histone kinase PRK1. The PRK1 inhibitor lestaurtinib shows a suppression of androgen dependent gene<br />

transcription in prostate cancer cells1 . A carbamate has been identified as a prodrug for hydroxamic acids with<br />

increased intracellular activity2 and we currently work on testing different protecting groups and their effect on cellular<br />

permeability.<br />

We thank the Deutsche Forschungsgemeinschaft (Ju295/7-1 and Ju295/9-1, within SPP 1463) for funding.<br />

References:<br />

1. Köhler, J. et al.: PLoS ONE <strong>2012</strong>, 7: e34973.<br />

2. Schlimme, S. et al. ChemMedChem 2011, 6: 1193-8.<br />

Sessionvorträge Targets 53


Helenalin derivatives and their impact on transcriptional control of mediators involved in rheumatic<br />

diseases<br />

Jäger C1 , Patel H1 , Humar M2 , Günther S3 , Merfort I1 1 Depart. Pharmaceutical Biology and Biotechnology, Inst. Pharmaceutical Sciences, Albert-Ludwigs University, Stefan-Meierstr. 19, 79104<br />

Freiburg, Germany<br />

2 Depart. Anesthesiology and Critical Care Medicine, University Hospital Freiburg, Freiburg, Germany<br />

3 Inst. Pharmaceutical Scienecs, Pharmaceutical Bioinformatics, Albert-Ludwigs University, Hermann-Herderstr. 9, 79104 Freiburg, Germany<br />

Helenalin derivatives are sesquiterpene lactones from the pseudoguaianolide type. They are considered as the<br />

effective compounds in Arnica montana flowers which preparations have been proven to exert beneficial effects in<br />

rheumatic complaints, such as osteoarthritis and rheumatoid arthritis (1). We here report studies on the molecular<br />

mode of actions which contribute to a better understanding of these beneficial effects.<br />

Both rheumatic diseases are characterized by chronic joint destruction. Whereas IL-1β and TNF-α are well-known<br />

players in this process it has become more and more obvious that IL-17 is also involved (2). This cytokine stimulates<br />

fibroblasts, macrophages and chondrocytes which for their part release various mediators with effects on<br />

inflammation, cartilage damage and bone erosion. Interestingly, IL-17 does not only induce TNF-α and IL-1β, but<br />

was shown to have also additive or even synergistic effects with TNF-α and IL-1β during the induction of cytokine<br />

expression and joint damage in vitro and in vivo.<br />

We could show with helenalinbutyrate that mRNA levels of IL-17 are reduced in primary human T-cells.<br />

Consequently, pretreatment of human primary chondrocytes with helenalinisobutyrate or<br />

dihydrohelenalinmethacrylate drastically reduced gene and protein expression of matrix metalloproteinase 1 (MMP1)<br />

and 13 (MMP13) at low concentrations with helenalinbutyrate being mostly active. Additionally, this sesquiterpene<br />

lactone lowered synergistically upregulated mRNA and protein of COX-2 by IL-17 and IL-1β. Transcription of MMPs<br />

is tightly regulated by the transcription factors AP-1 and NF-κB which were demonstrated to be directly targeted by<br />

the helenalin derivatives. The effect on NF-κB has already been studied intensively (3) in contrast that one on AP-1.<br />

Impacts on these transcription factors were supported by molecular docking studies.<br />

Acknowledgments: We are grateful to Bioforce company for financial support.<br />

References:<br />

1. Merfort, I. Curr. Drug Targets. 2011, 12: 1560-73.<br />

2. Koenders, M.I., Joosten, L.A.B., van den Berg, W.B., Ann. Rheum. Dis. 2006, 65: 29-33.<br />

3. Garcia-Pineres, A. et al., J. Biol. Chem. 2001, 276: 391113-20.<br />

54 Sessionvorträge Targets


Rescuing mutant p53 using halogen-enriched fragment libraries (HEFLibs)<br />

Boeckler F M1 ; Wilcken R1,2 ; Fersht A R2 ; Joerger A C2 1 Laboratory for Molecular Design and Pharmaceutical Biophysics, Department of Pharmaceutical and Medicinal Chemistry, Institute of<br />

Pharmacy, Eberhard-Karls-University Tuebingen, Auf der Morgenstelle 8, 72076 Tuebingen, Germany<br />

2 MRC Laboratory of Molecular Biology, Hills Road, Cambridge CB2 0QH, United Kingdom<br />

The destabilizing cancer mutation Y220C creates a druggable surface crevice in the DNA-binding core domain of the<br />

transcription factor and tumor suppressor p53 [1,2]. We developed a strategy exploiting halogen bonding for lead<br />

discovery to stabilize the mutant with small molecules. We designed halogen-enriched fragment libraries (HEFLibs)<br />

as starting points to complement classical approaches [3]. From screening of HEFLibs and subsequent structureguided<br />

design, we developed substituted 2-(aminomethyl)-4-ethynyl-6-iodophenols as p53-Y220C stabilizers. Crystal<br />

structures of their complexes highlight two key features: (i) a central scaffold with a robust binding mode anchored by<br />

halogen bonding of an iodine with a main chain carbonyl and (ii) an acetylene linker, enabling the targeting of an<br />

additional subsite in the crevice. The best binders showed induction of apoptosis in a human cancer cell line with<br />

homozygous Y220C mutation. Structure-activity relationships show correlations to QM-based interaction energies of<br />

halogen-bonding. Our structural and biophysical data suggest a more widespread applicability of HEFLibs in lead<br />

identification, yielding lead structures that feature binding modes hardly obtainable by other techniques.<br />

References:<br />

1. Joerger, A. C.; Ang, H. C.; Fersht, A. R.: Proc. Natl. Acad. Sci. U. S. A. 2006, 103(41): 15056-61.<br />

2. Boeckler, F. M. et al.: Proc. Natl. Acad. Sci. U. S. A. 2008, 105(30): 10360-10365.<br />

3. Wilcken, R. et al.: J. Am. Chem. Soc. <strong>2012</strong>, 134(15): 6810–6818.<br />

Sessionvorträge Targets 55


Novel leads for PPARs and FXR: a computer-aided drug design approach<br />

Proschak E1 , Steri R, Achenbach J, Lamers C, Rau O, Schneider G, Steinhilber D, Schubert-Zsilavecz M1 Goethe-University of Frankfurt, Institute of Pharmaceutical Chemistry, OSF/ZAFES, Max-von-Laue Str. 9, D-60438 Frankfurt, Germany<br />

Peroxisome proliferator-activated receptors (PPARs) and farnesoid X receptor are in the main focus of<br />

pharmaceutical research for a wide range of metabolic disorders. Novel findings in this area indicate that although<br />

these targets have been investigated intensively, there is still an unmet need for novel safe and efficient lead<br />

structures. This presentation demonstrates the impact of computer-aided drug design techniques applied to<br />

discovery and optimization of novel lead structures for PPARs and FXR. We present three different approaches to<br />

identify novel lead structures: virtual screening, de novo design and in silico drug repurposing.<br />

In our virtual screening study we were able to identify a potent and selective PPARa agonist using a combination of<br />

pharmacophore- and shape-based screening [1]. The software retrieved a potent hit (EC50 = 44 nM), with >100fold<br />

selectivity against PPARγ. In a second application, we designed a lead structure de novo, starting from the known<br />

PPARα agonist GW590735 [2]. In the design process, two moieties were replaced by bioisosteric groups suggested<br />

by SQUIRRELnovo, which are responsible for compound potency. The new lead structure yielded submicromolar<br />

activity on PPARα (EC50 = 0.51 µM) in a cell-based reporter-gene assay [3].<br />

Furthermore we were able to identify the molecular basis for the glucose-lowering effects of imatinib based on FXR<br />

activation using a machine-learning approach [4]. We applied self-organizing maps to self-organizing map (SOM) is a<br />

virtual screening method for correlation of molecular structure and potential biological activity [5]. We used a SOM<br />

trained with known FXR ligands to screen the DrugBank database [6] for potential ligands for FXR. We were able to<br />

identify six approved drugs out of the Drugbank as FXR modulators (ketoconazole, pentamidine, dobutamine,<br />

imatinib, papaverine and montelukast) by using a SOM for screening of the DrugBank database. We show FXR<br />

modulation by selected compounds in a full length FXR transactivation assay and modulation of a FXR target gene<br />

by imatinib.<br />

Acknowledgments: We gratefully acknowledge financial support from the Else-Kröner-Fresenius-Stiftung, Merz Pharmaceuticals, Lipid<br />

Signaling Forschungszentrum Frankfurt (LiFF), Oncogenic Signaling Frankfurt (OSF) and Fonds der Chemischen Industrie for financial<br />

support.<br />

References:<br />

1. Proschak, E., Zettl, H., Tanrikulu, Y., Weisel, M., Kriegl, J.M., Rau, O., Schubert-Zsilavecz, M., Schneider, G., ChemMedChem 2009, 4:<br />

41–44.<br />

2. Sierra ML, Beneton V, Boullay A-B, Boyer T, Brewster AG, Donche F, et al., J Med Chem 2007, 50: 685–95.<br />

3. Proschak, E., Sander, K., Zettl, H., Tanrikulu, Y., Rau, O., Schneider, P., Schubert-Zsilavecz, M., Stark, H., Schneider, G., ChemMedChem<br />

2009, 4: 45-48.<br />

4. Steri, R., Achenbach, J., Steinhilber, D., Schubert-Zsilavecz, M., Proschak, E., Biochem Pharmacol <strong>2012</strong> accepted.<br />

5. Schneider P, Tanrikulu Y, Schneider G., Curr Med Chem 2009, 16: 258–66.<br />

6. Knox C, Law V, Jewison T, Liu P, Ly S, Frolkis A, et. Al., Nucleic Acids Res 2011, 39: D1035–1041.<br />

56 Sessionvorträge Targets


Inhibitors of class I histone deacetylases induce 5-lipoxygenase expression<br />

Steinhilber D<br />

Goethe-University Frankfurt, Institute of Pharmaceutical Chemistry/ZAFES, Max-von-Laue Str. 9, D-60438 Frankfurt, Germany<br />

5-lipoxygenase (5-LO) is the key enzyme in the formation of leukotrienes. We have previously shown that the histone<br />

deacetylase (HDAC) inhibitor trichostatin A (TSA) activates 5-LO transcription via recruitment of Sp1, Sp3 and RNA<br />

polymerase II to the proximal promoter [1, 2].<br />

In order to identify the HDACs involved in the regulation of 5-LO promoter activity isoform-specific HDAC inhibitors<br />

were applied. 5-LO promoter activity and mRNA expression was upregulated by the class I HDAC inhibitors apicidin<br />

and MS-275 but not by class II inhibitors.<br />

In order to analyze the chromatin modifications at the 5-LO promoter associated with HDAC inhibition, the time<br />

course of 5-LO mRNA induction by trichostatin A was analyzed and the concomitant changes in histone<br />

modifications at the 5-LO promoter in HL-60, U937 and Mono Mac6 cells was determined. ChIP analysis revealed<br />

that trichostatin A increases acetylation of histones H3 and H4 at the 5-LO core promoter in HL-60 and U937 cells<br />

whereas no significant changes were observed in Mono Mac6 cells. The appearance of H3 and H4 acetylation<br />

preceded the 5-LO mRNA induction whereas in all three cell lines, induction of 5-LO mRNA expression correlated<br />

with histone H3 lysine 4 trimethylation (H3K4me3), a marker for transcriptional activity of gene promoters.<br />

Induction of 5-LO mRNA expression by class I HDAC inhibitors coincides with the upregulation of 5-LO promoter<br />

activity as determined by reporter gene assays. The data suggest that in contrast to calcitriol and TGFβ [3], induction<br />

of 5-LO mRNA expression by HDAC inhibition mainly occurs on the 5-LO promoter level.<br />

Acknowledgments: We gratefully acknowledge financial support from the Else Kröner-Fresenius-Stiftung and DFG (GRK757).<br />

References:<br />

1. Schnur, N., Seuter, S., Katryniok, C., Rådmark, O. and Steinhilber, D., Biochim Biophys Acta 2007, 1771: 1271-1282.<br />

2. Klan, N., Seuter, S., Schnur, N., Jung, M. and Steinhilber, D., Biol Chem 2003, 384: 777-785.<br />

3. Stoffers, K.L., Sorg, B.L., Seuter, S., Rau, O., Radmark, O. and Steinhilber, D., J Mol Biol 2010, 395: 884-896.<br />

Sessionvorträge Targets 57


Session Thrombozytenfunktion und Gerinnung<br />

Novel oral anticoagulants – State of the art and aspects of their application<br />

Alban S<br />

Institute of Pharmacy, Christian-Albrechts-University of Kiel, Gutenbergstr. 76, 24118 Kiel, Germany<br />

Since thromboembolic diseases, i.e. myocardial infarction, stroke and pulmonary embolism (PE), represent the major<br />

cause of death in the Western world, anticoagulants are a class of life-saving drugs. Whereas there have been some<br />

advances in short- and medium-term anticoagulation during the last two decades, the only option for long-term<br />

anticoagulation were the vitamin K antagonists (VKA) so far.<br />

This has changed in 2008 with the approval of two novel oral anticoagulants (NOA): the direct thrombin inhibitor<br />

(DTI) dabigatran etexilate (Pradaxa ® ) und the first direct Factor Xa-Inhibitor (DXI) rivaroxaban (Xarelto ® ) for<br />

prevention of venous thromboembolism (VTE) after hip and knee replacement operation. In 2011, apixaban<br />

(Eliquis ® ), another oral DXI, was approved for this proof of concept indication and dabigatran etexilate and<br />

rivaroxaban were licenced for stroke prevention in atrial fibrillation and rivaroxaban additionally for treatment of deep<br />

vein thrombosis (DVT) and prevention of recurrent DVT and PE following acute DVT.<br />

Therefore, it’s time for an update: How the anticoagulation in clinical practice changes with the NOA? What are the<br />

differences between the NOA and the VKA as well as between the three NOA? Which patients are candidates for<br />

switching from VKA to NOA? What is about practical aspects like monitoring anticoagulant efficacy, interruption for<br />

surgical or invasive procedures and management of bleeding? Finally, are Marcumar ® and heparin injections about<br />

to fold?<br />

58 Sessionvorträge Targets


Novel platelet inhibitors – State of the art and aspects of their application<br />

Greinacher A<br />

Sessionvorträge Targets 59


Naturally occurring PF4/polyanion complexes prime for heparin-induced thrombocytopenia as a<br />

misdirected host defense mechanism<br />

Krauel K1,2 ; Weber C3 ; Jaax M1 ; Hackbarth C 1 ; Brandt S2 ; Block S2 ; Hammerschmidt S3 ; Greinacher A1 1 Institut für Immunologie und Transfusionsmedizin<br />

2 Zentrum für Innovationskompetenz – Humorale Immunreaktionen bei kardiovaskulären Erkrankungen<br />

3 Abteilung Genetik der Mikroorganismen, Interfakultäres Institut für Genetik und Funktionelle Genomforschung, <strong>Ernst</strong>-<strong>Moritz</strong>-<strong>Arndt</strong>-<strong>Universität</strong><br />

<strong>Greifswald</strong>, Germany<br />

Heparin-induced thrombocytopenia (HIT) is caused by highly immunogenic complexes of negatively charged heparin<br />

and positively charged platelet factor 4 (PF4). Some of the resulting antibodies against PF4/heparin complexes<br />

activate platelets via the platelet FcγIIa receptor, leading to thrombin generation and a prothrombotic state. We have<br />

recently proposed that HIT might be the result of a misdirected bacterial defense mechanism, as PF4 bound to<br />

bacteria also exposes the epitopes to which PF4/heparin antibodies bind [1]. Consistently, we showed that<br />

PF4/heparin antibodies are highly significantly associated with periodontitis, one of the most prevalent human<br />

infections mostly induced by Gram-negative bacteria [2].<br />

We now identified potential PF4 binding structures on the surface of Gram-negative bacteria using different bacterial<br />

mutants with defined variations in their lipopolysaccharide (LPS) sequences [3]. Binding of biotinylated PF4 was<br />

quantified by flow cytometry, and exposure of the antigenic epitope was assessed by adsorption and elution of<br />

PF4/heparin antibodies. Bacterial mutants lacking the O-antigens and the core of LPS but still exposing lipid A<br />

showed strongest PF4 binding, mediated by the phosphate groups of lipid A. As both the lipid A on the surface of<br />

Gram-negative bacteria and the amino acids of PF4 contributing to polyanion binding are highly conserved, our<br />

results further support the hypothesis that neoepitope formation on PF4 after binding to bacteria is an ancient host<br />

defense mechanism. The distance of two phosphate groups on lipid A is rather similar to the distance of phosphate<br />

groups on DNA or RNA. We therefore assessed the interaction of PF4 and nucleic acids. By enzyme-immunoassay<br />

and functional studies, we found cross-reactivity of PF4/heparin antibodies with PF4/DNA complexes in vitro.<br />

Systematic assessment of different RNA- and DNA-constructs revealed length and number of double-stranded<br />

segments of nucleic acids as risk factors for complex formation with PF4. This has consequences for drug<br />

development, as we accordingly found immunogenicity as a class effect of aptamers (DNA/RNA based therapeutics).<br />

PF4/aptamer complexes bound PF4/heparin antibodies in vitro and induced PF4/aptamer antibodies in a mouse<br />

model.<br />

We next asked the question, which structural changes of PF4 may trigger B-cell activation using circular dichroism<br />

(CD) spectroscopy. Pronounced structural changes of PF4, leading to expression of anti-parallel beta sheets,<br />

occurred during the interaction with highly charged polyanions known to be immunogenic in vivo. Only minor or no<br />

changes were found for weakly charged or neutral polymers. This allows to predict immunogenicity of drugs exposing<br />

polyanions by CD spectroscopy.<br />

Through desulfation of heparin, the new compound 2-O, 3-O desulfated heparin (ODSH, ParinGenix) has low<br />

anticoagulant activity but the residual negative charge still allows interaction with PF4. We found that ODSH inhibits<br />

platelet activation by PF4/heparin antibodies by disruption of PF4/heparin complexes [4]. In addition, ODSH does not<br />

induce the conformational changes of PF4 analyzed by CD spectroscopy. Therefore a mixture of heparin and ODSH<br />

might be an interesting option to reduce the risk of HIT which has to be addressed in clinical studies.<br />

In summary, polyanions induce structural changes of PF4 leading to an adverse immune reaction, which is a<br />

misdirected humoral host defense mechanism. CD spectroscopy can be applied to screen new drugs for their<br />

potential to induce immunogenic structural changes of PF4 and partially desulfated heparin can be applied to<br />

antagonize the adverse biological effects of PF4/polyanion complex antibodies.<br />

References:<br />

1. Krauel, K. et al.: Blood 2011, 117(4): 1370-1378.<br />

2. Greinacher, A. et al.: Blood 2011, 118(5): 1395-1401.<br />

3. Krauel, K. et al.: Blood <strong>2012</strong>, epub.<br />

4. Krauel, K. et al.: Blood <strong>2012</strong>, 119(5): 1248-1255.<br />

60 Sessionvorträge Targets


Session Neue Targets und Testsysteme<br />

PfGSK-3: A target for new antimalarial drugs?<br />

Kunick C1 , Fugel W1 , Ratin M2 , Kruggel S3 , Oberholzer A E4 , Dzikowski R5 , Pressburger N5 , Ferandin Y2 , Lemcke T3 ,<br />

Meijer L2 1 Technische <strong>Universität</strong> Braunschweig, Institut für Medizinische und Pharmazeutische Chemie, Beethovenstraße 55, 38106 Braunschweig,<br />

Germany<br />

2 Protein Phosphorylation & Human Disease Group, CNRS, Station Biologique, Place Georges Teissier, B.P. 74, 29682 Roscoff, France<br />

3 <strong>Universität</strong> Hamburg, Institut für Pharmazie, Bundesstraße 45, 20146 Hamburg, Germany<br />

4 Structural Biology Community Laenggasse (sbcl), 3000 Bern, Switzerland<br />

5 Department of Microbiology & Molecular Genetics, The Kuvin Center for the Study of Infectious and Tropical Diseases, IMRIC, The Hebrew<br />

University-Hadassah Medical School, Jerusalem 91120, Israel<br />

Malaria is one of the most threatening infectious diseases in tropical areas, causing hundreds of thousands fatal<br />

cases each year. [1] Although efficacious medicines are available for treatment and prophylaxis, the emergence of<br />

resistance is urging for new drugs with alternative pharmacological mechanisms. The inhibition of plasmodial protein<br />

kinases has repeatedly been suggested as a strategy for the development of antimalarial drugs throughout the last<br />

decade. [2-4] In spite of several efforts in this direction, protein kinase inhibitors have not yet reached clinical trials as<br />

antimalarial drugs. PfGSK-3, the plasmodial homologue of the human glycogen synthase kinase-3 (HsGSK-3), was<br />

recently identified from Plasmodium falciparum, the causative agent of malaria tropica. With a view to evaluate<br />

PfGSK-3 as potential target for new antimalarial agents, several known GSK-3 inhibitors were tested on PfGSK-3. [5]<br />

Unfortunately, these compounds were either none-selective or showed selective inhibition of the human enzyme.<br />

Therefore, compound libraries were tested to identify new selective PfGSK-3 inhibitors. A hit compound from this<br />

campaign was employed as structural template for the design of a congener series. Analysis of structure-activity<br />

relationships and structure-guided design enabled the preparation of selective PfGSK-3 inhibitors with in vitro<br />

antiplasmodial activity in the single-digit micromolar concentration range.<br />

Acknowledgments: The work was funded in part by the Commission of the European Communities (Contracts LSHB-CT-2004-503467 and<br />

Health-F3-2008-223414, to L. M. and C. K.). This joint project was financially supported by the State of Lower-Saxony, Hannover, Germany (to<br />

R. D., N. P., and C. K). S. K. was supported by a PhD grant from the University of Hamburg, Germany.<br />

References:<br />

1. WHO: World Malaria Report 2010 (http://www.who.int/malaria/world_malaria_report_2010/worldmalariareport2010.pdf).<br />

2. Doerig, C., Meijer, L.: Expert Opin Ther Targets 2007, 11(3): 279–290.<br />

3. Zhang, V. M., Chavchich, M., Waters, N. C.: Curr Top Med Chem <strong>2012</strong> 12(5): 456–472.<br />

4. Jirage, D., Keenan, S. M., Waters, N. C.: Infect Disord Drug Targets 2010, 10(3): 134–146.<br />

5. Droucheau, E. at al.: Biochim Biophys Acta 2004, 1697: 181–196.<br />

Sessionvorträge Targets 61


Chemical Oncology – Converging Cancer Genetics, Structural Biology and Medicinal Chemistry<br />

Rauh D<br />

Technische <strong>Universität</strong> Dortmund, Fakultät Chemie, Chemische Biologie, Otto-Hahn-Str. 6, 44227 Dortmund<br />

The 512 protein kinases encoded by the human genome are a prime example of nature's ability to create diversity by<br />

introducing variations to a highly conserved theme. Layers of regulatory mechanisms involving different combinations<br />

of post-translational modifications, intramolecular contacts, and intermolecular interactions control the activity of each<br />

kinase domain. Ultimately, they all achieve their effect by favoring particular conformations that promote or prevent<br />

the kinase domain from catalyzing protein phosphorylation. The central role of kinases in various diseases has<br />

encouraged extensive investigations of their biological function and three-dimensional structures, yielding a more<br />

detailed understanding of the mechanisms that regulate protein kinase activity by conformational changes.[1] I will<br />

discuss these regulatory mechanisms and show how conformational changes can be exploited for the design of<br />

specific inhibitors that lock protein kinases in inactive conformations.[2, 3] In addition, I highlight our recent<br />

developments to monitor ligand-induced structural changes in protein kinases and for screening and identifying<br />

inhibitors that stabilize enzymatically incompetent kinase conformations.[4-8]<br />

References:<br />

1. Rabiller, M., et al., Arch Pharm 2010, 343(4): 193-206.<br />

2. Getlik, M., et al., J Medicinal Chem 2009, 52(13): 3915-26.<br />

3. Getlik, M., et al., Eur J Med Chem <strong>2012</strong>, 48: 1-15.<br />

4. Simard, J.R., et al., Nature Chem Biol 2009, 5(6): 394-6.<br />

5. Simard, J.R., et al., J Am Chem Soc 2009, 131(51): 18478-88.<br />

6. Simard, J.R., et al., J Am Chem Soc 2010, 132(12): 4152-60.<br />

7. Simard, J.R., et al., J Am Chem Soc 2009, 131(37): 13286-96.<br />

8. Schneider, R., et al., J Am Chem Soc <strong>2012</strong>, 134(22): 9138-41.<br />

62 Sessionvorträge Targets


Development of lysine mimic quinazoline-based selective inhibitors of a Jmj-C histone demethylase<br />

family<br />

Rotili D1 ; Cheng X2 ; Mai A1 1 Pasteur Institute–Cenci Bolognetti Foundation, Department of Chemistry and Technology of Drugs, “Sapienza” University of Rome, Aldo Moro<br />

Square 5, 00185 Rome, Italy<br />

2 Department of Biochemistry, Emory University School of Medicine, 1510 Clifton Road, Atlanta, GA 30322, USA<br />

The histone proteins are subjected to various post-translational modifications, such as methylation, acetylation,<br />

phosphorylation, and ubiquitylation. These modifications, interfering with RNA polymerase activity and DNA<br />

transcription, represent one of the main mechanisms of the epigenetic modulation of gene expression. Aberrant<br />

expression of histone-modifying enzymes has been linked to various human diseases, such as neurological<br />

disorders and cancer. While studies on histone acetylation are known from years, studies on histone<br />

methylation/demethylation and relative modulators are at their infancy [1].<br />

N<br />

N<br />

HN<br />

HN<br />

MeO<br />

N<br />

MeO<br />

N<br />

MeO N N<br />

N<br />

H2N O N NH<br />

N<br />

BIX-01294 MC3076<br />

N<br />

H2N MeO<br />

O<br />

HN<br />

N<br />

N Cl<br />

H2N<br />

MeO<br />

O<br />

N<br />

N<br />

N NH<br />

N<br />

MC3073 MC3074<br />

BIX-01294, a diazepin-quinazoline-amine derivative able to inhibit G9a and G9a-like protein (GLP) lysine<br />

methyltransferases and to reduce methylation levels of H3K9 at several G9a target genes [2], was our starting point<br />

to design novel putative inhibitors of histone methyltransferases and/or demethylases acting on H3K9, (G9a/GLP and<br />

KIAA1718, respectively). Indeed, G9a and KIAA catalyze opposite reactions on H3K9 (the first adding and the latter<br />

removing the Me2 histone mark), and may recognize Lys in either un-methylated or methylated state, it being the<br />

substrate or reaction product, alternatively. Thus, we replaced the C7-methoxy group of BIX with a Lys-mimic side<br />

chain (5-aminopentyloxy chain), obtaining increased G9a/GLP inhibition (MC3076) respect to BIX [3]. Since MC3076<br />

as well as BIX were found by us able to inhibit also the demethylase KIAA1718, on the basis of crystallographic<br />

studies performed on MC3076 complexed with either GLP or KIAA, we prepared a new series of quinazoline analogs<br />

in order to improve their potency and selectivity against KIAA demethylase.<br />

We proceeded to the simplification of the MC3076 structure by insertion of small substituents at the C2 and/or C4<br />

quinazoline ring positions, so obtaining new quinazolines (MC3073, MC3074, etc.) highly potent against KIAA1718<br />

and devoid of any GLP inhibiting activity [4]. Some of these derivatives (MC3074) were also inactive against the<br />

JARID family of Jmj-C histone demethylases so demonstrating a promising capability to discriminate between<br />

different Jmj-C histone demethylase families [4].<br />

References:<br />

1. Varier, R.A.; Timmers, H.T.M. Biochim Biophys Acta 2011, 1815(1): 75-89.<br />

2. Kubicek, S. et al.: Mol Cell 2007, 25(3): 473-481.<br />

3. Chang, Y. et al.: J Mol Biol 2010, 400(1): 1-7.<br />

4. Upadhyay, A.K. et al.: J Mol Biol <strong>2012</strong>, 416(3): 319-327.<br />

Sessionvorträge Targets 63


Profiling of epigenetic targets using peptide microarrays<br />

Schutkowski M; Rauh D; Roessler C; Masch A<br />

Martin-Luther-University Halle-Wittenberg, Institute of Biochemistry & Biotechnology, Department Enzymology, Kurt-Mothes-Strasse 3, 06120<br />

Halle, Germany<br />

Acetylation of lysine residues in proteins is one of the most frequently occurring post-translational modifications<br />

playing a major role in protein-protein-interaction as well as in regulating DNA transcription and metabolic processes.<br />

Using acetyl coenzyme A as donor for the acetyl group, lysine acetyltransferases are transferring the acetyl group<br />

onto the ε-amino-group of lysine residues in proteins. The positive charge of the side chains is neutralized, thus<br />

remodeling biological activity of enzymes and protein-protein-interaction. Bromodomains as protein interaction<br />

modules are recognizing specifically the acetylated lysine side chains.<br />

To understand substrate specificity of lysine acetyltransferases and binding specificity of bromodomains, we created<br />

a high-density peptide microarray displaying 6800 peptides derived from all known acetylation sites in human<br />

proteins in both unmodified and acetylated form resulting in more than 13.600 different features per microarray pair<br />

(acetylome microarray). The acetylome microarrays were treated with lysine acetyltransferases in the presence of<br />

acetyl-CoA followed by an optimized mixture of anti-acetyl-lysine-antibodies.<br />

Additionally, treatment of the acetylome microarray with Sirtuins, NAD + -dependent protein lysine-deacetylases<br />

involved in regulation of central physiological functions, such as energy metabolism, cell cycle progression, and<br />

aging processes, resulted in decrease of signals for several of the acetylated peptides in comparism to the control<br />

experiment in the absence of NAD + . We were able to identify subsite specificities and substrate sequences for all of<br />

the seven human Sirtuin isoforms. Besides known substrates our results provide novel substrate candidates for the<br />

different Sirtuin isoforms which could be confirmed in subsequent solution phase experiments.<br />

Principally, each enzyme transfering residues to the side chains of lysines could be analysed with the acetylome<br />

microarray using either radioisotopic labeling (lysine-methyltransferases and labelled AdoMet, poly-ADPribosyltransferases<br />

and labelled NAD + ) or antibody-based readout (poly-ADP-ribosyltransferases and ethenoNAD +<br />

followed by anti-ethenoNAD + -antibody, ubiquitin/SUMO transfering enzymes followed by anti-ubiquitin/SUMOantibodies).<br />

64 Sessionvorträge Targets


Session Tumortherapie und Resistenzmechanismen<br />

Anti-infectives research to combat drug resistant tumors?<br />

Hartmann R W 1,2<br />

1 Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), Campus C2.3, 66123 Saarbrücken, Germany<br />

2 Pharmaceutical and Medicinal Chemistry, Saarland University, Campus C2.3, 66123 Saarbrücken, Germany<br />

In the last decades there have been great efforts to develop new tumor therapeutics and in the last 25 years<br />

approximately 100 new drugs (NCEs) have been admitted to the market. The clinical situation however has only<br />

improved slightly which is due to the fact that the tumors develop resistance against the new drugs as well. The big<br />

amounts of money that have been invested into basic research to elucidate resistance mechanisms have brought<br />

new interesting findings but (so far) have not resulted in more efficient drugs. Also in antibiotics therapy the<br />

emergence of resistant bacterial strains is a severe problem and research is focused on strategies to investigate how<br />

the development of resistant bacterial strains can be avoided. In this context recent results from our group will be<br />

presented aiming at a novel therapy for Pseudomonas aeruginosa infections by interference with its unique PQS<br />

quorum sensing system. Finally the question will be raised whether insights from antiinfectives research could help to<br />

discover novel strategies to combat drug resistant tumors.<br />

A new class of glutathione peroxidase inhibitors is able to reverse resistance to chemotherapeutics<br />

in human B-cell lymphoma cell lines<br />

Schulz R1 ; Emmrich T1 ; Lemmerhirt H1 ; Hirt C2 ; Kiefer T2 ; Link A1 ; Bednarski P J1 1 Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy, <strong>Ernst</strong>-<strong>Moritz</strong>-<strong>Arndt</strong> University <strong>Greifswald</strong>, 17489 <strong>Greifswald</strong>,<br />

Germany<br />

2 Clinic for Haematology and Oncology, <strong>Ernst</strong>-<strong>Moritz</strong>-<strong>Arndt</strong> University <strong>Greifswald</strong>, 17475 <strong>Greifswald</strong>, Germany<br />

The development of chemoresistance in cancer cells poses a major risk for chemotherapy failure. A combination of<br />

anticancer drugs and agents that suppresses adaptation in cancer cells is a potent method to increase the efficiency<br />

of therapies. GUMBUS and DOGUM, two cell lines from patients with non-Hodgkin lymphomas, were used to<br />

investigate the process of drug resistance. Cells were treated with the anticancer drugs methotrexate, etopiside,<br />

cisplatin and bortezomib to induce resistance. Assays showed a significantly increased amount of glutathione<br />

peroxidase 1 (GPx1) in treated cells compared to untreated. The approach now is to inhibit GPx1 to target the antioxidative<br />

pathway in these cancer cells. A virtual screening of a self-composed compound library docked to the<br />

active site of the X-ray crystal structure of bovine GPx1 yielded promising structures. In an enzyme assay a class of<br />

acylhydrazone heterocyclic proved to be GPx inhibitors. Furthermore, lymphoma cells lost their resistance to anticancer<br />

drugs when treated with these hydrazones. This concept could be refined to a new strategy in<br />

chemotherapies.<br />

Sessionvorträge Targets 65


Chemoresistance of ovarian cancers against platinum complexes<br />

Kassack M U; Hamacher A<br />

Heinrich-Heine-<strong>Universität</strong> Düsseldorf, Institut für Pharmazeutische und Medizinische Chemie, Pharmazeutische Biochemie,<br />

<strong>Universität</strong>sstrasse 1, 40225 Düsseldorf, Germany. matthias.kassack@uni-duesseldorf.de<br />

Ovarian cancer is the most lethal gynaecological cancer. Surgery and a combination chemotherapy including<br />

carboplatin (cisplatin) are the primary treatment options. Initially, most ovarian cancers are chemosensitive. However<br />

rather soon, resistance mechanisms develop and lead to relapse and therapeutic failure. Many resistance<br />

mechanisms against platinum compounds are known but this has not yet been translated into improved therapy. One<br />

reason is a lack of systematic studies to overcome chemoresistance using resistance mechanisms as additional<br />

therapeutic targets and second, relevant mechanisms driving the development of chemoresistance have only partly<br />

been studied. Comprehensive analysis of cancer cells on a genomic, transcriptional and translational level show that<br />

the development of chemoresistance is a complex biological process involving both, selection and adaptation<br />

processes (1, 2).<br />

cisplatin resistance factor<br />

7<br />

6<br />

5<br />

4<br />

3<br />

2<br />

1<br />

0<br />

1 2 3 4 5 6 7 8 9 10 12 16 20 24<br />

No of treatment cycles [weeks]<br />

Resistance development over 6 months in A2780 ovarian cancer cells<br />

To study the development of chemoresistance, we<br />

have undertaken a novel approach of generating<br />

resistant cell lines. Ovarian cancer cell lines were<br />

treated intermittently according to a clinically<br />

relevant dosing scheme over a period of 6<br />

months. By that procedure, we have obtained<br />

pairs of sensitive and resistant cell lines that were<br />

analysed using MTT assay, whole genome<br />

expression microarrays, and (phospho) proteome<br />

profiling of kinases. Bioinformatic analysis<br />

revealed a number of potential novel targets to<br />

overcome chemoresistance. Subsequent inhibitor<br />

studies in combination with carbo/cisplatin could<br />

demonstrate a reversal of chemoresistance<br />

against platinum compounds.<br />

In conclusion, bioinformatic analysis allows establishing drug resistance signatures with potential prognostic value.<br />

Furthermore, novel therapeutic targets have emerged establishing a new approach to overcome chemoresistance<br />

against carbo/cisplatin in ovarian cancer.<br />

References:<br />

1. Eckstein N, et al. Cancer Res. 2009; 69(7): 2996-3003.<br />

2. Gosepath EM, et al. Int J Cancer. 2008; 123(9): 2013-9<br />

66 Sessionvorträge Targets


Medicinal Chemistry with Organometallics – From Catalysts to Anticancer Drugs?<br />

Ott I<br />

Institute of Medicinal and Pharmaceutical Chemistry, Technische <strong>Universität</strong> Braunschweig, Beethovenstr. 55, 38106 Braunschweig, Germany<br />

Despite the tremendous success of platinum based cancer chemotherapeutics, transition metal based drugs are rare<br />

on the current drug market as well as in medicinal chemistry and drug design. However, metal complexes offer<br />

interesting opportunities for drug development based on their structural and kinetic properties, which differ in many<br />

ways from traditional “organic” compounds.[1]<br />

Organometallics are metal complexes with metal-carbon bonds, which have been attracting increasing attention in<br />

general chemistry over the last decades and enabled an enormous progress in the field of catalysis. Not surprisingly<br />

this has led to a broader knowledge of the properties of available organometallic functional groups and thereby also<br />

new approaches for inorganic medicinal chemistry have emerged.[2,3]<br />

We have been working over the last years on several types of organometallics including metal complexes with Nheterocyclic<br />

carbene (NHC) ligands or gold alkynyl complexes as new antitumor drugs (see the figure for some<br />

examples).[4-6] For many of these compounds we could confirm distinct biological properties such as antiproliferative<br />

effects in cultured tumor cells, selective inhibition of the enzyme thioredoxin reductase, triggering of reactive oxygen<br />

species (ROS) formation or anti-angiogenic effects. Binding to albumin, cellular uptake and intracellular distribution<br />

have been studied for selected examples by high resolution atomic absorption spectroscopy (HRCS-AAS) in order to<br />

provide a more detailed picture of their behaviour on the cellular level.<br />

In the presentation a short overview of the topic will be given and selected examples of our current projects will be<br />

presented.<br />

N<br />

N<br />

Au<br />

Cl<br />

O<br />

Au P<br />

Figure: examples for bioactive organometallics<br />

Sessionvorträge Targets 67<br />

N<br />

N<br />

Ru<br />

Cl Cl<br />

Acknowledgments: Financial support by the DFG (project FOR 630, “Biological Function of Organometallic Compounds”) is gratefully<br />

acknowledged.<br />

References:<br />

1. Alessio, E: Bioinorganic Medicinal Chemistry (Wiley) 2011.<br />

2. Gasser G, Ott I, Metzler-Nolte N: J Med Chem 2011, 54(1): 3–25.<br />

3. Noffke, AL et al.: Chem Comm <strong>2012</strong>, 48: 5219–5246.<br />

4. Rubbiani, R et al.: J Med Chem 2011, 54(24): 8646–8657.<br />

5. Oehninger, L et al.: ChemMedChem 2011, 6: 2142–2145.<br />

6. Meyer, A et al.: Angew Chem <strong>2012</strong>, accepted.


NHC-Silver(I) and NHC-gold(I) complexes as bioorganometallic anticancer and antibacterial drugs<br />

Tacke M; Hackenberg F; Patil S; Lally G; Streciwilk W<br />

UCD School of Chemistry and Chemical Biology, University College Dublin, Belfield, Dublin 4, Ireland<br />

The synthesis of N-heterocyclic carbene (NHC) silver(I) acetate complexes with varying lipophilic benzyl-substituents<br />

at the 1 and 3 positions of the (benz)imidazole ring was achieved by reaction of silver(I) acetate with the<br />

corresponding (benz)imidazolium bromide or iodide salts [1-7]. These NHC-silver(I) acetate derivatives exhibit<br />

interesting structural motifs in the solid state and proof to be soluble and stable in biological media. The preliminary<br />

antibacterial activity of all the compounds was studied against Gram-negative bacteria Escherichia coli, and Grampositive<br />

bacteria Staphylococcus aureus using the Kirby-Bauer disk-diffusion method. Almost all the NHC-silver(I)<br />

acetate complexes have shown high antibacterial activity compared to the NHC-precursors. The antibiotic lead<br />

compound SBC3, which is synthesised from 4,5-diphenyl-1,3-dibenzyl-imidazole, reaches an antibacterial activity<br />

comparable to marketed modern antibiotics. In addition, the NHC-silver complexes had their cytotoxicity investigated<br />

through MTT based preliminary in vitro testing on the human renal cancer cell line Caki-1 in order to determine their<br />

IC50 values. Here, NHC-silver(I) acetate complexes were found to have IC50 values as low as 1.2 μM for (1-methyl-<br />

3-(p-cyanobenzyl) benzimidazole-2-ylidene) silver(I) acetate, which has become the anticancer lead structure SBC1.<br />

The best corresponding NHC-Au(I)Cl complex SBC2 reaches an IC50 value of 10 μM against the cancer cell line<br />

CAKI-1 [8,9]. In addition, results of SBC1 against drug-resistant cancer cell lines, transport and target assays as well<br />

as in vivo testing in zebrafish and mouse will be communicated.<br />

The authors acknowledge IRCSET and UCD for funding.<br />

References:<br />

1. F. Hackenberg, A. Deally, G. Lally, S. Malenke, H. Müller-Bunz, F. Paradisi, S. Patil, D. Quaglia, M. Tacke, Int J Inorg Chem <strong>2012</strong>,<br />

DOI: 10.1155/<strong>2012</strong>/121540<br />

2. S. Patil, A. Deally, B. Gleeson, F. Hackenberg, H. Müller-Bunz, F. Paradisi, M. Tacke., Z Allg Anorg Chem 2011, 637: 386–396.<br />

3. S. Patil, A. Deally, B. Gleeson, H. Müller-Bunz, F. Paradisi, M. Tacke. Novel Benzyl-Substituted N-Heterocyclic Carbene–Silver Acetate<br />

Complexes: Synthesis, Cytotoxicity and Antibacterial Studies, Metallomics, 2011, 3: 74–88.<br />

4. S. Patil, K. Dietrich, A. Deally, B. Gleeson, H. Müller-Bunz, F. Paradisi, M. Tacke, Helv Chim Acta 2010, 93: 2347–2364.<br />

5. S. Patil, A. Deally, B. Gleeson, H. Müller-Bunz, F. Paradisi, M. Tacke, Appl Organomet Chem 2010, 24: 781–793.<br />

6. S. Patil, J. Claffey, A. Deally, B. Gleeson, M. Hogan, L. M. Menéndez Méndez, H. Müller-Bunz, F. Paradisi, M. Tacke, Eur J Inorg Chem<br />

2010, 1020–1031.<br />

7. S. Patil, M. Tacke., „NHC-Silver(I) Acetates as Bioorganometallic Anticancer and Antibacterial Drugs. Insights into Coordination,<br />

Bioinorganic and Applied Inorganic Chemistry.“ Edited by M. Melník, P. Segľa, M. Tatarko., Press of Slovak University of Technology,<br />

Bratislava, 2011: 555–566.<br />

8. L. Kaps, B. Biersack, H. Müller-Bunz, K. Mahal, J. Münzner, M. Tacke, T. Mueller, R. Schobert., J Inorg Biochem <strong>2012</strong>, 106: 52–58.<br />

9. S. Patil, A. Deally, F. Hackenberg, L. Kaps, H. Müller-Bunz, R. Schobert, M. Tacke., Helv Chim Acta 2011, 94: 1551–1562.<br />

68 Sessionvorträge Targets


Sessionvorträge Tabletten<br />

Session Nanomedizin<br />

Nanomedicines for targeted drug delivery to the inflamed intestinal mucosa<br />

Collnot E M1 ; Leonard F2 ; Hare E1,3 ; Mell N1 ; Ali H2 ; Lehr C M1,2 1 Department of Drug Delivery (DDEL), Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infectious<br />

Research (HZI), Saarland University, Campus A4.1, 66123 Saarbruecken, Germany<br />

2 Biopharmacy and Pharmaceutical Technology, Saarland University, 66123 Saarbruecken, Germany<br />

3 Hebrew University of Jerusalem, Faculty of Medicine, School of Pharmacy, Institute for Drug Research, Jerusalem 91120, Israel<br />

Inflammatory bowel diseases (IBD) such as Crohn’s disease or Ulcerative colitis are autoimmune, chronic, episodic,<br />

inflammatory conditions of the gastrointestinal tract. Classical therapy of IBD with steroids or immunomodulators<br />

delivered via pellets, capsules, or tablets is often inefficient mainly due to limited drug release time and enhanced<br />

elimination of drug carriers as the result of diarrhoea. The emergence of biologicals has significantly increased the<br />

treatment options for IBD in recent years. Still, these new therapeutics have to be applied systemically resulting in<br />

sometimes severe adverse effects. Nanomedicine may enhance the efficacy of conventional IBD therapeutics and<br />

open up new routes of application for next generation drugs: Passively targeting the inflamed intestinal areas,<br />

nanoparticles of ~100 nm size have been shown to accumulate in the inflamed tissue, while bigger particles of 1 or<br />

10 µm showed no specificity for diseased intestinal areas [1]. Moreover, polylactide-co-glycolide (PLGA)<br />

nanoparticles loaded with rolipram showed higher and prolonged anti-inflammatory activity and reduced central<br />

nervous adverse effects in a TNBS rat model [1].<br />

Further investigating this EPR (enhanced permeability and retention) – like effect we developed a co-culture model of<br />

the inflamed intestine based on Caco-2 enterocytes and blood derived macrophages and dendritic cells [2]. Via<br />

addition of the cytokine interleukin-1β a reversible inflammation can be induced. In response to the stimulus a<br />

decrease in barrier function, a re-organization of tight junctions, a release of inflammation markers and increased<br />

mucus production can be observed mimicking pathophysiological phenomena observed in IBD in vivo.<br />

Different nanoformulations of the glucocorticoid budesonide were applied in the in vitro system and evaluated for<br />

their mechanism of accumulation and anti-inflammatory activity [3]. Both budesonide loaded PLGA nanoparticles and<br />

the free drug solution were able to recover epithelial barrier function (quantified via TEER) and to reduce release of<br />

inflammation marker IL-8. However, treatment with free budesonide solution was only effective for a short time as<br />

after 48 h a rebound of IL-8 release was observed. In contrast PLGA budesonide treated cells retained low IL-8<br />

levels. The prolonged activity of the nanoparticulate formulation can be ascribed to the formation of local drug<br />

depots, as particles accumulate between the cells in the area of the tight junctions. A liposomal budesonide<br />

formulation seemed to be preferentially taken up and processed by the immune cells in the co-culture model. The<br />

resulting dose dump negatively affected the barrier function and resulted in increased IL-8 levels.<br />

Aiming to further enhance the efficacy of nanocarrier accumulation, active targeting ligands for the inflamed intestinal<br />

mucosa were investigated. In the inflamed colonic mucosa of colitis patients an increased expression of transferrin<br />

receptor (TfR) has been reported both in the basolateral aspects of crypts and on the apical surface [4]. Indeed anti-<br />

TfR immunoliposomes applied the luminal side showed greatly enhanced adherence in vivo compared to nonspecific<br />

immunoliposomes.<br />

As a first topical application of a biological in IBD therapy a nanoformulation of the potent, anti-inflammatory cytokine<br />

IL-10 was developed. IL-10 is a promising IBD therapeutic, but failed in clinical trials upon subcutaneous injection,<br />

due to its short serum half-life and severe adverse effects at higher doses. Drug loaded albumin nanoparticles were<br />

prepared by spray drying and embedded in Eudragit S-100 microparticles to ensure stability during gastrointestinal<br />

passage. The nanoparticle in microparticle oral system succeeded in protecting IL-10 integrity during the preparation<br />

process and releasing bioactive cytokine at the desired pH of 7.4.<br />

References:<br />

1. Collnot, E.M., H. Ali, and C.M. Lehr, J Control Release <strong>2012</strong>.<br />

2. Leonard, F., E.M. Collnot, and C.M. Lehr, Mol Pharm 2010. 7(6): 2103–2119.<br />

3. Leonard, F., et al., ALTEX <strong>2012</strong>, 29(3): 275–285.<br />

4. Harel, E., et al., PLoS ONE 2011, 6(9): e24202.<br />

Sessionvorträge Tabletten 69


Selective gene vehicles for enhanced DNA and siRNA delivery<br />

Schümmelfeder J2 , Dayyoub E1 , Marxer E1 , Brüßler J1 , Schäfer J1 , Bakowsky U1 1Marburg University, Department of Pharmaceutical Technology and Biopharmaceutics, Ketzerbach 63, D-35037 Marburg, Germany<br />

2Department of Cardiology, St. Georg Hospital Eisenach, Mühlhäuser Strasse 94/95, D-99817 Eisenach, Germany<br />

Gene therapy is believed by many to be the therapy of the 21st century. Successful gene therapy or gene<br />

knockdown depends on the delivery of nucleic acid molecules into cell. While viral vectors have shown high gene<br />

transfer efficacies, their drawbacks include immunogenic/inflammatory responses, limited loading capacity and<br />

issues with regards to large scale manufacturing and quality control. Consequently, numerous non-viral gene delivery<br />

vectors including liposomes (lipoplexes), polycationic polymers (polyplexes) and organic or inorganic nanoparticles<br />

(nanoplexes) have been developed and reported in recent years [1,2,4]. General issues in non-viral transfection<br />

systems are the delivery of DNA for gene transfection into the nucleus or small interfering RNAs (siRNAs) for the<br />

induction of RNA interference (RNAi). RNAi is a powerful method that has been applied successfully for the downregulation<br />

of genes in both functional genetic analysis and gene therapy. It relies small interfering RNAs (siRNAs),<br />

and since all other components of the RNAi machinery are provided by the target cell /target tissue, the efficient<br />

siRNA delivery is of critical importance and has proven to be the bottleneck for successful RNAi applications.<br />

Atherosclerosis is characterized by the buildup of lipid-rich plaques within the blood vessel walls of large arteries, and<br />

underlies the clinical conditions of myocardial infarction, chronic stable angina, stroke and peripheral vascular<br />

disease. The process of atherogenesis is induced by a low-grade inflammatory process in the vascular wall, leading<br />

through various steps to the eventual formation of atheromatous plaque. These plaques may also become<br />

progressively unstable due to local reaction to pro-inflammatory cytokines and proteinases (e.g. IFN-gamma,IL-<br />

18,CD40–CD154, or IL 1 etc.), leading to plaque ulceration or rupture. Additionally activated endothelial cells express<br />

adhesion molecules such as P-selectin and E-selectin, VCAM or ICAM leading to a pro-inflammatory environment<br />

and a mediated activation of clotting cascade [3]. The gene overexpression of such target molecules make them<br />

promising candidates for the treatment by siRNA formulations.<br />

The aim of this research was to develop a gene-transfection system based on site specific lipoplexes and<br />

lipopolyplexes which would possess a nano-scaled size below 200 nm. For the preliminary experiments a luciferase<br />

reporter gene bearing plasmid DNA (pcmv beta) was used instead of siRNA for the therapeutic application (down<br />

regulation of the targets TNF-alpha, VEGF). The pDNA was pre-condensed with cationic polymers [1] and/or<br />

complexed [2,4] with different lipid mixtures (DPPC, DOPC, DSPC, Saint, Tetraetherlipids or Cholesterol etc.) to<br />

improve the stability/efficiency and lowering the toxicity. The gene vehicles were targeted to E-selectin via surface<br />

modification with E-Sel antibodies. The transfection efficacy and cell viability was investigated on the HUVEC and<br />

CHO-E cells under shear flow conditions.<br />

Acknowledgments: This study was supported by “Deutscher Akademischer Austauschdienst“ (DAAD) and by “Deutsche Forschungsgemeinschaft”<br />

(Forschergruppe FG 495 and FG 629, UB). We are grateful to Prof. Dr. J. Aigner for the development of the polyplex vehicles<br />

and the technical support.<br />

References:<br />

1. Schäfer J, Höbel S, Bakowsky U, Aigner A: Biomaterials 2010, 31(26): 6892–6900.<br />

2. Kneuer C et al.: J Nanosci Nanotechnol 2006, 6(9-10): 2776–2782.<br />

3. Kneuer C et al.: Drug Discov Today 2006, 11 (21-22): 1034–1040.<br />

4. Tabatt K et al.: J Controlled Release 2004; 97: 321–332.<br />

70 Sessionvorträge Tabletten


Modulating immune responses of human cells by adjuvant loaded particulate carriers<br />

Wischke C1,2 , Mathew S1,2 , Roch T1 , Frentsch M2 , Lendlein A1,2 1Center for Biomaterial Development, Helmholtz-Zentrum Geesthacht, Kantstr. 55, 14513 Teltow, Germany<br />

2Berlin-Brandenburg Centre for Regenerative Therapies, Berlin and Teltow, Germany<br />

Building an efficient immunological memory to avoid infections remains a challenge<br />

for various types of pathogens, particularly when purified, poorly immunogenic antigenic<br />

structures should be used for safety reasons. Therefore, antigens are often<br />

combined with cocktails of immunostimulating compounds, which in a rather uncontrolled<br />

process induce a proinflammatory environment and frequently have led to<br />

discussions about adjuvant induced severe vaccine reactions. For enabling tailored<br />

immune responses including efficacy against intracellular pathogenes, carrier systems<br />

are of high interest, as they may co-deliver the antigen(s) and a specific adjuvant<br />

[1]. Particles from poly[(rac-lactide)-co-glycolide] (PLGA) have shown their<br />

suitability as protein carriers as they are very efficiently taken up into cells by<br />

Fig. 1: Intracellular localization of<br />

MP, reproduced from [3] with<br />

permission, Copyright 2006.<br />

phagocytosis [2] (Fig. 1) and, moreover, facilitated phagolysosomal escape to the cytosol as relevant to induce<br />

immunity against intracellular pathogens. The carrier size should be in the upper nanosize range or, preferentially,<br />

microparticles (MP) < 10 µm, in order to efficiently enable phagocytosis as uptake mechanism and deliver sufficient<br />

levels of payload.<br />

Importantly, the MP carriers themselves were shown to be inert and did not induce activation of antigen presenting<br />

cells (APC) such as human primary dendritic cells (DC) [3], which is required to tailoring the immune response by the<br />

selectively added adjuvant. When poly[(riboinosinic acid)-co-(ribocytidylic acid)] as a Toll-like receptor 3 (TLR3)<br />

agonist was bound to surface-functionalized MP [3] in a layer-by-layer approached, only ligand loaded but not blank<br />

particles induced a full antigen-independent maturation of DC [4].<br />

As the safety profile of TLR ligands in general is occasionally subject to<br />

concerns and addressed presently in clinical studies, alternative defined<br />

adjuvants may be required. Therefore, in this study, N-acetylmuramyl–<br />

L-alanyl–D-isoglutamine (MDP) and γ-D-glutamyl-meso-diaminopimelic<br />

acid (iE-DAP) as ligands of cytosolic receptors sensing nucleotide and<br />

oligomerization domains (NOD) were evaluated for their capability to<br />

induce DC maturation and cytokine release after encapsulation into<br />

PLGA MP [5]. This concept was rationalized by the carriers’ capability<br />

to support phagolysosomal escape, thus making available their payload<br />

to the compartment where NOD receptors are expressed. Microencapsulated<br />

NOD agonists but not blank MP or soluble NOD agonists<br />

induced a dose-dependent maturation of DC (Fig. 2), illustrating the<br />

efficiency of the particulate carrier in combination with phagocytosis as<br />

transport mechanism. A proinflammatory DC phenotype with<br />

Fig. 2: Dose dependent maturation of DC. remarkable high levels of released cytokines particularly for iE-DAP<br />

were detected. Importantly, for selectively detecting effects of the studied adjuvant, a strategy is required to exclude<br />

immunogenic impurities and prevent erroneous interpretations of cell activation in the case of impurity-burdened MP.<br />

By different reporter cell lines not sensing MDP and iE-DAP but selectively expressing other pathogen associated<br />

molecular pattern, the purity of the employed carriers and the specificity of results were proven. Based on this, further<br />

exploration of these carriers is planned in vivo.<br />

Acknowledgments: D. Lorenzen, Institute for Tumor Therapy, Duderstadt, Germany, for support in previous cell studies [Fig. 1; Ref. 2-4].<br />

References:<br />

1. Rappuoli, R., Aderem, A.: Nature 2011, 473: 463–469.<br />

2. Wischke, C. et al.: Eur J Pharm Biopharm 2006, 62: 247–253.<br />

3. Wischke, C. et al.: J Contr Rel 2006, 114: 359–368.<br />

4. Wischke, C. et al.: Int J Pharm 2009, 365: 61–68.<br />

5. Wischke, C. et al.: <strong>2012</strong> manuscript in revision.<br />

6. Mathew, S. et al.: <strong>2012</strong> manuscript submitted.<br />

Sessionvorträge Tabletten 71


Acid-Degradable Dextran Particles for the Delivery of Biotheropeutics<br />

Wich P R1 ; Fréchet J M J2 1 Institut für Pharmazie und Biochemie, Johannes Gutenberg-<strong>Universität</strong> Mainz, Staudingerweg 5, 55128 Mainz, Germany<br />

2 College of Chemistry, University of California Berkeley, CA 94720-1460, USA<br />

Polymer-based drug, vaccine and gene delivery systems can be easily modified and are particularly attractive<br />

because of their ability to perform several critical functions simultaneously. These include delivering therapeutic or<br />

other bioactive agents to a specific site via a targeting mechanism, increasing the circulation times of drugs in the<br />

body, protecting drugs from degradation, or increasing the bioavailability of poorly soluble drugs.<br />

Recently, a new class of modular and acid-degradable, polymeric particles using acetal-modified dextran (Ac-DEX)<br />

was developed.[1,2] This biocompatible material can be formulated into particles using a variety of common<br />

emulsion-based techniques enabling control over both size and morphology. Due to their pH sensitivity, Ac-DEX<br />

particles can selectively and rapidly release their encapsulated payload under mildly acidic conditions including those<br />

found in sites of inflammation, tumor tissue, or endocytic vesicles. Upon hydrolytic degradation, Ac-DEX reverts back<br />

to FDA-approved dextran without the generation of acidic byproducts that could damage the encapsulated payload or<br />

lead to inflammation, as has been observed with some other commonly used polymer systems. In addition, Ac-DEX<br />

particle degradation rate can be easily tuned within the time scale of relevant cellular processes. This release<br />

tunability combined with their low-toxicity and payload versatility make Ac-DEX particles an ideal platform for a wide<br />

range of biotherapeutic delivery applications including immunotherapy [2] and gene delivery.[3]<br />

References:<br />

1. Bachelder E.M. et al.: J Am Chem Soc 2008, 32: 10494–10495.<br />

2. Broaders, K.E. et al.: Proc Natl Acad Sci USA, 2009, 106: 5497–5502.<br />

3. Cohen, J. L. et al.: Bioconj Chem 2011, 13: 1902–1905.<br />

72 Sessionvorträge Tabletten


Characterization of Bacterial Nanocellulose as Drug Delivery System for Low and High Molecular<br />

Weight Drugs<br />

Müller A1 ; <strong>Moritz</strong> S1 ; Wiegand C 2 ; Wesar F 3 ; Munteanu M 4 ; Hessler N 4 ; Kralisch D 4 ; Müller F A 3 ; Hipler U-C 2 ;<br />

Fischer D1 1 Department of Pharmaceutical Technology, Friedrich-Schiller-University Jena, Otto-Schott-Str. 41, 07745 Jena, Germany<br />

2 Department of Dermatology, Friedrich-Schiller-University Jena, Erfurter-Str. 35, 07743 Jena, Germany<br />

3 Institute of Materials Science and Technology, Friedrich-Schiller-University Jena, Loebdergraben 32, 07743 Jena, Germany<br />

4 Institute of Technical Chemistry and Environmental Chemistry, Friedrich-Schiller-University Jena, Lessingstr. 12, 07743 Jena, Germany<br />

Synthesized by bacteria of the genus Gluconacetobacter the biopolymer bacterial nanocellulose (BNC) has already<br />

been proven to be advantageous for various biomedical applications as tissue engineering, implant materials, skin<br />

substitutes and wound care [1, 2]. The use of this biomaterial as drug delivery system offers the opportunity to<br />

combine its beneficial material properties (e.g. high purity, mechanical stability and biocompatibility) with the positive<br />

effects of incorporated drugs and opens up new promising fields of application.<br />

Comparative loading and release studies with high molecular weight protein drugs (bovine serum albumin: BSA,<br />

luciferase) as well as different low molecular weight antiseptics (polihexanide: PHMB, PVP-iodine, octenidine) were<br />

performed using planar BNC fleeces produced in a static cultivation process. Therefore BNC samples were loaded<br />

by immersion in aqueous drug solutions and released after different incubation times. Influence of drug<br />

concentration, water content of the BNC samples, incubation time as well as temperature were investigated.<br />

Quantification of the different drugs was performed by UV spectrophotometry (BSA, 278 nm; PHMB, 235 nm; PVPiodine,<br />

352; octenidine, 281 nm), BCA assay and luminescence measurements (luciferase assay system). Drug<br />

distribution in the samples was analyzed by staining of BSA loaded samples with BCA assay reagent and the use of<br />

red brown coloured PVP-iodine. Sample morphology was studied by SEM analysis. Furthermore, drug release data<br />

were examined according to Ritger-Peppas equation. Polyacrylamide gel electrophoresis of released BSA as well as<br />

measurement of biological activity of released luciferase was conducted to study protein stability. Antibacterial<br />

efficacy of antiseptic BNC extracts prepared according to DIN EN ISO 10993-12 against Staphylococcus aureus was<br />

tested by microplate laser nephelometry. Furthermore, a direct challenge test (JIS L 1902:2002) was performed for<br />

measuring the antibacterial efficacy of PVP-iodine loaded samples. Cell proliferation of human HaCaT keratinocytes<br />

incubated with extracts of BNC loaded with antiseptics was quantified using a luminometric ATP-luciferase assay for<br />

cytotoxicity studies. The proteins as well as the low molecular antiseptic drugs showed the same release pattern,<br />

characterized by a burst in the beginning followed by a slower release rate until 48 hours. An overlay of diffusion- and<br />

swelling-controlled drug transport mechanisms was found by mathematical analysis (Ritger-Peppas equation) for<br />

both types of drugs. Optimized loading techniques allowed a homogenous drug distribution in the loaded samples.<br />

Protein stability could be maintained during loading and release as shown by gel electrophoresis as well as biological<br />

enzyme activity. Extracts of BNC hydrogels loaded with PHMB and octenidine showed a strong inhibition of<br />

Staphylococcus aureus growth. Antiseptic efficacy of PVP-iodine loaded BNC could be confirmed with the direct<br />

challenge test. Cell proliferation of HaCaT keratinocytes in cytotoxicity assays was found to be dependent on<br />

antiseptic drug type and extraction ratio. Whereas PVP-iodine extracts had no influence on cell proliferation, PHMB<br />

and octenidine extracts exhibited a decrease of cell proliferation at different extraction ratios (PHMB: 0.1 g:50 mL,<br />

octenidine: 0.001 g:50 mL), which has to be considered for determination of active loading concentration in wound<br />

dressing development.<br />

In conclusion, obtained results characterize drug loading to and release from the BNC biomaterial and confirm the<br />

suitability of BNC for drug delivery applications for high molecular as well as low molecular weight drugs.<br />

Acknowledgments: This study was funded by the Thuringian Ministry of Education, Science and Culture (B714-10032) as well as the European<br />

Fund for Regional Development. Additionally, the authors would like to thank Ramona Brabetz as well as Elena Pfaff for the excellent technical<br />

assistance.<br />

References:<br />

1. Kralisch, D. et al.: Biotechnol Bioeng 2010, 105(4): 740–747.<br />

2. Klemm, D. et al.: Angew Chem Int Ed 2011, 50(24): 5438–5466.<br />

Sessionvorträge Tabletten 73


Biomedical applications of magnetite in the respiratory system – Toxicological considerations and<br />

mechanistic studies in A549 lung cells<br />

Könczöl M1,2 , Gminski R1 , Gieré R3 , Merfort I2 , Mersch-Sundermann V1 1 Institut für Umweltmedizin und Krankenhaushygiene, <strong>Universität</strong>sklinikum Freiburg<br />

2 Institut für Pharmazeutische Biologie und Biotechnologie, <strong>Universität</strong> Freiburg<br />

3 Institut für Geowissenschaften, <strong>Universität</strong> Freiburg<br />

Magnetite, a ferromagnetic iron oxide, represents a promising material within the broad field of nanomaterials for<br />

biomedical applications [1]. Beside its use in magnetic resonance imaging or hyperthermia, research has been<br />

focused on its potential to serve as an inhalative carrier system for drugs or vaccines. The lack of consistent data on<br />

pulmonary toxicity requires more systematic investigation of magnetite nano- and micrometer particles. In a former<br />

study, magnetite was thoroughly investigated with regard to its potential to induce toxic effects and to influence<br />

signaling pathways. It was clearly demonstrated that reactive oxygen species (ROS)-formation led to mitochondrial<br />

damage and genotoxic effects in A549 cells [2]. Based on these findings, we here report studies which elucidate the<br />

origin of magnetite-mediated ROS-formation and their influence on cell cycle of A549 human lung epithelial cells.<br />

Additionally, effects on p53 and p21 activation were evaluated by western blots.<br />

After exposure of A549 to magnetite particles, an increased superoxide production, measured by electron<br />

paramagnetic resonance (EPR), was detected, while the glutathion (GSH) level decreased significantly. Moreover,<br />

the particles were able to induce ROS-formation in an acellular environment. Mitochondria or soluble iron did not<br />

contribute considerably to the ROS production. We could show that incubation of A549 cells with inhibitors of<br />

NADPH-oxidase (NOX) prior to particle treatment led to decreased ROS formation measured by EPR and DCFH-DA<br />

assay. Analysis of cell cycle distribution yielded a pronounced sub-G1-peak, which cannot be linked to increased cell<br />

death. Western blot analysis showed no activation of p53 but upregulation of p21 in A549. Surprisingly, exposure to<br />

magnetite leads to p21-mediated G1-like arrest, which has only reported before for low concentrations of microtubule<br />

stabilisation drugs, but so far never after particle exposure. Importantly, the arrested sub-G1 cells were viable and<br />

showed no caspase activation. In summary, magnetite induced a variety of cellular responses in A549 cells in vitro.<br />

Since it cannot be excluded that these toxic effects may be also of relevance in vivo, a fundamental safety profile has<br />

to be compiled before considering nanomaterials for biomedical use in human.<br />

References:<br />

1. Kim, J. et al., Arch Toxicol <strong>2012</strong>, 86(5): 685–700.<br />

2. Könczöl, M. et al., Chem Res Toxicol 2011, 24(9): 1460–1475.<br />

74 Sessionvorträge Tabletten


Session Pharmaverfahrenstechnik<br />

Formulation screening with colloidal drug carrier systems<br />

Bunjes H<br />

Institute of Pharmaceutical Technology, TU Braunschweig, Mendelssohnstraße 1, 38106 Braunschweig, Germany<br />

As a large fraction of newly identified drug candidates is poorly water soluble the use of enabling formulation<br />

technologies becomes increasingly important. In principle, this applies to all potentially relevant routes of<br />

administration but dosage forms for systemic administration, in particular via the parenteral (especially intravenous)<br />

and oral route are usually of special interest. Usually, the development of viable dosage forms for poorly soluble<br />

substances is a comparatively time-consuming process. This creates a kind of dilemma in regard to the rapid<br />

availability of a rather large number of potential drug candidates by modern drug discovery methods. The situation is<br />

further complicated by the usually very limited amount of drug substance available for formulation studies in early<br />

development. In order to rapidly provide suitable formulations for the required tests with poorly water soluble drug<br />

candidates the availability of formulation screening methods for the fast development of formulations with a minimum<br />

amount of drug candidate substance would thus be highly desirable. Ideally, the formulations identified this way<br />

would also bear the potential to be developed into marketable products.<br />

Colloidal carrier systems like drug nanosuspensions or different types of lipid dispersions (such as liposomes, mixed<br />

micellar systems or several types of lipid nanoparticles) are a prominent formulation option for poorly soluble drug<br />

candidates. Properly designed colloidal formulations are, in principle, applicable via all potential routes of<br />

administration and may also be developed into drug products. Compared with more conventional dosage forms (e.g.,<br />

drug solutions) most of them are, however, also comparatively demanding, in particular with regard to their way of<br />

preparation and their physicochemical stability. Only a few of them can be prepared by simple mixing (e.g. mixed<br />

micellar solutions) that would allow for easy screening. For others (like liposomes), small scale dispersion methods<br />

are well established but the whole process of formulation is still quite time-consuming.<br />

The preparation of colloidal carrier systems with micro- and nanostructured devices like by high-pressure<br />

microchannel or premix membrane emulsification [1, 2] or in miniaturized milling devices [3] offer novel approaches<br />

for small scale formulation preparation. Beyond formulation screening with a drug candidate of interest, they are also<br />

very useful for the optimization of formulations and in basic formulation research. The work with such preparation<br />

methods may, however, still require several experiments to determine the loading capacity of a lipid carrier system<br />

for a specific drug. In order to solve this task in a more efficient way, loading of pre-formed colloidal carrier systems is<br />

an interesting option [4]. This presentation will introduce and discuss different approaches for formulation screening<br />

with a special focus on the development of colloidal lipid carrier systems for poorly water soluble substances.<br />

References:<br />

1. Finke, J. et al.: Chem Eng J <strong>2012</strong>, accepted for publication.<br />

2. Joseph, S., Bunjes, H.: J Pharm Sci <strong>2012</strong>, 101(7): 2479–2489.<br />

3. Juhnke, M., Berghausen, J., Timpe, C.: Chem Eng Technol 2010, 33(9): 1412–1418.<br />

4. Kupetz, E. et al.: 8 th PBP World Meeting <strong>2012</strong>, Istanbul, Turkey.<br />

Sessionvorträge Tabletten 75


Pharmaceutical Extrusion Technology – Recent developments<br />

Thommes M<br />

Institute of Pharmaceutics and Biopharmaceutics, Heinrich-Heine-University, Universitaetsstrasse 1, 40225 Duesseldorf, Germany<br />

Pharmaceutical extrusion technology has become an important processing technology within the last few decades.<br />

For this reason, academia, as well as industry, is focusing research efforts on this area. It is likely that extrusion will<br />

become one of the most important technologies for many pharmaceutical applications.<br />

In the last few years the co-rotating twin screw extruder has become the common extruder type due to its versatility.<br />

Its modular setup allows for broad manufacturing process customization to meet different product requirements.<br />

Such flexibility allows multiple unit operations to be performed in a single machine, thereby increasing efficiency.<br />

Based on the plastification method, extrusion processes can be differentiated as wet, hot melt, and cold extrusion.<br />

These have been investigated and used during the past 30 years. Each of these methods has its own advantages<br />

leading to specific applications, such as pelletization, taste masking, and solubility enhancement.<br />

Recent investigations deal with the process understanding (QbD) and downstream processing such as film extrusion<br />

and injection moulding. A lot of research efforts also deal with down-scaling of extrusion equipment to make it<br />

suitable for small-scale production and early development.<br />

Pharmaceutical extrusion technology is an innovative technology that continues to become more relevant, due to its<br />

many assets as compared with other pharmaceutical technologies.<br />

76 Sessionvorträge Tabletten


Hot melt extruded dosage forms<br />

Rein H<br />

Pharmazeutische Technologie, <strong>Universität</strong> Bonn, Gerhard-Domagk-Straße 3, D-53121 Bonn-Endenich, Germany<br />

1. Introduction<br />

Hot melt extrusion is gaining more and more interest in pharmaceutical industry. One of the main advantages, apart<br />

from improving the bioavailability of an API, is the possibility to move away from batch processing towards<br />

continuous production. Mostly the extrusion process itself is already well adjusted. One of the major problems right<br />

now is giving shape to the final products.The aim of our work was to develop continuous processes for the<br />

production of pellets and extruded tablets (extrudets).<br />

2. Pellets [1]<br />

Spherical starch pellets were directly and continuously produced using hot-melt extrusion and die-face pelletisation.<br />

In contrast to conventional pelletisation procedures, a discontinuous spheronisation step is not necessary. Pellets<br />

were produced based on different starches and several other polysaccarids, four different active ingredients<br />

(ibuprofen, paracetamol, phenazon and tramadol-HCl) and various additives. The resulting pellets exhibit a large<br />

mechanical stability, low porosity, small surface area and a a very narrow particle size distribution even in the micron<br />

scale. The drug is either dispersed or dissolved in the polysaccharid melt. Drug loadings of up to 80% are<br />

achievable. The dissolution rate depends on pellet size, pellet porosity, the velocity with which the penetrant diffuses<br />

into the pellet-core, the water solubility of the API, and the molecular structure, composition and the swelling behavior<br />

of the used polysaccarid. The fractional drug release from non eroding starch pellets can be described with the<br />

Noyes-Whitney equation. [2]<br />

3. Tablets/extrudets<br />

Biplan tablets were produced by using a co-rotating twin-screw extruder (ZSE 27 HP-PH, Leistritz AG, D-Nürnberg).<br />

For the final shaping of the tablets, the still elastic strands were cut with a rotary fly-knife cutting machine (Dynamat,<br />

Metzner, D-Neu Ulm), which is generally used in plastics industry for cutting of wires, cables or seals. [3] Another<br />

important aspect of our work concerns data-acquisition during the cutting process. Monitoring the energy of each cut<br />

gives an interesting insight into the process. This might be applied as a useful in-process tool.<br />

Tablets were produced based on several starches and various different polysaccharides, which were well known in<br />

the food industry, e.g. gelatine, gummi arabicum, pectine or locust bean gum and several additives. Depending on<br />

the used polysaccharide the production of melting tablets, lozenges or dosage forms with fast or prolonged API<br />

release is possible.<br />

Acknowledgments:<br />

1, Roquette, F-Lestrem, donation of starches.<br />

2. Leistritz Extrusionstechnik, D-Nürnberg, allocation of a twin-screw extruders (ZSE 18 HP-PH and ZSE 27 HP-PH).<br />

3. Metzner Maschinenbau, D-Neu-Ulm, allocation of a rotary fly-knife cutting machine (Dynamat 20/40).<br />

References:<br />

1. Bialleck S, Rein H,: Eur J Pharm Biopharm 2011, 79: 440–448.<br />

2. Bialleck S, Rein H,: Stärke <strong>2012</strong>, 64: 408–419.<br />

3. Kudernatsch, H., 2011, Pharm Ind 2011, 73: 2050–2060.<br />

Sessionvorträge Tabletten 77


Nanogrinding of naproxen: influence of process parameters on product quality<br />

Bitterlich A1 , Komoß C2 , Dengler M, Bunjes H 2 , Kwade A1 1Institute for Particle Technology, TU Braunschweig, Volkmaroder Str. 5, 38104 Braunschweig, Germany<br />

2Institute of Pharmaceutical Technology, TU Braunschweig, Mendelssohnstraße 1, 38106 Braunschweig, Germany<br />

Nanosized active pharmaceutical ingredients (APIs) increasingly attract interest in pharmaceutical industries. Up to<br />

now, five drug-containing nanoparticulate formulations are on the market [1]. In micronized form, all corresponding<br />

APIs exhibit a poor water solubility and, consequently, a low bioavailability after oral administration. By reducing the<br />

particle size, the dissolution rate can be strongly increased which leads to an improved biopharmaceutical profile.<br />

Wet media milling is often used to produce API nanoparticle suspensions. In this study the influence of the process<br />

parameters on product quality during the nanogrindnig process was investigated. A planetary ball mill (PM400,<br />

Retsch GmbH) was modified according to [2] to enable parallel small scale screening with 12 samples of 25 mg API<br />

each. Inside the grinding chambers, the anti-inflammatory API naproxen (kindly provided by Novartis Pharma AG) in<br />

aqueous suspension experienced high shear and compression forces between grinding media of different size,<br />

material and shape which were brought into relative motion by centrifugal forces.<br />

In a first step screening experiments were conducted, using various polymers and surfactants as stabilizers to<br />

identify an optimum formulation against agglomeration phenomena. With this formulation (10 % naproxen, 2.5 %<br />

Kollidon ® 30) the influence of process parameters on product quality was investigated. Within a certain range, the<br />

product quality (regarding particle size and distribution width) increased by using small grinding beads, spherical<br />

grinding media, high centrifugal forces and a high density of the grinding media.<br />

In addition, some interesting stabilizer and process parameter dependent phenomena were observed during the<br />

grinding process: Different product shapes were obtained by varying the stabilizer. When using PVP (Kollidon ® 30) or<br />

PVA (Mowiol ® 3-85) grades, the product particles exhibited a round morphology after the grinding process. By using<br />

Tween ® 80 or HPMC (Pharmacoat ® 603) as stabilizer, however, the product particle shape was more needle-like<br />

(figure 1). Furthermore, the product particle shape could be influenced by adjusting the process parameters: By<br />

applying grinding media made from yttrium stabilized zirconia with a diameter of 100 µm at high centrifugal forces of<br />

the mill, even the morphology of the product particles stabilized with Kollidon ® 30 changed from “round” over “needlelike”<br />

to finally “disc-like” over a grinding time of 960 min in the modified planetary ball mill. Apparently, the high<br />

(mechanic) energy transferred to the product led to a growth of the API crystals under these process conditions<br />

rather than to a real comminution process.<br />

Figure 1: TEM micrograph of a Mowiol ® 3-85 (left) and Tween ® 80 (right) stabilized<br />

naproxen suspension after 4 h grinding time<br />

References:<br />

1. Merisko-Liversidge, E., Liversidge, G.G.: Adv Drug Deliv Rev 2011, 63: 427–440.<br />

2. Juhnke, M., Berghausen, J., Timpe, C.: Chem Eng Technol 2010, 33: 1412–1418.<br />

78 Sessionvorträge Tabletten


Session Bioanalytik: Testsysteme und Datenqualität<br />

Image Processing in medicinal microscopy<br />

Matz M1 , Schumacher K2 , Rustenbeck I2 , Baumann K1 1Institute of Medicinal and Pharmaceutical Chemistry, TU Braunschweig, Mendelssohnstraße 1, 38106 Braunschweig, Germany.<br />

2Institute of Pharmacology, Toxicology and Clinical Pharmacy, TU Braunschweig, Mendelssohnstraße 1, 38106 Braunschweig, Germany.<br />

In the last decade, total internal reflection fluorescence microscopy (TIRF-M) was established as a standard method<br />

for observing molecular phenomena close to the cell membrane. It is used here to study the behavior of insulin<br />

granules. The obtained TIRF images and video sequences are usually evaluated with commercial software<br />

packages, which allow counting of the labeled cell components and tracking of single labeled particles (i.e. insulin<br />

granules).<br />

Here we present a new and sophisticated program for the exploration of TIRF video sequences, enabling the user to<br />

simultaneously count and track all visible labeled objects. Moreover, the tracked objects can be characterized with<br />

respect to velocity, size, kinetic behavior, and exocytosis. The program starts with background subtraction. Next, a<br />

3D convolution is carried out for noise reduction. Afterwards all fluorescing objects are tracked down and are being<br />

followed. A specifically tailored clustering routine in combination with an edge filter, which detects jumps, is used<br />

exocytosis detection.<br />

While methods for object identification and single particle tracking are well established in the literature, this talk<br />

focuses on new methods for the characterization of granule tracks. The goal is to estimate whether a particular object<br />

moves due to diffusion or some kind of directed movement.<br />

A detailed analysis of the detection of exocytitic events will also be presented. Simply using an edge filter on the<br />

tracks yields too many false positive and false negative events. The clustering routine employed here, is able to<br />

identify even vague increases in fluorescence intensity, which is highly related to membrane fusion and the release<br />

of vesicle content.<br />

Sessionvorträge Tabletten 79


The use of microscale thermophoresis (MST) for protein characterization and formulation<br />

Besheer A1 , Abstiens K1 , Winter G1 , Baaske P2 , Duhr S2 1 Pharmaceutical Technology and Biopharmaceutics, Department of Pharmacy, Ludwig-Maximilians-<strong>Universität</strong>, Butenandtstrasse 5-13, D-<br />

81377 Munich, Germany.<br />

2 NanoTemper Technologies GmbH, Flößergasse 4, D-81369 Munich, Germany.<br />

Microscale thermophoresis (MST) is the drift of dissolved molecules along a micron-sized temperature gradient [1]<br />

(Figure 1). The strength and direction of this molecular thermal diffusion depend mainly on the molecular surface<br />

area, the strength of ionic shielding and on the molecule’s hydration shell [2]. Changes in these properties lead to<br />

alterations in the thermophoretic motion, which make MST a very sensitive tool to analyse structural changes in<br />

proteins. Accordingly, MST was successfully applied for the measurement of macromolecular interactions and<br />

binding affinities [3]. By Applying MST to investigate protein unfolding, we found out that the changes in protein<br />

hydration accompanying unfolding lead to significant changes in thermophoresis, allowing the determination of the<br />

protein’s melting point (Tm). For instance, it was possible to determine Tm for recombinant human granulocyte colony<br />

stimulating factor (rh-GCSF) at different pHs and sucrose concentrations, and validate the results by fluorescence<br />

and microcalorimetric measurements. In this respect, MST offers a number of advantages as it is a label-free<br />

technique, requiring very small sample volumes (< 3 µl) and is able to measure protein thermophoresis over a wide<br />

concentration range (nM to mM). Furthermore, it is an entirely optical method, which is contact-free and therefore<br />

minimizes contamination of the sample. In comparison to other methods, it is not dependant on the surroundings of<br />

tryptophan and does not require the use of external fluorescent dyes like in fluorescence-based techniques, nor is it<br />

limited by low concentration ranges like microcalorimetry and circular dichroism, or the need for relatively<br />

concentrated solutions as in ATR-FTIR. The potential of MST to deliver information about protein unfolding can thus<br />

be exploited over a wide range of applications in protein science.<br />

Figure 1. A) Experimental setup<br />

for label-free MST involves the<br />

use of UV-LED fluorescence for<br />

concentration measurement<br />

through the intrinsic tryptophan<br />

fluorescence. An IR-Laser<br />

produces a temperature<br />

gradient on the order of<br />

0.1 K/µm. Capillaries containing<br />

< 3 µl samples are heated to<br />

the desired temperature.<br />

Samples can show B) a<br />

thermophobic behaviour, where<br />

molecules diffuse away from the<br />

IR-Laser or C) a thermophilic<br />

behaviour moving towards it.<br />

References<br />

1. Iacopini, S., Rusconi, R., & Piazza, R., Eur Phys J E Soft Matter 2006 19: 59–67.<br />

2. Duhr, S. & Braun, D., Proc Natl Acad Sci USA 2006 103: 19678–19682.<br />

3. Wienken, C .J., Baaske, P., Rothbauer, U., Braun, D., & Duhr, S., Nat Commun 2010 (1): 100.<br />

80 Sessionvorträge Tabletten


High-Throughput Protein Melting Temperature Analysis by Differential Scanning Fluorimetry<br />

Menzen T, Frieß W<br />

Ludwig-Maximilians-<strong>Universität</strong> München, Department of Pharmacy, Pharmaceutical Technology & Biopharmaceutics; Butenandtstraße 5, D-<br />

81377 Munich, Germany<br />

Unfolding and aggregation of therapeutic proteins are severe problems in drug formulation, because they are linked<br />

to immunogenic reactions and reduced efficiency. Thus, the investigation of stabilizing conditions is essential during<br />

development. The protein melting temperature (Tm) resembling the temperature at which half of the protein is in a<br />

denatured state, is commonly used to assess overall thermal stability. Stabilizing conditions will shift Tm to higher<br />

values whereas destabilizing effects reduce Tm. Due to the wealth of formulation parameters, such as buffers, pH,<br />

excipients, and combinations thereof a high-throughput Tm assay would be beneficial. Differential Scanning<br />

Calorimetry (DSC) as the gold standard for Tm analysis even in an automatized capillary system is limited to a few<br />

samples per day of several hundred microliters of volume. Differential Scanning Fluorimetry (DSF) was introduced in<br />

2001 by Pantoliano and coworkers and allows the determination of Tm by tracking the thermal unfolding of the<br />

protein by means of an environmental sensitive fluorescent dye [1]. The application of DSF for high-throughput Tm<br />

analysis of therapeutic proteins with low consumption of both time and material has been described [2-4]. But this<br />

method to determine Tm is limited in presence of surfactants which are frequently used as stabilizers in protein<br />

formulation. SYPRO® Orange is commonly utilised because it shows bright fluorescence after binding to the<br />

exposed hydrophobic structures of the unfolded protein. However, the unfolding transition of the protein is covered by<br />

a high background fluorescence of the dye trapped inside the hydrophobic core of the micelles formed by the<br />

surfactants present. In this study we demonstrate how to overcome this limitation to determine Tm of a therapeutic<br />

monoclonal antibody (MAb) with DSF in presence of surfactants typically used as stabilizing agent polysorbate 20,<br />

polysorbate 80, and poloxamer 188 in concentrations above the CMC. The molecular rotor probe DCVJ (4-<br />

(dicyanovinyl)julolidine) is sensitive to changes in protein structure and shows less interactions with micelles [5]. By<br />

careful correction of the background fluorescence of corresponding placebo samples, Tm values of surfactant<br />

containing protein formulations can be determined, whereas it is not possible solely based on the original sample<br />

traces. The resulting Tm values correlate very well with Tm values obtained by DSC and in surfactant free<br />

formulations. Tm values could be determined in a concentration range between 4 and 40 mg/ml MAb with DCVJ and<br />

SYPRO® Orange in presence of surfactants. Despite the limited sensitivity of DCVJ compared to SYPRO® Orange,<br />

DCVJ allowed Tm determination in various cases in which SYPRO® Orange failed. Additionally, we observed that<br />

the appearance of turbidity marking the formation of aggregates of this MAb is linked to the second unfolding<br />

transition (Tm2). This indicates that unfolding of specific domains is decisive for aggregation. We conclude that DSF<br />

is a very promising method for high-throughput Tm analysis during therapeutic protein formulation development and<br />

can also be used in presence of surfactants, a previously critical limitation.<br />

References:<br />

1. Pantoliano, M. W. et al.: J Biomol Screen 2001, 6(6): 429–440.<br />

2. He, F. et al.: J Pharm Sci 2010, 99(4): 1707–1720.<br />

3. Goldberg, D. S. et al.: J Pharm Sci 2011, 100(4): 1306–1315.<br />

4. Li, Y. et al.: J Pharm Sci 2011, 100(6): 2120–2135.<br />

5. Hawe, A. et al.: Pharm Res 2010, 27(2): 314–326.<br />

Sessionvorträge Tabletten 81


NMR spectroscopy of high molecular weight protein complexes<br />

Nagaraj M1 ; Schubeis T1 ; Ahmed M1 ; Zimmer A1,2 ; Ritter C1 1 Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany<br />

2 current address: Karolinska Institutet, SE-171 77 Stockholm, Sweden<br />

High-resolution structural information of protein targets is of crucial importance for rational drug design approaches.<br />

X-ray crystallography and solution Nuclear Magnetic Resonance (NMR) are standard techniques to determine the<br />

structures of soluble proteins. However, large protein complexes are still extremely challenging to study, as they are<br />

difficult to crystallize or inherently non-crystalline, as is the case e.g. for fibrillar proteins. Recent advances in NMR<br />

spectroscopy, isotope labelling schemes and protein sample preparation strategies have enabled new approaches to<br />

obtain atomic-resolution structural details, and even high-resolution structures, of ordered, high molecular weight<br />

protein complexes. Examples include fibrillar aggregates, microcrystalline samples, oligomeric assemblies and<br />

membrane proteins.<br />

Here, we present solution- and solid state NMR based approaches [1-3] and specific isotope labelling strategies to<br />

study the structural details of the amyloid-like bacterial virulence factor curli. Amyloid fibrils are ordered aggregates<br />

that have a high content of specific eta-sheet secondary structure. Long associated with fatal human diseases such<br />

as Alzheimer’s, Parkinson’s and prion diseases, the amyloid fold is now known to be formed natively by a diverse set<br />

of proteins without toxic side-effects. Amyloid fibrils appear to be particularly abundant as adhesive surface<br />

structures on bacteria forming multi-cellular communities called biofilms. Understanding the structural properties of<br />

these amyloid fibrils, as well as the details of their biogenesis, is therefore of great interest for infection research. In<br />

addition, they allow the straight-forward correlation of structure and function within the native organism. They are<br />

thus promising targets to study general aspects of amlyoid formation.<br />

T. Schubeis greatly acknowledges a scholarship from the HZI Graduate School. M. Ahmed greatly acknowledges the funding from the German<br />

Research Foundation.<br />

References:<br />

1. Ritter, C. et al.: Nature 2005, 435: 844–848.<br />

2. Siemer, A.B. et al.: Angew Chem Int Ed 2005, 44: 2441–2444.<br />

3. Wasmer, C. et al.: J Mol Biol 2010, 402(2): 311–325.<br />

82 Sessionvorträge Tabletten


Session Spezielle Darreichungsformen<br />

Orodispersible dosage forms – novel concepts, innovative products, new therapeutic options<br />

Breitkreutz J<br />

Heinrich Heine University Düsseldorf, Institute of Pharmaceutics and Biopharmaceutics, 40225 Düsseldorf, Germany<br />

Orodispersible dosage forms are pharmaceutical drug formulations which rapidly disintegrate into multiple particles in<br />

the oral cavity. Whereas oral lyophilisates 1 and orodispersible tablets (ODTs) 2 have been used for years in various<br />

indications throughout the world, orodispersible films (ODFs) 3 and orodispersible minitablets (ODMTs) 4 have been<br />

licensed most recently for the European and the national German market. Orodispersible films have been recently<br />

included in the Ph.Eur. for the first time. Some further production methodologies such as electrospinning are<br />

currently under investigation, but have not reached industrial production scale yet.<br />

The gained popularity of orodispersible drug formulations is due to the fact that these rapidly disintegrating dosage<br />

forms combine beneficial properties of both liquid and solid dosage forms. They enable improved drug stability over<br />

the shelf-life and usually do not contain any potential harmful ingredients like preservatives or antioxidants, but offer<br />

precise dosing, ease of administration and swallowing. Hence, they are ideal dosage forms for patients with diseaserelated<br />

swallowing issues 5 , in particular for the paediatric and geriatric population 6 .<br />

Some recent advances in material science and improved engineering processes seem to overcome the previously<br />

used energy and time consuming freeze-drying technology for preparing orodispersibles. Ready-to-use tabletting<br />

excipients such as Ludiflash ® , Pharmaburst ® , Pearlitol Flash ® or Parteck ® ODT promise easy and safe production by<br />

simple mixing and direct compression. However, the resulting disintegration times are usually much higher as for the<br />

oral lyophilisates. More recently the ODMT concept was introduced by our group using orodispersible minitablets of<br />

only 2 mm in diameter or even less made from ready-to-use excipients or self-prepared mixtures which exhibit short<br />

disintegration times below 5 s which is very similar to the marketed oral lyophilisates. As the total minitablet mass is<br />

restricted to about 7 mg, the maximum drug load is also limited to approximately 2.5 mg. Complying with the Ph.Eur.<br />

specifications regarding uniformity of dosage units is a major issue for low-dosed drug substances. A subsequent<br />

granulation step may improve the content uniformity. Orodispersible films can be produced by various methods, such<br />

as solvent-casting, melt extrusion or drug printing. Disintegration times are much higher than for lyophilisates and<br />

ODMTs, but often the film is not sensed at the application site and therefore, the disintegration times might be of<br />

minor importance for patients’ acceptance and therapy adherence. The drug dose is defined by the film area and<br />

thickness, usually containing up to 15-20 mg drug substance. Inclusion of taste masking agents such as ionexchange<br />

resins or cyclodextrins may further lower the maximum drug load.<br />

Although the recent developments and products are quite promising and offer new therapeutic options in various<br />

conditions, the specifications in the pharmacopoeias and the requirements of the regulatory bodies are still not<br />

harmonized and mostly even useless. Especially mechanical properties of the orodispersible dosage forms,<br />

disintegration and dissolution testing need new validated equipment and analytical methods to ensure the quality of<br />

the advanced drug dosage forms.<br />

References:<br />

Seager H., J Pharm Pharmacol 1998, 50: 375–382.<br />

Brown D., Drug Deliv Technol 2003, 3: 58–61.<br />

Hoffmann E.M., Breitenbach A., Breitkreutz J., Exp Opin Drug Deliv 2011, 8: 299–316.<br />

Stoltenberg I., Breitkreutz J., Eur J Pharm Biopharm 2011, 78: 462–469.<br />

Stegemann S., Gogol M., Breitkreutz J., Int J Pharm <strong>2012</strong>, 430: 197–206.<br />

Breitkreutz J., Boos J., Exp Opin Drug Deliv 2007, 4: 37–45.<br />

Sessionvorträge Tabletten 83


Challenges in Developing Gastroretentive Dosage Forms<br />

Neumann M; Schneider F, Weitschies W<br />

Institute of Biopharmaceutics and Pharmaceutical Technology, Department of Pharmacy, University of <strong>Greifswald</strong>, Center of Drug Absorption<br />

and Transport, F.-Hausdorff-Str. 3, 17487 <strong>Greifswald</strong>, Germany<br />

Similarly to food, solid oral dosage forms show highly variable gastric emptying, depending e.g. on their size, volume<br />

and viscosity of gastric contents, intragastric flows, pH-profiles in the gastrointestinal (GI) tract or caloric content of<br />

coadministered food [1]. Furthermore, due to the interdigestive migrating motor complex (IMMC), especially the<br />

housekeeping waves, both particles and monolithic dosage forms are subject to antral forces in the stomach [2].<br />

Once they are emptied through the pylorus, absorption of drugs from the human intestine is a complex and also<br />

highly variable process. Hence, consideration of human physiology is the major prerequisite in the design of dosage<br />

forms for drugs that may benefit from the targeted delivery in the stomach.<br />

Since years, it is an ongoing challenge to overcome physiological processes of gastric emptying, and thereby<br />

achieving prolonged gastric residence times of oral dosage forms in a controlled and reproducible manner.<br />

In general, so called gastroretentive dosage forms are supposed to remain in the stomach over a long period of time,<br />

and thus enable controlled drug delivery by delayed gastric emptying. Furthermore, gastroretention is suitable for<br />

drugs that exhibit e.g. regio specific absorption in the upper small intestine (absorption windows), low solubility or<br />

stability at high pH-values or short plasma half-lives [3]. By means of prolonging gastric residence time the<br />

bioavailability of drugs can be increased.<br />

During the last decades different gastroretentive dosage forms have been designed, which can be classified into four<br />

major categories: (i.) floating systems that are able to swim on top of gastric contents; (ii.) mucoadhesive systems<br />

that attach to the stomach walls; (iii.) high density systems that deposit in the antrum; and (iv.) unfolding or<br />

expanding systems that enlarge in size, and thus cannot pass the pylorus [4].<br />

Our aim is to develop and evaluate new gastroretentive dosage forms, which remain in the stomach in a controlled<br />

manner. On one side, we focus on the design of fast expandable hydrogels of different shapes and the formulation of<br />

a swelling gastroretentive dosage form for pH independent release of acyclovir. This antiviral drug is widely used in<br />

therapy and prophylaxis of herpes infections and exhibits low bioavailability of only 10-30 % primarily due to its<br />

narrow absorption window in the upper small intestine [5].<br />

On the other side, another challenge is the development of in vitro and in vivo test methods to evaluate potential<br />

gastroretentive dosage forms. Critical physiological conditions cannot be reflected by the application of rather simple<br />

in vitro tests as they were used in the past. However, data from tracking of dosage forms in the human GI tract via<br />

commonly applied techniques like gamma scintigraphy, X ray, MRI and magnetic marker monitoring (MMM) raised<br />

the need for more biorelevant in vitro test methods [6]. Therefore, our dosage forms are tested by use of a novel,<br />

mechanical stomach model developed in our laboratory, which simulates antral pressures and housekeeping waves.<br />

Financial support from the German Federal Ministry of Education and Research is gratefully acknowledge (FKZ 13N11370).<br />

References:<br />

1. Streubel, A., Siepmann, J., Bodmeier, R.: Curr Opin Pharmacol 2006, 6: 501–508.<br />

2. Wilson, C.G., Crowley, P.J. (eds): Controlled Release in Oral Drug Delivery (Controlled Release Society) 2011.<br />

3. Klausner, E.A. et al.: J Control Release 2003, 90: 143–162.<br />

4. Bardonnet, P.L. et al.: J Control Release 2006, 111: 1–18.<br />

5. Arnal, J. et al.: J Pharm Sci 2008, 97(12): 5061–5073.<br />

6. Kagan, L., Hoffman, A.: Expert Opin Drug Deliv 2008, 5(6): 681–692.<br />

84 Sessionvorträge Tabletten


Innovative nanoparticular dosage forms – advantages in pharmacokinetics and administration<br />

Rischer M<br />

Losan Pharma GmbH, Department of Drug Delivery & Innovation Projects, Otto-Hahn-Strasse 13, 79395 Neuenburg am Rhein, Germany<br />

Oral and parenteral dosage forms containing nanoparticles in the pharmaceutical development have been proposed<br />

and used over more than 10 years. However, very often results have been published for the application of<br />

nanosuspensions only not considering the difficulties (e.g. agglomeration, loading, scale-up, manufacturing costs)<br />

which may appear if the downprocessing to the final dosage form is performed. In addition sometimes surfactants<br />

or polymers as stabilizers have been used which could not be regarded as safe and toxicological assessments or<br />

studies need to be performed. Basically, these reasons should be mainly responsible that only a few dosage forms<br />

containing nanoparticles have entered the final commercial market yet. Losan Pharma has now completed several<br />

studies with up-to-date Active Pharmaceutical Ingredients (BCS II compounds) and also projects with New<br />

Chemical Entities from Research collaborations and has implemented a process of a cost-effective production of<br />

nanoparticles in oral dosage forms (e.g. tablets). Key figures of this innovative process will be presented as well as<br />

case studies proving the advantages of nanoparticulate dosage forms. Also a comparison of in-vitro with in-vivo<br />

data will be made for certain cases. Finally, new convenient water-free “on the go” applications for such<br />

formulations will be presented. Although, many research companies nowadays try to avoid insoluble new<br />

compounds, there is still a majority of API´s present which do need new and innovative formulations and<br />

applications. Nanoparticulate formulations do also help to reduce environmental contamination by reducing the<br />

dosage for application an aspect which is be more and more considered (e.g. by AMNOG) for future developments.<br />

Sessionvorträge Tabletten 85


Development and pharmaceutical technological applications of Diels-Alder hydrogels<br />

Brandl F1,2 ; Kirchhof S2 ; Hammer N2 ; Messmann V; Goepferich A2 1 Massachusetts Institute of Technology, David H. Koch Institute for Integrative Cancer Research, Cambridge, MA 02139<br />

2 Universitaet Regensburg, Department of Pharmaceutical Technology, 93040 Regensburg<br />

Since the pioneering work of Otto Wichterle in the 1950s, polymeric hydrogels received much interest due to their<br />

chemical versatility and favorable properties. Hydrogels essentially consist of water-insoluble polymers with infinite<br />

molecular weight that bind large amounts of water. Their rheological behavior is between those of liquids and solids,<br />

which makes them ideal candidates for injectable soft tissue implants. In fact, hydrogels have been proposed as<br />

wound dressings, as carrier systems for the controlled release of drugs, as three-dimensional scaffolds for cell<br />

transplantation, and for many other biomedical or technical applications. However, despite many years of research,<br />

finding suitable cross-linking reactions is still an issue. On the one hand, the chosen reaction has to be highly<br />

efficient in order to obtain high cross-linking densities. But on the other hand, the cross-linking reaction must occur<br />

under mild conditions to be compatible with peptides, proteins, or living cells.<br />

O<br />

O<br />

O<br />

O<br />

O<br />

O<br />

N<br />

O<br />

O<br />

O<br />

N<br />

During our research on hydrogels, the Diels-Alder reaction came into our focus. The Diels-Alder reaction is a [4+2]<br />

cycloaddition reaction between a conjugated diene and a substituted alkene which occurs in water without any<br />

catalysts or initiators. The high efficiency and selectivity makes this reaction an ideal, biocompatible cross-linking<br />

reaction. In our group, star-shaped poly(ethylene glycol) is functionalized with furyl and maleimide groups yielding<br />

two complementary macromonomers. Upon mixing the two macromonomers, highly elastic hydrogels with low<br />

swelling ratio are formed within minutes. Currently, we are investigating the suitability of these hydrogels as carrier<br />

systems for the controlled release of pharmacologically active proteins. Special emphasis is put on the delivery of<br />

antibodies, e.g. to treat common eye diseases such as age-related macular degeneration. Other potential<br />

applications are the treatment of postoperative wounds or in the field of tissue engineering.<br />

Acknowledgements: Financial support from Deutsche Forschungsgemeinschaft (grant number GO 565/16-1) is gratefully acknowledged.<br />

O<br />

O<br />

86 Sessionvorträge Tabletten<br />

O<br />

O<br />

N<br />

H<br />

O<br />

N<br />

H<br />

O<br />

O<br />

O<br />

O


Session Optimierte Patientenversorgung<br />

Medication safety research as challenge for pharmaceutical scientists<br />

Jaehde U<br />

Institute of Pharmacy, Clinical Pharmacy, University of Bonn<br />

Various studies have shown that drug therapy is a high-risk process demanding particular attention by all health care<br />

professionals. Many adverse drug events are caused by medication errors, such as overdosing in patients with organ<br />

dysfunctions or inadequate consideration of drug-drug interactions. Whereas there are strict regulatory requirements<br />

to assure the safety of drugs before and after approval (product safety), there are only few systematic approaches to<br />

enhance the safety of the entire medication (patient safety). The Federal Ministry of Health (BMG) has therefore<br />

published an action plan for "Improving medication safety in Germany” which defines specific measures to be taken<br />

by professional institutions and organizations dealing with drug therapy.<br />

As part of this action plan the BMG supports various individual projects and encourages scientists to initiate research<br />

aiming at developing new concepts and models for the improvement of medication safety. At the University of Bonn<br />

various research projects have been conducted in collaboration with pharmacy practitioners as well as physicians<br />

from various medical disciplines. Based on two examples, the challenges and opportunities of medication safety<br />

research for pharmaceutical scientists will be discussed.<br />

Clinical pharmacists to optimise patient safety in drug therapy<br />

Bertsche T<br />

Department of Clinical Pharmacy, University of Leipzig, Germany<br />

Objectives<br />

Drug-related problems (DRP) are frequent in hospitals. Particularly, they occur in prescription and administration and<br />

lead to death, prolonged hospital stay, and severe adverse drug reactions (ADRs). Different reasons, such as<br />

knowledge deficits, account for DRPs. In a setting of limited resources, however, intervention strategies to improve<br />

patient safety have to be prioritised to the most frequent and severe DRPs and have to be tailored to the causes.<br />

Participants and methods<br />

At a university hospital, we identified DRPs in routine care by instructed monitors (clinical pharmacists) and assessed<br />

knowledge deficits by a questionnaire survey. DRPs were classified according to their prevalence, potential risk, and<br />

reasons for their occurrence using a decision-matrix model. Tailored interventions such as teaching sessions, clinical<br />

pharmacist interventions, and newly developed clinical decision support systems were implemented in hospital<br />

settings including e.g. intensive care units, paediatric wards, or cancer patients.<br />

Results<br />

In drug administration, we identified physico-chemical incompatibilities and drug administration by gastric tube as<br />

most prevalent DRPs (n = 1376 processes). DRP prevalence decreased by 59% (p = 0.003, incompatibilities, adult<br />

ICU patients, n = 1108 drug pairs) and by 94% (p < 0.001, gastric tube administration in children, n = 1164<br />

processes). In a follow-up analysis these effects were sustainable. In drug prescription, guideline adherence in pain<br />

treatment for cancer patients increased by 81% (p < 0.001, n = 100 patients), ADRs caused by drug interactions<br />

decreased by 43% (p = 0.001, n = 265 patients) and excessive dosing in patients with renal insufficiency by 51%<br />

(p < 0.001, n = 68).<br />

Conclusions<br />

Tailored interventions in routine care by clinical pharmacists based on teaching sessions and newly developed<br />

electronic systems decreased rates of DRPs and DRP-related ADRs.<br />

Sessionvorträge Tabletten 87


Safety of pharmacotherapy in nursing homes<br />

Thürmann P A<br />

Clinical Pharmacology, Faculty of Health (Department of Human Medicine), HELIOS Clinical Centre, 42283 Wuppertal, Germany<br />

Nursing home residents are seen as a vulnerable population, due to their old age, multimorbidity, high frequency of<br />

dementia and subsequent polypharmacy. Moreover, nursing home residents receive the highest number of<br />

psychotropics. Many of these psychotropic drugs are so-called potentially inappropriate medication for the elderly<br />

(PIM) as defined by Beers (1) and adopted to the German market by Holt et al. (2). Moreover, nursing home<br />

residents are prone to experience drug-related problems (DRPs) and adverse drug-related events (AEs) (3). So far,<br />

no data were available on the frequency and severity of AEs and DRPs in German nursing homes. We thus aimed to<br />

investigate DRPs and AEs in German nursing homes and to develop a concept for an inter-disciplinary intervention in<br />

order to improve safety of drug therapy in nursing homes.<br />

Methods: Drug- related problems and drug-related adverse events were detected by trained clinical pharmacists in<br />

nursing homes willing to participate over a period of four weeks. The study protocol was approved by the responsible<br />

ethics committee, written informed consent was obtained from the residents or their responsible caregivers. Medical<br />

documentation and nurses notes were screened by trained pharmacists, medication was documented and potential<br />

DRPs and AEs were documented in an ACCESS database. Potential AEs were discussed in a multi-disciplinary<br />

team, were causality assessment was performed. Frequency of ongoing and new AEs during the 30-days period was<br />

calculated as the 30-day-prevalence of AEs per 100 nursing home residents months (NHRMs), whereas the 30-daysincidence<br />

included only those AEs which started during the 30-days period and were also related to 100 NHRMs.<br />

Results of the cross-sectional survey were presented in a multi-disciplinary expert workshop and served to develop a<br />

concept to improve safe prescribing and monitoring of pharmacotherapy in nursing homes. This intervention was<br />

implemented in 4 nursing homes, where the prescribing general practitioners, nurses and pharmacists received<br />

special training in geriatric pharmacotherapy. In nursing homes Drug Safety Teams (DSTs) were built consisting of<br />

trained nurses and pharmacists, who followed a pre-specified protocol of medication checks, residents monitoring<br />

and communication with physicians.<br />

Results: 778 nursing home participants in 11 nursing homes participated in the first survey. In total 102 AEs were<br />

detected in 80 seniors (10.3 %). 37 of these AEs (59.7 %) were judged as preventable by the experts and 4 AEs (6.5<br />

%) were ameliorable. The 30-days-prevalence of AEs was estimated with 12.94 AEs/100 NHRMs, the incidence<br />

came to 7.87 AEs/100 NHRMs. 1.493 DRPs were detected, where almost 50 % concerned the documentation, 35 %<br />

the storage and 15 % dispension of drugs. Three months after the implementation intervention the 30-daysprevalence<br />

came to 15.93 AEs per 100 NHRMs and the incidence to 7.62 AEs/100 NHRMs. In contrast to the first<br />

cross-sectional survey a much higher number of adverse events concerning the central nervous system was<br />

observed, probably due to the increased awareness of the nurses. The intervention proved to be feasible, most<br />

nurses would recommend it for other nursing homes.<br />

Discussion: Data on frequency of AEs in nursing homes obtained in this study are in accordance with data published<br />

in the literature (3). A prospective, cluster-randomized trial will be performed in two different regions of Germany to<br />

demonstrate the effectiveness of the intervention with reduction of AEs as primary outcome variable.<br />

The study was funded by the German Ministry of Health, grant no.: 1501/54401.<br />

References:<br />

1. Beers MH, Ouslander JG, Rollingher I, Reuben DB, Brooks J, Beck JC., Arch Intern Med 1991, 151: 1825–32.<br />

2. Holt S, Schmiedl S, Thürmann PA., Dtsch Arztebl In. 2010, 107: 543–51.<br />

3. Gurwitz JH, Field TS, Judge J et al., Am J Med 2005, 118: 251–258.<br />

88 Sessionvorträge Tabletten


Identification of predictors for drug related problems in the context of home medication review<br />

Fiß T1 ; Hoffmann W2 1 German Center for neurodegenerative diseases (DZNE), site Rostock/<strong>Greifswald</strong>; Ellernholzstreet 1/2; 17487 <strong>Greifswald</strong>, Germany<br />

2 University <strong>Greifswald</strong>, Institute for Community Medicine; Ellernholzstreet 1/2 , 17487 <strong>Greifswald</strong><br />

Introduction: Drug related problems (DRP) are affecting success of pharmacotherapy, cause drug-induced<br />

hospitalisations high costs, and limit health related quality of life. Identification of DRP and their predictors is needed<br />

for focussed intervention and implementation of innovative health supply concepts. Several data sources are<br />

available for identification of predictors for DRP. Especially routine data from the statutory health insurances have a<br />

very strong role in this field. However, these data are limited due to incomplete data. Routine data only reflect health<br />

care services which are paid by the statutory health insurance. Clinical tests as plasma level of several vital<br />

parameter or cognitive tests are not available. Hence there is a need for drug utilisation studies in the context of<br />

health services research.<br />

Methods: We used datasets from two studies (AGnES: Arzt-entlastende, Gemeinde-nahe, E-Health-gestützte,<br />

systemische Intervention; IDemUck: inegriertes Demenznetzwerk Uckermark) to evaluate determinants for drug<br />

related problems and estimated needs for a structured medication management. Patients received a structured<br />

medication review1 in their households and subsequent intervention. The pharmaceutical care was delegated in the<br />

AGnES-studies to the local pharmacy whereas the patient’s GP conducted intervention in IdemUck.<br />

Results: We found a high proportion of drug related problems. Especially the intake of potentially inappropriate<br />

medication as well as intake of anticholinergig drugs was found as problematic. Out of the AGnES-patients aged > 65<br />

years a total of 18% (n= 134) of the patients received 163 inappropriate drugs according to the Beers’ criteria. The<br />

intake of PIM was slightly associated with self-reported falls (Phi: 0.1074; P= 0.0244). Multivariate logistic regression<br />

showed significant results for the number of taken substances (OR = 1.176; 95% CI 1.121-1.234, P< 0.001).<br />

Additionally we found the intake of anticholinergics (according to the ACBS list) as determinant for impaired cognition<br />

(OR 10; 95-CI 1.2-85.3). As a 2nd problem we identified the occurrence of potentially clinically relevant drug-drug<br />

interaction. In n=454 patients (58.3%) we found at least one drug-drug interaction with serious or moderate<br />

relevance. Adjusted for age and gender, multiple binary logistic regression showed significant results for the number<br />

of taken active substances (OR 1.48; 95%-CI 1.38-1.58), metabolic diseases (OR 1.52; 95%-CI 1.04-2.2), or a<br />

diagnose of the muscular-skeletal disease (OR 1.74; 95%-CI 1.2-2.5).<br />

Due to the pharmacist’s intervention in the AGnES-studies we found significantly improved DRP (adherence, drugdrug<br />

interaction). In contrast to these results we found an increased prevalence of potentially clinically relevant drugdrug<br />

interaction from 42.1% to 47.4% in the IDemUck-study.<br />

Discussion: In the context of home medication reviews we identified a large array of DRP. Due to several additional<br />

tests we could find some associations to clinical outcomes as falls and impaired cognition. Overall the number of<br />

taken drugs was the main risk factor for the occurrence of DRP. Subsequent intervention by a pharmacist is<br />

necessary to improve DRP.<br />

However data are limited due to small number of patients and short follow-up period. We could only find first clues for<br />

determinants which must be evidenced in further analysis. Hence linkage with routine data could help for long term<br />

follow-up. Clinical outcomes as hospitalisation and mortality could be measured with these data.<br />

As one model of evaluate efficacy of tailored intervention including long term effects we have included medication<br />

management into the study DelpHi-MV (dementia: life and person centred help in Mecklenburg Western Pomerania,<br />

clinicaltrials.gov identifier NCT01401582). Results of the analysis of determinants for the occurrence of DRP were<br />

integrated into the study algorithms.<br />

References:<br />

1. Fiss T, Ritter CA, van den Berg N et al. Pa Wo Sci 2010, 32(5):566-74.<br />

Sessionvorträge Tabletten 89


Session Arzneimittelüberwachung/Regulatory Affairs<br />

Analyzing the use of (Q)SAR models for the evaluation of potential genotoxic impurities: A<br />

cookbook in support of ICH M7<br />

Sutter A<br />

Bayer AG<br />

• Assessing the (Q)SAR systems used<br />

• Defining what exactly is expert knowledge<br />

• Evaluating current practice in pharma companies: PhRMA and EFPIA surveys<br />

• Discussing (Q)SAR reporting<br />

Control strategy for an active coating process based on systematic process development (DoE) and<br />

in-line Raman spectroscopy (PAT)<br />

Funke A<br />

Bayer Pharma AG, Global Pharmaceutical Development, Müllerstraße 178, 13353 Berlin, Germany<br />

Introducing active ingredients into pharmaceutical dosage forms via film coating onto tablets (active coating) is<br />

unusual and challenging due to high quality requirements with regard to both intra-tablet coating uniformity and<br />

accuracy of average coating amount per tablet. The variability has been reduced by systematic process development<br />

using design of Experiments (DoE). A process control strategy has been elaborated using in-line Raman<br />

spectroscopy as an IPC for the endpoint determination of the coating process (PAT). As a result, the active coating<br />

process reproducibly leads to a drug product fulfilling the predefined quality requirements.<br />

90 Sessionvorträge Tabletten


Human Tissues and Cells – Advanced Therapies Medicinal Products<br />

Astner I1, 2<br />

1 GAA, Petzvalstr. 18, 38104 Braunschweig, Germany.<br />

2 ZLG – EFG 04, Sebastianstr. 189, 53115 Bonn, Germany.<br />

Tissues and Cells for transplantation offer a wide and expanding field of medicinal products for treating a variety of<br />

diseases in humans. Alike the better known organ transplants tissues and cells are exchanged worldwide to<br />

overcome transplant shortages or to ensure better donor–recipient matching.<br />

Furthermore human or xenogenous tissues and cells are used as starting material for manufacturing (personalized)<br />

advanced therapies medicinal products (ATMPs) or Medical Devices. Gene therapy, somatic cell therapy and tissue<br />

engineering medicinal products offer new treatment opportunities for human diseases or dysfunctions like<br />

regeneration, repairing or replacement of human tissues.<br />

For tissues, cells and ATMPs it is essential to establish high standards of quality and safety for donors and patients,<br />

on the one hand and to provide exchange of these products within the (sometimes short) expiry date of the cell<br />

products, on the other hand.<br />

As human and xenogenous materials are heterogeneous similar to biologicals the quality management and the<br />

validated manufacturing process is an essential part of the product.<br />

References:<br />

1. DIRECTIVE 2004/23/EC OF THE EUROPEAN PARLIAMENT AND OF THE COUNCIL of 31 March 2004 on setting standards of quality<br />

and safety for the donation, procurement, testing, processing, preservation, storage and distribution of human tissues and cells: OJ L 102,<br />

2004: 48<br />

2. COMMISSION DIRECTIVE 2006/17/EC of 8 February 2006 implementing Directive 2004/23/EC of the European Parliament and of the<br />

Council as regards certain technical requirements for the donation, procurement and testing of human tissues and cells, OJ L 38, 2006: 38<br />

3. REGULATION (EC) No 1394/2007 OF THE EUROPEAN PARLIAMENT AND OF THE COUNCIL of 13 November 2007 on advanced<br />

therapy medicinal products and amending Directive 2001/83/EC and Regulation (EC) No 726/2004, OJ L324, 2007: 121<br />

Sessionvorträge Tabletten 91


Analysis of illegal medicinal products using NMR<br />

Schramek N<br />

Bavarian Health and Food Safety Authority, Veterinärstr. 2, Oberschleißheim, 85764, Germany<br />

The number of adulterated food supplements and illegal medicinal products to be analyzed at the Official Medicinal<br />

Control Laboratories raises steadily. More and more often unknown substances can be found as illicit additives in<br />

food supplements [1]. Fast and feasible analytical methods are required to deal with this challenge. Despite its lack of<br />

sensitivity Nuclear Magnetic Resonance (NMR) spectroscopy can be used as a fast screening method. Moreover<br />

structural information can be obtained from NMR experiments required for the identification of unknown substances<br />

[2, 3].<br />

This presentation gives a short overview on how 1- and 2-dimensional NMR spectroscopy can be useful to official<br />

medicinal control laboratories. A novel sildenafil-analogue, found as adulterant in a food supplement is presented [4].<br />

References:<br />

1. Venhuis B. J., de Kaste D.: J Pharm Biomed Anal <strong>2012</strong>, DOI:10.1016/j.jpba.<strong>2012</strong>.02.014.<br />

2. Trefi S., Gilard V., Balayssac S., Malet-Martino M., Martino R.: Magn Reson Chem 2009, 47: 163–173.<br />

3. Holzgrabe U., Malet-Martino M.: J Pharm Biomed Anal 2011, 55: 679–687.<br />

4. Wollein U., Eisenreich W., Schramek N.: J Pharm Biochem Anal 2011, 56: 705–712.<br />

92 Sessionvorträge Tabletten


Poster


001<br />

An in vitro test system to examine release in the vitreous humor<br />

Loch C1; Nagel S1; Guthoff R²; Seidlitz A1; Weitschies W1 1 University of <strong>Greifswald</strong>; Center of Drug Absorption and Transport (C_DAT), Institute of Pharmacy, Felix-<br />

Hausdorff-Strasse 3, <strong>Greifswald</strong>, 17487; Germany<br />

2 University of Rostock; Department of Ophthalmology, Doberaner Strasse 140, Rostock, 18055; Germany<br />

Introduction<br />

The vitreous body has become an interesting location for the administration of injections or<br />

implants in modern ophthalmology [1]. Such intravitreal dosage forms are loaded with<br />

active agents, which are released over a prolonged period of time. A test method simulating<br />

the situation in vivo is needed to observe the release behaviour of these new dosage forms.<br />

To simulate this situation the Vitreous Model (VM) was developed.<br />

Methods<br />

The corpus of the VM consists of glass. Size and shape are assimilated to the in vivo<br />

situation. Gel insertion and sampling can be performed via a vent at the top. To simulate<br />

the movement of the eye the VM was placed on an altered orbital shaker. The modified<br />

polyacrylamide gel (PAA-gel), which is used as vitreous substitute in this study, consists of<br />

a 30% solution of acryl amide and standard Ringer buffer solution. The viscose solution<br />

was filled in the corpus and the acryl amide monomers were cross-linked. The water<br />

content, pH, relative density, refractive index and viscosity of the PAA-gel were examined<br />

and compared experimentally to porcine and to human vitreous humor by literature<br />

analysis. Furthermore, during a period of 3 hours the distribution behaviour of fluorescein<br />

sodium solution (1.25 mM) injected into the PAA-gel and in the porcine vitreous humor was<br />

investigated by using the VM.<br />

Results<br />

The water content (99%) and pH (7.4) of the PAA-gel were equal to the values obtained for<br />

porcine and human vitreous humor. Also, the results for the relative density (1.0013) and<br />

refractive index (1.3385) showed good accordances (deviation < 1%). Slight deviations<br />

were found for the viscosity. The shape and volume of the VM correspond to the natural<br />

vitreous body.<br />

In the distribution study further agreements were achieved. For analysis the porcine<br />

vitreous body and the substitute were divided in two halves. 82% of the detected amount of<br />

fluorescein sodium was found at the side at which the injection occurred for the PAA-gel<br />

and 75% for the natural vitreous humor.<br />

Conclusion<br />

The results of the distribution study and the tested physical and chemical properties of the<br />

PAA-gel were in good accordance with the properties of porcine and human vitreous<br />

humor. Along with the related shape of the VM, the new model seems suitable to perform in<br />

vitro experiments, which are capable of simulating in vivo conditions. Through the<br />

possibility of insertion of intravitreal injections and implants in the VM, biorelevant<br />

distribution and release studies may become feasible.<br />

Acknowledgments: The authors would like to thank to the staff members of the glazier’s workshop of the<br />

Faculty of Mathematics and Natural Sciences for their excellent technical assistance. This project was<br />

funded by the Federal Ministry of Education and Research (BMBF) within REMEDIS “Höhere Lebensqualität<br />

durch neuartige Mikroimplantate” (FKZ: 03IS2081).<br />

References:<br />

1. Baino, F. et al.: Acta Biomaterialia 2011, 7(3): 921-935.<br />

002<br />

Diversified influences of arginine and guanidine on proteins<br />

Redweik S; Wätzig H<br />

Institute of Medicinal and Pharmaceutical Chemistry, TU Braunschweig, Beethovenstr. 55, 381106<br />

Braunschweig, Germany<br />

Although arginine and guanidine exhibit the same partial structure their influence on<br />

proteins is very diversified [1]. In this study these influences on the proteins BSA and ßlactoglobulin<br />

were analyzed by Affinity Capillary Electrophoresis (ACE). These influences<br />

on the proteins were also tested in the presence of urea in the Tris-buffer system because<br />

there is a concentration dependent inhibition of arginine to the urea induced protein<br />

unfolding of BSA [1].<br />

ACE is a powerful tool to detect interactions between proteins and various ligands due to<br />

mobility shifts of the protein [2]. Therefore the difference of the protein electrophoretic<br />

mobility is detected without any ligand in the buffer and with the addition of a defined ligand<br />

concentration. In order to compensate for changes on the migration time, which are not<br />

due to ligand binding, the mobility ratio of an EOF-marker and the protein is used [3]. Six<br />

repeats were done with a very good precision due to the use of a special rinsing procedure<br />

[4]. The differences of the mobility ratios Rf (without addition of ligand) and Ri (in presence<br />

of a defined ligand concentration) were normalized and the confidence intervals were<br />

estimated to demonstrate the significance of these interactions. The achieved small<br />

confidence intervals showed clearly different influences of the tested ions on BSA and ßlactoglobulin<br />

with and without urea in the buffer.<br />

References:<br />

1. Gosh, R. et al.: Biochemistry 2009, 48(5): 1135-1143.<br />

2. Busch, M. H. A. et al.: J Chromatogr A 1997, 777(2): 311-328.<br />

3. Bose, S. et al.: J Chromatogr B 1997, 697(1-2): 77-88.<br />

4. El-Hady, D. et al.: J Pharm Biomed Anal 2010, 52(2): 232–241.<br />

003<br />

The new cardioactive compounds from Leonurus cardiaca L. (Ph.Eur.)<br />

Savtschenko A1 2, Dhein S1, Rauwald H W2 1 Clinic for Cardiac Surgery, University of Leipzig, Strümpellstraße 39, D-04289 Leipzig, Germany<br />

2 Department of Pharmaceutical Biology, University of Leipzig, Johannisallee 21–23, D-04103 Leipzig,<br />

Germany.<br />

Just recently, aqueous preparations of Leonurus cardiaca – such as a newly developed<br />

refined extract – were reported to exert a series of effects on cardiac electrophysiology,<br />

such as ICa.L blockade, a reduction of the repolarising current IK.r, as well as the prolongation<br />

of both AP-duration and the activation time constant of the If current, while INa was strikingly<br />

unaffected [1]. This synergistic action on multiple electrophysiological targets may limit<br />

potential adverse effects, especially the risk of proarrhythmia, and is thus in accordance<br />

with the traditional use of this medicinal plant as an antianginal in European herbalism [2].<br />

Therefore, several extract samples were examined for substances potentially contributing<br />

to these pharmacological activities. In this screening procedure, ferulic acid (4-hydroxy-3methoxy-trans-cinnamic-acid)<br />

and stachydrine were detected as cardioactive compounds of<br />

the refined extract. Although a variety of pharmacological actions on the cardiovascular<br />

system have been reported for this caffeic acid derivative [3], specific molecular<br />

pharmacological actions on cardiac ion channels – as observed for the L. cardiaca refined<br />

extract [1] – have not been reported for the isolated compound up to now. In subsequent<br />

voltage clamp experiments carried out according to a protocol previously developed in our<br />

group [4], ferulic acid at concentration of 3 µM influences neonatal rat ventricular<br />

cardiomyocytes by ICa.L blockade and AP-duration shortening while INa was not affected.<br />

Epicardial mapping experiments on isolated rabbit hearts perfused according to the<br />

Langendorff technique showed an increase of basic cycle length and relative coronary flow,<br />

as well as a reduction of left ventricular pressure. Thus mimicking parts of the effect of the<br />

previously described L. cardiaca refined extract. In voltage clamp experiments stachydrine<br />

at concentration of 1 mM and depending on cell type increases the ICa.L current and<br />

prolongs the AP-duration of neonatal rat ventricular cardiomyocytes, while INa was<br />

unaffected. Thus, ferulic acid and stachydrine contribute to the cardiac effects of L.<br />

cardiaca; however, other compounds from L. cardiaca may also contribute according to a<br />

‘multi-component / multi-target’ active principle [1].<br />

References:<br />

1. Ritter, M. et al.: Planta Med. 2010, 76: 572-82.<br />

2. Fuchs, L.: New Kreutterbuch (Basel edit on) 1543.<br />

3. Wang, BH., Ou-Yang JP.: Cardiovascular Drug Reviews 2005, 23(2): 161-172.<br />

4. Scheffler, A. et al.: J. Ethnopharmacol. 2008, 120: 233-240.<br />

004<br />

Quantification of tumor-promoting and skin irritating diterpene esters in<br />

homeopathic mother tinctures<br />

Kummer, J., Melzig, M. F.<br />

Institute of Pharmacy - Pharmaceutical Biology, Freie <strong>Universität</strong> Berlin, Königin-Luise-Str. 2+4,<br />

14195 Berlin, Germany<br />

Some plants from the family Euphorbiaceae were used in the past as traditional remedies.<br />

With ingenol melbutate there is now a highly active therapeutic for the topical field therapy<br />

of actinic keratosis [1]. The active ingredients are diterpene esters, which are known to be<br />

tumor-promoting and skin irritating since Hecker et al. discovered TPA (12-Otetradecanoylphorbol-13-acetate)<br />

in the oil of Croton tiglium L. seeds [2].<br />

There are two basic chemical structures such as the alcohols phorbol and ingenol. These<br />

two alcohols form a great variety of different esters with different activities. Available<br />

preparations of these esters are homeopathic mother tinctures. For standardization and the<br />

monographic description in pharmacopoeias it is necessary to establish an easy method to<br />

quantify these esters from herbal preparations, e.g. homeopathic mother tinctures.<br />

To avoid the difficult analysis of the different esters and to quantify all the toxic esters it´s<br />

necessary to hydrolyse them and to determine the resulting alcohols by HPLC. The alkaline<br />

hydrolysis is not suitable for all structures because of a bad recovery [3].<br />

We will discuss the different approaches for gentle methods to hydrolyze the esters. The<br />

enzymatic hydrolysis with different enzymes such as pectinase from Aspergillus niger and<br />

esterase from pig liver and Bacillus subtilis didn t result in complete hydrolysis. Other<br />

esterases like enzymes from rabbit or rat liver seem to be more selective for diterpene<br />

esters [4].<br />

Ingenol Phorbol<br />

References:<br />

1. Lebwohl, M. et al., N Engl J Med <strong>2012</strong>, 366: 1010-1019.<br />

2. Hecker, E., Schmidt, R., Prog Chem Org Nat Prod 1974, 31: 377-467.<br />

3. Gläser, S., Untersuchungen zu einem möglichen Gesundheits- und Krebsrisiko durch pflanzliche<br />

Arzneimittel sowie industriell genutze Rohstoffe aus Euphorbiaceen – Quantitative Bestimmung von<br />

irritierenden und tumorpromovierenden Diterpenestern und Evaluierung durch biochemische und<br />

biologische Tests (Thesis, <strong>Universität</strong> Heidelberg) 1991.<br />

4. Saito, M., Egawa, K., J Biol Chem 1984, 259: 5821-5826.<br />

005<br />

Various Modes of High Performance Liquid Chromatography for Analysis of<br />

Monoclonal Antibodies<br />

Grotefend S1; Wätzig H1 1 Technische <strong>Universität</strong> Braunschweig, Institute of Medicinal and Pharmaceutical Chemistry, Beethovenstr.<br />

55, 38106 Braunschweig, Germany<br />

Protein quantitation as well as the detection of protein aggregates becomes more and more<br />

important since protein based pharmaceuticals (biologicals), especially monoclonal<br />

antibodies (mAb), were established as potent drugs in anticancer therapy or for treatment<br />

of autoimmune based diseases. Accurate and precise methods have to be found for this<br />

purpose, due to the high efficiency of these drugs and its behaviour to form aggregates,<br />

which may lead to adverse effects like anaphylaxis or injection side reactions after drug<br />

application even in small amounts [1]. Quality control purposes could not been reached by<br />

classic protein quantification methods such as the Bradford assay or SDS-PAGE, yet. In<br />

contrast different HPLC separation modes possess great selectivity as well as high<br />

precision in monoclonal antibody analysis. An IgG1-type antibody could be precisely<br />

analyzed (RSD % Monomer = 2.54 % day-to-day precision, n=60) by using High<br />

Performance Size Exclusion Chromatography (HP-SEC), which also enables the complete<br />

separation from monomer, aggregates and a low molecular weight fragment [2]. Weak<br />

Cation Exchange Chromatography (WCX), which is also already successfully applied in<br />

protein, especially antibody analysis, will be analyzed for its suitability in precise and<br />

selective monoclonal antibodies analysis. The high selectivity of this mode allows<br />

separating antibodies with different C-terminal lysine residues (proved by carboxypeptidase<br />

B addition) [3]. The separation modes will be compared in terms of precision, selectivity,<br />

analysis time as well as effort during sample and mobile phase preparation.<br />

94 Poster


References:<br />

1. Carpenter, J.F. et al, J. Pharm. Sci. 2010, 99 (5): 2200-2208.<br />

2. Grotefend, S. et al, Oral presentation on HPLC 2011 Budapest, Hungary.<br />

3. Weitzhandler, M. et al, Proteomics 2001, 1: 179-185.<br />

006<br />

Synthesis of novel Janus Kinase Inhibitors via Copper Catalyzed Azide-Alkyne<br />

Cycloaddition<br />

Gehringer, M. and Laufer, S.<br />

University of Tuebingen, Auf der Morgenstelle 8, 72076 Tuebingen, Germany<br />

Janus Kinases (JAKs) are non-receptor protein tyrosine kinases mediating signalling<br />

through the JAK-STAT (Signal Transducer and Activator of Transcription) pathway. The<br />

Janus Kinase family has four members: JAK1,2,3 and TYK2. Being crucial signal<br />

transducers for a variety of cytokines, growth factors, and interferons, JAKs are involved in<br />

numerous pathologies including malignancies, myeloproliferative disorders and<br />

autoimmune diseases.<br />

In contrast to the ubiquitous expression of the other JAK family members, JAK3 is<br />

predominantly expressed in hematopoietic cells. In mammals, the lack of functional JAK3<br />

causes immunodeficiencies while not disrupting the function of non-immune cells.<br />

Therefore, targeting JAK3 is a promising strategy to generate a novel class of<br />

immunosupressant drugs with limited side effects. [1]<br />

Recently, Ruxolitinib a small-molecule JAK1/2 inhibitor was approved by the US Food and<br />

Drug Administration for the treatment of patients with intermediate or high-risk<br />

myelofibrosis. [2]<br />

In search for novel JAK3 inhibitors we replaced the Ruxolitinib pyrazole ring by a 1,4disubstituted<br />

1,2,3-triazole accessible thought copper catalyzed azide-alkyne<br />

cycloaddition. [3] Compared to the laborious synthesis of the corresponding pyrazoles, click<br />

chemistry offers a rapid and efficient access to triazols with various substitution patterns<br />

facilitating their optimization towards JAK3 inhibition.<br />

References:<br />

[1] Cornejo , M. G. et al., Int. J. Biochem. Cell. Biol. 2009, 41, 2376-2379.<br />

[2] Mesa, R. A. et al., Nature Rev. Drug Discov. <strong>2012</strong>, 11, 103-104.<br />

[3] Sharpless, K.B. et al., Angew. Chem. Int. Ed., 2002, 41, 2596–2259.<br />

007<br />

Synthesis of water soluble aziridine-2,3-dicarboxylate-based cysteine protease<br />

inhibitors with leishmanicidal activity<br />

Fey, P. 1; Schad, C. 2; Schurigt, U. 3; Moll, H. 3; Schirmeister, T. 1<br />

1Institute of Pharmacy and Biochemistry, University of Mainz, Staudingerweg 5, 55099 Mainz,<br />

Germany<br />

2Institute of Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, 97074<br />

Wuerzburg, Germany<br />

3Institute for Molecular Infection Biology, Infection Immunology Unit, University of Wuerzburg,<br />

Josef-Schneider-Str. 2, 97080 Wuerzburg, Germany<br />

Cysteine proteases of Leishmania are proven targets for disease control of leishmaniasis,<br />

because they play a key role in parasite growth, differentiation, pathogenicity and<br />

virulence.[1]<br />

Starting from two selective protease inhibitors RV122C (Boc-(S)-Leu-(R)-Pro-(S,S)-<br />

Azi(OBn)2) and RV212C (Boc-(R)-Leu-(S)-Pro-(S,S)-Azi(OBn)2) of cathepsin L and CL-like<br />

proteases, a series of structural isomers, stereoisomers and derivatives was synthesized<br />

and tested for antileishmanial activity against L. major. [2] Out of this series CS128 (Boc-<br />

(S)-Leu-(R)-Pro-(R,R)-Azi(OBn)2) is the most promising candidate. The improvement of the<br />

water solubility of this compound is the main goal of the present work.<br />

According to docking results with RV122C in the active site of human cathepsin L[3] the<br />

Boc protecting group as well as one of the benzyl esters are not necessary for productive<br />

interactions with the enzyme. CS128, only differing in the configuration at the aziridine ring<br />

carbons, is proposed to have a comparable binding mode and therefore will be modified at<br />

the identified positions.<br />

We first focussed on the synthesis of aziridin-2,3-dicarboxylates with different ester<br />

moieties. One of these is used to yield a free acid-group after hydrogenolysis, which will<br />

increase water solubility. The other is designed to be stable against hydrogenolysis, but<br />

enabling the introduction of different liphophilic esters increasing affinity by deeply<br />

extending into the hydrophobic S2-pocket of the enzyme.<br />

Additionally, substitution of the protecting group with hydrophilic groups like glycols was<br />

chosen as alternative possibility to increase water solubility.<br />

O<br />

O<br />

HN<br />

(S)<br />

N<br />

O<br />

(R)<br />

N<br />

HO<br />

O<br />

R<br />

O O<br />

(S S) + (R R)<br />

References<br />

1. Ponte-Sucre, A. et al., Antimicrob. Agents Chemother. 2006, 50, 2439-2447.<br />

2. Schurigt U. et al., Antimicrob. Agents Chemother. 2010, 54, 5028-5041.<br />

3. Vicik R. et al., ChemMedChem 2006, 1, 1126-1141.<br />

008<br />

Synthesis, characterization and screening of Dengue Virus Type 2 NS2B-NS3<br />

protease inhibitors<br />

Holloway, S. 1; Wu, H. 1; Bodem, J. 2; Snitko, M. 2; Welker, A. 3; Sotriffer, C. 3; Klein, C. 4;<br />

Schirmeister, T. 1<br />

1 Institute of Pharmacy and Biochemistry, University of Mainz, Staudingerweg 5, D-55099 Mainz,<br />

Germany<br />

2 Institute of Virology and Immunology, University of Würzburg, Versbacher Strasse 7, D-97078<br />

Würzburg, Germany<br />

3 Institute of Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, D-97074<br />

Würzburg, Germany<br />

4 Institute of Pharmacy and Molecular Biotechnology, University of Heidelberg, Im Neuenheimer<br />

Feld 364, D-69120 Heidelberg, Germany<br />

Dengue, a mosquito-borne viral infection of humans transmitted by Aedes aegypti and<br />

Aedes albopticus mosquitoes, has become a continuing global threat, since 2.5 billion<br />

people living in tropical and subtropical regions of the world (predominantly in Asia, Africa<br />

and the Americas) are at risk of infection. [1,2] It is caused by a flavivirus with a single<br />

positive-stranded RNA genome [3,4] and four distinct serotypes. [5] The dengue virus<br />

infection can result in classic dengue fever, dengue hemorrhagic fever (DHF) or dengue<br />

shock syndrome (DSS). [6,7] The dengue virus genome encodes a serine protease with a<br />

classical catalytic triad (His51, Asp75 and Ser135) [8,9] which is responsible for the posttranslational<br />

proteolytic processing of the polyprotein precursor and essential for the viral<br />

replication [10,11], making it an important and attractive therapeutic target. [12]<br />

Our projects include syntheses, characterization and testing of DENV-2 NS2B-NS3pro inhibitors based on the structure of a) cinnamic acid amides, with the aim to analyze the<br />

influence on enzyme inhibition due to a variation of the amine residue on the one hand and<br />

the aromatic cinnamic acid residue on the other hand, and b) diaryl thioethers. The possible<br />

binding modes are analyzed by docking studies. The synthesized compounds are screened<br />

in fluorometric enzyme assays and HPLC-based assays using a fluorogenic AMC-derived<br />

substrate. Since a good specifity for the DENV-2 NS2B-NS3pro should be achieved, other<br />

serine proteases, such as trypsin, chymotrypsin and thrombin are included in our screening<br />

procedures.<br />

References<br />

1. Guzman, M. G. et al.: Nat. Rev. Microbiol. 2010, 8: S7–S16.<br />

2. Halstead, S. B.: The Lancet 2007, 370: 1644–1652.<br />

3. Li, H. et al.: J. Virol. 1998, 73(4): 3108–3116.<br />

4. Chambers, T. J. et al.: Annu. Rev. Microbiol. 1990, 44: 649–688.<br />

5. Morita, K., Tanaka, M., Igarashi, A.: J. Clin. Microbiol. 1991, 29: 2107–2110.<br />

6. Gubler, D. J.: Clin. Microbiol. Rev. 1998, 11(3): 480–496.<br />

7. Rigau-Pérez, J. G. et al.: The Lancet 1998, 352: 971–977.<br />

8. Bazan, J. F., Fletterick, R. J.: Virology 1989, 171: 637–639.<br />

9. Melino, S., Paci, M.: FEBS Journal 2007, 274: 2986–3002.<br />

10. Falgout, B. et al.: J. Virol. 1991, 65: 2467–2475.<br />

11. Zhang, L., Mohan, P. M., Padmanabhan, R.: J. Virol. 1992, 66: 7549–7554.<br />

12. Zheng, Y. et al: Bioorg. Med. Chem. Lett. 2006, 16: 36–39.<br />

009<br />

Substituted vinyl sulfones as covalent and reversible cysteine protease<br />

inhibitors<br />

Kesselring, J. 1; Schneider, T. 1; Engels, B. 2; Sotriffer, C.; 3 Schirmeister, T. 1<br />

1 Institute of Pharmacy and Biochemistry, University of Mainz, GERMANY<br />

2 Institute of Physical and Theoretical Chemistry, Universiy of Würzburg, GERMANY<br />

3 Institute of Pharmacy and Food Chemistry, University of Würzburg, GERMANY<br />

Cysteine proteases play vital roles for the life cycles, nutrition and pathogenesis of a variety<br />

of parasites causing infectious tropical diseases [1,2]. Therefore a promising strategy for<br />

the treatment of these diseases is the inhibition of these proteases.<br />

At present a large number of potent and irreversible cysteine protease inhibitors of different<br />

classes, including peptidyl vinyl sulfones, have been reported [3]. The inhibition mechanism<br />

of such vinyl sulfones results from the addition of the enzyme’s active site cysteine residue<br />

in a Michael type reaction [4].<br />

QM and QM/MM calculations have proposed substituted vinylsulfones which should be able<br />

to form a covalent, but reversible bond with the cysteine sulfur of the protease’s active site.<br />

Such a reversible reaction should be possible by choosing suitable substituents and leaving<br />

groups attached to the α- and β-position of the vinyl sulfone moiety, e.g. nitrile groups, and<br />

thiolates, respectively. This leads to a thermoneutral or slightly endergonic vinylic<br />

substitution or addition reaction. To confirm these calculations a series of highly<br />

functionalized vinyl sulfones with varying peptidic residues were synthesized and reacted<br />

with phenylethanethiol as an analogue of the active site’s cysteine residue. The progression<br />

of the reaction was monitored by NMR- and quantitative IR spectroscopy, which allows the<br />

determination of rate constants, equilibrium constants, and reaction energies. Furthermore<br />

the inhibitory potencies of the synthesized compounds were evaluated by enzyme assays<br />

with various cysteine proteases.<br />

References:<br />

[1] R.P. Hanzlik, J. Med. Chem. 1984, 27(6), 711.<br />

[2] P.J. Rosenthal and coworkers, Ant microb. Ag. Chemother. 2003, 47(1), 154.<br />

[3] J.C. Powers, J.L. Asgian, O.D. Ekici, K.E. James Chem. Rev. 2002, 12, 4639.<br />

[4] J.T. Palmer, D. Rasnick, J.L. Klaus, D. Bromme J. Med. Chem. 1995, 38 (17), 3193.<br />

010<br />

Genome-inspired drug discovery in the predatory bacterium Cupriavidus<br />

necator<br />

Nett, M. 1; Kreutzer, M. F. 1; Kage, H. 1<br />

1 Junior Research Group “Secondary Metabolism of Predatory Bacteria”, Leibniz Institute for<br />

Natural Product Research and Infection Biology / Hans-Knöll-Institute, Beutenbergstr. 11a, 07745<br />

Jena, Germany<br />

Whole genome sequencing has illuminated the secondary metabolic proficiency of<br />

Poster 95


microbes – underappreciated for years based on conventional isolation programs – and has<br />

helped set the foundation for a new natural product discovery paradigm based on genome<br />

mining [1]. Furthermore, it is becoming increasingly evident that the potential for the<br />

biosynthesis of bioactive compounds is more abundant than previously anticipated. In<br />

recent years, many neglected microorganisms have emerged as alternative resources of<br />

chemical diversity [2].<br />

Bacteria that prey on other organisms as a food source are particularly appealing in this<br />

context. We assume that a large fraction among these micropredators evolved or acquired<br />

antibiotic biosynthesis genes to promote their feeding strategy. As a proof-of-concept, we<br />

analyzed the genome of the predatory bacterium Cupriavidus necator for the presence of<br />

such genes. C. necator had previously been reported to secrete a peptidic molecule, which<br />

sequesters copper from the environment and is important for the killing of competing<br />

predacious species [3], but the structure of the involved metabolite was never resolved. We<br />

now identified a promising gene cluster that could account for the biosynthesis of this<br />

copper chelating natural product. The candidate locus includes genes for the nonribosomal<br />

assembly of a peptide as well as for the incorporation of bidentate ligand groups. To<br />

retrieve the associated natural product, we disrupted one of the biosynthesis genes by<br />

insertional mutagenesis and subsequently compared the metabolite profiles of mutant and<br />

wild-type strain. This approach led to the discovery of a family of linear lipopeptides,<br />

exemplified by the main metabolite cupriachelin [4]. When tested in an agar diffusion assay,<br />

cupriachelin and its congeners exhibited strong effects against numerous bacteria,<br />

including all identified prey organisms of C. necator.<br />

Acknowledgments: We gratefully acknowledge the Bundesministerium für Bildung und Forschung for<br />

supporting our research involving genomics and genome-inspired drug discovery (BMBF GenoMik-Transfer<br />

programme; grant no. 0315591A).<br />

References:<br />

1. Nett, M.; Ikeda, H.; Moore, B. S.: Nat. Prod. Rep. 2009, 26(11): 1362-1384.<br />

2. Winter, J. M.; Behnken, S.; Hertweck, C.: Curr. Opin. Chem. Biol. 2011, 15(1): 22-31.<br />

3. Casida, L. E.: Appl. Environ. Microbiol. 1987, 53(7): 1515-1518.<br />

4. Kreutzer, M. F.; Kage, H.; Nett, M.: J. Am. Chem. Soc. <strong>2012</strong>, 134(11): 5415-5422.<br />

011<br />

Aziridine-based inhibitors of HIV-1 protease<br />

Grün, J. 1; Rieger, M. 2; Welker, A. 3; Bodem, J. 4; Schneider, T. 1; Engels, B. 2 ; Sotriffer, C. 3;<br />

Kiefer, W. 1; Schirmeister, T. 1;<br />

1 Institute of Pharmacy and Biochemistry, University of Mainz, GERMANY<br />

2 Institute of Physical and Theoritical Chemistry, Universiy of Würzburg, GERMANY<br />

3 Institute of Pharmacy and Food Chemistry, University of Würzburg, GERMANY<br />

4 Institute of Virology and Immunology, University of Würzburg, GERMANY<br />

The HIV-1 protease represents an aspartate protease essential for proper virion assembly<br />

and maturation. [1-3] Many competitive inhibitors of this protease are available with FDA<br />

approval. [4,5] But there is a rapid rise of strains that encode mutant proteases resistant to<br />

these reversible protease inhibitors. [6,7] We explored the ability of QM/MM models to<br />

accurately describe the inhibition reaction of the HIV-1 protease by epoxide- and aziridinebased<br />

inhibitors. In contrast to their epoxide counterparts, the mechanisms and binding<br />

modes of aziridine-based inhibitors have much more rarely been the subject of<br />

investigations, e.g. no X-ray data for complexes with HIV-1 PR or SIV PR exist.<br />

Computations [8-11] predict their inhibition mechanism to be similar to that of epoxides, but<br />

differences result from the stronger basicity of the aziridine. Accordingly, aziridine-based<br />

inhibitors should be ideally suitable for aspartate proteases which act in more acidic<br />

environments. [12] This was indeed shown by recent work. Employing docking approaches<br />

the HIV-1 PR structure is used to predict possible substitution patterns of such new<br />

inhibitors with improved binding affinities. We present synthetic approaches for these new<br />

optimized aziridine-based inhibitors.<br />

References:<br />

1. Frankel, A. D.; Young, J. A. T. Annu Rev B ochem 1998, 67, 1.<br />

2. Seelmeier, S.; Schmidt, H.; Turk, V.; Vonderhelm, K. P Natl Acad Sci USA 1988, 85, 6612.<br />

3. Brik, A.; Wong, C. H. Org Biomol Chem 2003, 1, 5.<br />

4. Rose, J. R.; Craik, C. S. Am J Resp Crit Care 1994, 150, 176.<br />

5. Wlodawer, A.; Erickson, J. W. Annu Rev Biochem 1993, 62, 543.<br />

6. Metzner, K. J.; Rauch, P.; von Wyl, V.; Leemann, C.; Grube, C.; Kuster, H.; Boni, J.; Weber, R.;<br />

Gunthard, H. F. J Infect Dis 2010, 201, 1063.<br />

7. Bihani, S. C.; Das, A.; Prashar, V.; Ferrer, J. L.; Hosur, M. V. Biochem Bioph Res Co 2009, 389, 295.<br />

8. Helten, H.; Schirmeister, T.; Engels, B. J Phys Chem A 2004, 108, 7691.<br />

9. Helten, H.; Schirmeister, T.; Engels, B. J Org Chem 2005, 70, 233.<br />

10. Vicik, R.; Helten, H.; Schirmeister, T.; Engels, B. Chemmedchem 2006, 1, 1021.<br />

11. Vicik, R.; Busemann, M.; Baumann, K.; Schirmeister, T. Curr Top Med Chem 2006, 6, 331.<br />

12. Dunn, B. M. Chem Rev 2002, 102, 4431.<br />

012<br />

Bicyclic acetals – potential inhibitors of Golgi α-mannosidase II<br />

Juchum, M. 1; Kiefer, W. 1; Schneider, T. 1; Seibel, J. 2; Sotriffer, C. 2; Engels, B. 3; Schirmeister,<br />

T. 1;<br />

1 Institute of Pharmacy and Biochemistry, University of Mainz, GERMANY<br />

2 Institute of Pharmacy and Food Chemistry, University of Würzburg, GERMANY<br />

3 Institute of Physical and Theoretical Chemistry, University of Würzburg, GERMANY<br />

Cancer – predominantly trachea, bronchus and lung cancer – causes 5.9 % (3rd place in the<br />

top ten after ischemic heart disease and stroke) of deaths in high-income countries and 2.4<br />

% (7 h place) of deaths in the world [1]. Unregulated spreading of cells, invasion of healthy<br />

tissue and especially metastasis characterize the pathology of these diseases [2].<br />

Selectins, carbohydrate recognizing proteins, play a crucial role in binding metastasizing<br />

cancer cells [3]. Ligands for selectins are often modified glycosylation patterns on the<br />

surface of the cancer cells [4]. Studies have shown an overexpression of different sugar-<br />

hydrolyzing enzymes in these cells, making such enzymes promising targets for new anticancer<br />

drugs [5]. Especially inhibition of the Golgi alpha-mannosidase II (GM II) has shown<br />

tumor repression [6].<br />

GM II, a glycosyl hydrolase, is a 125 kDa type II transmembrane protein [7] that plays an<br />

essential role in the N-glycosylation pathway of asparagine side chains. The high specific<br />

cleavage of two mannose units (α-(1,3) and α-(1,3)) of the intermediate<br />

GlcNAcMan5(GlcNAc)2 takes place in the active site of the enzyme, two aspartate residues<br />

and a zinc cation involved. GM II is a retaining glycosidase and cleaves the sugars in a twostep-SN2-mechanism<br />

that preserve the configuration of the anomeric C-atom [8].<br />

Already available inhibitors, mostly derivatives of swainsonine, have side effects, due to low<br />

selectivity [9]. The goal of the presented project is the synthesis of selective, covalentreversible<br />

inhibitors with a long resting time in the catalytic site of the enzyme. QM<br />

calculations and docking simulations have shown that bicyclic acetals are promising<br />

candidates, in terms of both, high affinity to the target enzyme and reaction kinetics. Based<br />

on L-gulose, we synthesized 1-2 and 1-6 bridged species. New strategies like cycloaddition<br />

and especially olefin ring-closing metathesis (RCM) were also evaluated as new<br />

synthetic pathways to yield the desired acetals.<br />

References:<br />

1. World Health Organization. Media Centre: The top ten causes of death (Updated June 2011).<br />

http://www.who.int/mediacentre/factsheets/fs310/en/index.html (accessed March 19, <strong>2012</strong>).<br />

2. Kohn, E. C., Liotta, L. A.: Cancer Res 1995, 55: 1856-1862.<br />

3. Kim, Y. J. et al.: PNAS 1998, 95: 9325-9330.<br />

4. Kim, Y. J., Varki, A.: Glycoconjugate J. 1997, 5: 569-576.<br />

5. Rambaruth, N. D., Dwek, M. V.: Acta Histochem. 2011, 113: 591-600.<br />

6. Goss, P. E. et al.: Clin. Cancer Res, 1997, 3: 1077-1086.<br />

7. Moremen, K. W., Touster, O.: J. Biol. Chem. 1986, 261: 10945-10951.<br />

8. Petersen, L. et al.: J. Am. Chem. Soc. 2010, 132: 8291-8300.<br />

9. Kuntz, D. A. et al.: ChemBioChem 2010, 11: 673-680.<br />

013<br />

Tacrine Derivatives and its dimers as Potential Antiprotozoic Agents<br />

Schmidt I1; Schurigt U2; Pradel G2; Holzgrabe U1 1Institute of Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, 97074<br />

Würzburg, Germany<br />

2Institute for Molecular Infection Biology, University of Würzburg, Josef-Schneider-Str. 2, 97080<br />

Würzburg, Germany<br />

Three of the most important tropical infectious diseases are Malaria with an estimated 655<br />

000 deaths in 20101, Leishmaniasis with an estimated incidence of 2 million new cases per<br />

year2 and Human African Trypanosomiasis with 7139 new cases as reported in 20103. The<br />

development of new and cheap agents for the treatment is urgently needed. In our previous<br />

investigations antiprotozoal activity of tacrine (e.g. IC50 = 12.5 µM for Plasmodium<br />

falciparum) has been shown. Its homodimeric hexyl-linked derivative N1,N6-bis(1,2,3,4 tetrahydroacridin-9-yl)hexane-1,6-diamine, R1 = H, (1) was found to have increased activity<br />

(e. g. IC50 = 0.095 µM for Plasmodium falciparum). This dimer and derivatives with<br />

carbonyl- and hydroxyfunctions at the saturated ringsysteme (R1) exhibit antiprotozoal<br />

activity partially in nanomolar range of concentration, but associated with moderate<br />

cytotoxicity. Hence our first task was to identify the optimal length of the linking alkyl chain.<br />

Therefore we synthesized the above mentioned compounds with various chain length from<br />

C6 to C12. The antiprotozoal activities of this dimers against Plasmodium falciparum,<br />

Leishmania major, Trypanosoma brucei and cytotoxicity on macrophages J774.1 were<br />

determined in vitro and showed that the length of 7 to 9 methylene units is most suitable.<br />

Furthermore our results demonstrated that the introduction of substituents like nitro or<br />

chlorine in the aromatic moiety leads to a similar antiprotozoal effect and reduced<br />

cytotoxicity. Therefore our future goal will be the design and synthesis of active compounds<br />

with decreased cytotoxicity.<br />

1<br />

Acknowledgments: We acknowledge the Deutsche Forschungsgemeinschaft (SFB 630) for support.<br />

References:<br />

1. WHO; www.who.int; World Malar a report 2011.<br />

2. WHO; www.who.int; Technical Report Series 949.<br />

3. WHO; www.who.int; Fact Sheet N° 259.<br />

014<br />

Development of MIP-Inhibitors<br />

Seufert F1; Juli C1; Norville I H2; Jenner D2; Sarkar-Tyson M2; Weiwad M3; Schweimer K4; Rösch P4; Holzgrabe U1 1 Institute of Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, 97074<br />

Würzburg, Germany<br />

2 Defence Science and Technology Laboratory, Porton Down, Salisbury SP4 0JQ, UK<br />

3 Research Center for Enzymology of Protein Folding, Max-Planck Institute Halle, Weinbergweg 22,<br />

06120 Halle, Germany<br />

4 Department of Biopolymers, University of Bayreuth, <strong>Universität</strong>sstrasse 30, 95447 Bayreuth,<br />

Germany<br />

Major virulence factors known as macrophage infectivity potentiators (MIPs) facilitate the<br />

infection of cells with different pathogenic bacteria like Legionella pneumophila or<br />

Burkholderia pseudomallei. Both pathogens manifest in different diseases, L. pneumophila<br />

is the causative agent of Legionnaires‘ disease, whereas B. pseudomallei induces<br />

melioidosis which emerges in Southeast Asia and Northern Australia. An infection with<br />

B. pseudomallei may occur by inoculation through skin lesions or occasionally by<br />

inhalation. Human infection with Legionellosis transpires as well by inhalation of<br />

contaminated aerosols, typically being air-conditioning systems, whirlpools or even<br />

showers, i.e. systems with stagnant, warm water. The MIP of L. pneumophilia assists the<br />

Legionella to transmigrate epithelial cells and extracellular matrix (ECM) of lung tissue and<br />

to intrude alveolar macrophages. Both MIPs of L. pneumophila and B. pseudomallei show<br />

peptidyl propyl cis/trans isomerase (PPIase) activity [1,2]. These surface proteins belonging<br />

to the family of the FKBP binding protein form a stable complex with immunosuppressive<br />

96 Poster


drugs FK506 or rapamycin which inhibit the enzymatic function of the mentioned MIPs [3].<br />

Due to the immunosuppressive effects Legionnaires‘ disease and melioidosis can not be<br />

treated with these drugs, thus novel inhibitors had to be applied. With the aid of<br />

computational analysis, subsequent syntheses and biological screening molecules with a<br />

pipecolin acid moiety (A, B) were established. These compounds showed an activity<br />

against L. pneumophila with an IC50 of 9.0 µM for A and 32 µM for B [1]. Also, a satisfying<br />

activity against B. pseudomallei was detected for both substances. In HSQC-NMR binding<br />

studies an interaction of synthesized compounds A, B and the MIP of L. pneumophila could<br />

be verified. Furthermore, an interaction was found with substance B and the MIP of<br />

B. pseudomallei. To continue studies NMR binding experiments with compound A and the<br />

MIP of B. pseudomallei will be carried out.<br />

Acknowledgments: Financial support DFG (SFB 630)<br />

References:<br />

1. Juli, C. et al. J. Med Chem. 2011, 54(1): 277–283.<br />

2. Norville, I.H. et al. Infection and Immunity 2011, 79(11): 4299–4307.<br />

3. Fischer, G. et al. Mol. Microbiol. 1992, 6(10): 1375–1383.<br />

015<br />

Discovery of novel protease inhibitors from the Caribbean sponge Plakortis<br />

halichondrioides.<br />

Oli S, 1 Tabares P, 2 Hentschel-Humeida U, 2 Schirmeister T3 1 Institut für Pharmazie und LMC, <strong>Universität</strong> Würzburg, Am Hubland, 97074 Würzburg, Germany<br />

2 Julius-von-Sachs Institut, <strong>Universität</strong> Würzburg, Julius-von Sachs Platz 3, 97082 Würzburg,<br />

Germany<br />

3 Institut für Pharmazie und Biochemie, Johannes Gutenberg-<strong>Universität</strong> Mainz, Staudinger Weg 5,<br />

55128 Mainz, Germany<br />

Marine sponges of the family Plakinadae are known to be rich sources of structurally<br />

unique and biologically active metabolites.[1] In this study, bioactive compounds with<br />

protease inhibiting properties were discovered from the sponge Plakortis halichondrioides<br />

following bioactivity-guided fractionation. The structures of the endoperoxides plakortide E<br />

and plakortin were identified by means of MS and NMR spectroscopy. Plakortide E showed<br />

inhibitory activities against papain-like proteases such as cathepsins B and L, as well as<br />

against SARS-CoV PLpro and rhodesain. The ester plakortin was identified due to its activity<br />

against the parasite protease rhodesain from Trypanosoma brucei rhodesiense.<br />

Procedures of isolation, spectroscopic data, enzyme inhibition data as well as in vitro<br />

results are presented.<br />

Plakortide E<br />

Plakortin<br />

References:<br />

[1] Noriko Sata. et al.: J. Nat. Prod. 2005, 68, 1400-1403.<br />

016<br />

New therapy against HIV: Syntheses of inhibitors of the vif - elongin-C<br />

interaction<br />

Menrath, C. 1; Matz, F. 2; Welker A. 2; Nowotny, B. 3; Bodem, J. 3; Rethwilm, A. 3, Sotriffer, C. 2,<br />

Schirmeister, T. 1<br />

1 Institute of Pharmacy and Biochemistry, University of Mainz, Staudingerweg 5, D-55099 Mainz,<br />

Germany<br />

2 Institute of Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, D-97074<br />

Würzburg, Germany<br />

3 Institute of Virology and Immunology, University of Würzburg, Versbacher Strasse 7, D-97078<br />

Würzburg, Germany<br />

AIDS (acquired immunodeficiency syndrome) is a disease caused by HIV-1 and HIV-2<br />

(human immunodeficiency virus). In the end of 2010, about 33 millions of people were<br />

infected with HIV. 1<br />

The today’s most successful therapy is HAART (highly active anti-retroviral therapy) using<br />

combinations of drugs inhibiting essential viral enzymes thus blocking virus replication.<br />

In the human immune system, the proteins of the APOBEC3 family play essential roles in<br />

the defence against retroviral infections, in case of HIV especially APOBEC3G and 3F are<br />

involved. These proteins possess cytidine deaminase acivity thus inducing various<br />

mutations in the viral nucleic acid. Such hypermutation ultimately destroys the coding and<br />

replicative capacity of the virus, resulting in nonviable virions. However, the viral protein vif<br />

(viral infectivity factor) interacts with APOBEC3G and triggers the ubiquitination and<br />

degradation of APOBEC3G via the proteasomal pathway, thus overcoming its native<br />

antiviral activity. APOBEC3G and vif complex formation also involves binding of the human<br />

protein elongin C.<br />

On the basis of the crystal structure of elongin C, we identified new inhibitors of the vif-eloC<br />

interaction by means of virtual screening and docking studies. We present syntheses and<br />

test results of these compounds exhibiting a new mechanism of action against retroviral<br />

infections. 2 3 4 5 6<br />

References:<br />

1. Epidemie Report Update (UNAIDS 2010).<br />

2. K. N. Bishop,R. K. Holmes, A. M. Sheehy, M. H. Malim Science 2004, 305 (5684), 645.<br />

3. S. L. Sawyer, M. Emerman, H. S. Malik, PLoS. Biol. 2004, Sep; 2(9), E275.<br />

4. A. M. Sheehy, N. C. Gaddis, J. D. Choi, M. H. Malim, Nature 2002, 418 (6898), 646 – 50.<br />

5. R. S. Harris, M. T. Liddament, Nat. Rev. Immunol. 2004, 4, 868 – 877.<br />

6. M. J. Wichroski, K. Ichiyama, T. M. Rana, J. Biol. Chem. 2005, 280 (9), 8387 – 8396.<br />

017<br />

Synthesis of 1-benzoxepines as NR2B selective NMDA receptor antagonists<br />

Wendling, F. 1; Wünsch, B. 1<br />

1 Institut für Pharmazeutische und Medizinische Chemie der Westfälischen Wilhelms <strong>Universität</strong><br />

Münster, Hittorfstraße 58-62, 48149 Münster, Germany<br />

While the NMDA receptor is responsible for a number of physiological effects, including<br />

neuronal development, overactivation of NMDA receptors has been associated with<br />

pathological conditions such as Alzheimer’s Disease, Parkinson’s Disease and depression.<br />

[1]<br />

Functional NMDA receptors are composed of four subunits containing at least one NR1(ah)<br />

subunit assembled with NR2 (A-D) and/ or NR3 (A, B). [2] The NR2 subunits vary in their<br />

distribution throughout the CNS. As the NR2B subunit is less distributed in the cerebellum<br />

[3], addressing it selectively has been reported to result in decreased cognitive and motoric<br />

side effects. [4]<br />

Therefore design, synthesis and structure-affinity relationships of NR2B selective<br />

antagonists are of great interest as these compounds represent possible therapeutic agents<br />

for various neurological conditions.<br />

Using Ro-25,6981 1 as a lead compound, which binds with high affinity (Ki = 6 nM) at the<br />

NR2B subunit [5], we develop structurally related molecules with various substituents and<br />

stereochemistry.<br />

On this poster we present a Dieckmann cyclisation as key step for the synthesis of<br />

benzoxepines 2 (X=O), which we developed as conformationally restricted Ro-25,6981<br />

analogues.<br />

1 2<br />

References:<br />

1. Dingledine, R., et al.: Pharmacol. Rev. 1999, 51(1): 7-62.<br />

2. Chatterton, J.E., et al.: Nature 2002, 415(6873): 793-798.<br />

3. Gogas, K.: Curr. Opin. Pharmacol. 2006, 6 (1):68-74.<br />

4.Nikam, S.S., Meltzer, N.T.: Curr. Pharm. Design 2002, 8(10): 845-855.<br />

5. Pinard, E., et al.: Bioorgan. Med. Chem. Lett. 2001, 11(16):2173-2176.<br />

018<br />

Identification of the bioactive principles underlying the vascular barrierprotecting<br />

action of the Crataegus extract WS ® 1442<br />

Fuchs, S. 1; Willer, E. A. 1; Hartung, E. 1; Vollmar, A. M. 1; Potterat, O. 2; Hamburger M. 2; Fürst,<br />

R. 1<br />

1 Department of Pharmacy, Center for Drug Research, Pharmaceutical Biology, University of<br />

Munich, Butenandtstr. 5-13, 81377 Munich, Germany<br />

2 Department of Pharmaceutical Sciences, Pharmaceutical Biology, University of Basel,<br />

Klingelbergstr. 50, 4056 Basel, Switzerland<br />

We have recently discovered that the hawthorn (Crataegus spp.) special extract WS ® 1442,<br />

widely used for the treatment of mild forms of heart failure, effectively prevents<br />

inflammation-induced endothelial hyperpermeability in vitro and in vivo [1]. WS ® 1442<br />

affects key systems of endothelial barrier regulation, such as the F-actin cytoskeleton,<br />

adherens junctions (VE-cadherin), the contractile machinery (myosin light chain), and the<br />

intracellular calcium concentration [1]. The extract targets two major endothelial signaling<br />

cascades: it blocks the barrier-destabilizing Ca2+/PKC/RhoA pathway and activates the<br />

barrier-protecting cAMP/Rap1/Rac1 signaling [1]. In the search for the constituents<br />

responsible for these effects, 4 fractions (A-D) of WS ® 1442 were prepared (Sephadex LH-<br />

20 column chromatography, kindly provided by Dr. Willmar Schwabe GmbH & Co. KG,<br />

Karlsruhe, Germany). We could show that the two signaling cascades affected by the<br />

extract are separately targeted by two different fractions: only fraction B, containing small<br />

phenolic compounds, flavonoids, and lipophilic constituents, blocked the calcium signaling<br />

and only fraction C, consisting of oligomeric proanthocyanidins (OPCs), activated the cAMP<br />

pathway in endothelial cells.<br />

Intention of the present study was to identify the bioactive compounds by bioguided<br />

fractionation. Regarding fraction B, 9 subfractions (preparative HPLC) were analyzed for<br />

their influence on a thrombin-evoked rise of the intracellular calcium concentration in<br />

primary human endothelial cells (ratiometric, microscopic analysis using Fura-2). Only one<br />

subfraction blocked the thrombin effect and was thus judged as active. A preliminary HPLC-<br />

DAD-MS study revealed that this fraction contains lipophilic, UV inactive substances,<br />

suggesting that phenolic compounds and flavonoids are not responsible for the observed<br />

action on calcium levels. A detailed phytochemical analysis of this subfraction (isolation,<br />

structure elucidation) is in progress.<br />

Fraction C was divided into 6 subfractions (preparative HPLC). The microscopical analysis<br />

of endothelial cortactin phosphorylation, a downstream event of the cAMP-triggered<br />

signaling evoked by WS ® 1442, served as read-out parameter for bioactivity. 2 fractions<br />

were highly active, 3 exhibited a slight activity, and one was completely inactive. Further<br />

separation (Sephadex LH-20 column chromatography) of the two highly active fractions<br />

resulted in 10 subfractions, of which 8 were active. Due to this very broad activity<br />

distribution and the well-known difficulties in precisely separating OPCs, we decided to test<br />

isolated OPCs of WS ® 1442. Procyanidin B2 (an OPC dimer) and procyanidin C1 (an OPC<br />

trimer) were confirmed to trigger endothelial cAMP signaling (cAMP ELISA, phosphocortactin<br />

Western blots and microscopical analysis).<br />

In summary, we could provide first insights into the bioactive principles that underlie the<br />

polypharmacological action of WS ® 1442. The inhibition of calcium signaling seems to be<br />

evoked by lipophilic, non-phenolic compounds, which we currently aim to identify.<br />

Moreover, we could show the OPCs procyanidin B2 and C1 to be responsible for the<br />

activation of endothelial cAMP signaling by WS ® 1442.<br />

Poster 97


Acknowledgments: This work is supported by the German Research Foundation (DFG, FU691/7-1). WS ®<br />

1442 was kindly provided by Dr. Willmar Schwabe GmbH & Co. KG, Karlsruhe, Germany.<br />

References:<br />

1. Bubik, M. F. et al.: J. Mol. Cell. Cardiol. 2011, 52(1): 196-205.<br />

019<br />

HPTLC of purified hyaluronan oligosaccharides: reagent-free derivatization<br />

and densitometric quantification on amino-modified silica, TLC–ESI–Q-TOF-MS<br />

coupling on normal phase<br />

Rothenhöfer, M. 1; Scherübl, R. 2; Bernhardt, G. 1; Heilmann, J. 2; Buschauer, A. 1<br />

1 Lehrstuhl für Pharmazeutische/Medizinische Chemie II, <strong>Universität</strong> Regensburg, 93040<br />

Regensburg, Germany<br />

2 Lehrstuhl für Pharmazeutische Biologie, <strong>Universität</strong> Regensburg, 93040 Regensburg, Germany<br />

By the development of powerful high performance thin layer chromatography (HPTLC)<br />

techniques, planar chromatography has become a cost-effective and convenient alternative<br />

to high performance liquid chromatography (HPLC). It allows parallel analysis of multiple<br />

samples and, owing to a multitude of derivatization possibilities, attractive options for the<br />

detection of unfavorable analytes. For instance, oligosaccharides derived from enzymatic<br />

cleavage of hyaluronan by hyaluronidases pose a particular analytical challenge.<br />

Nevertheless, there is a strong need for purified hyaluronan fragments as pharmacological<br />

tools to shed light on the physiological and pathophysiological role of hyaluronidases,<br />

hyaluronan, and its degradation products.<br />

Therefore, we developed and validated an efficient HPTLC method for qualitative and<br />

quantitative analysis of small saturated hyaluronan oligosaccharides consisting of 2 to 4<br />

hyalobiuronic acid moieties. Uniform and reagent-free in situ derivatization is enabled by<br />

the use of amino-modified silica as stationary phase. Subsequent quantification by<br />

densitometric scans in reflectance mode revealed very low limits of detection of 7 to 19<br />

pmol per band, depending on the oligosaccharide.<br />

In view of the applicability of HPTLC for the quality control of hyaluronan oligosaccharides,<br />

proof of principle was provided for unambiguous identification of the substances by direct<br />

ESI–Q-TOF-MS coupling. For this purpose standard silica TLC plates were used and high<br />

resolution mass spectra were obtained for all three analytes.<br />

020<br />

Effects of STW 5 and STW 6 on rat ileal and colonic preparations: A<br />

comparative study<br />

Nieber, K1, Voß, U. 1, Michael, S. 2, Kelber, O. 3, H. Abdel-Abziz3 Weiser, D. 3<br />

1Institut für Pharmazie, <strong>Universität</strong> Leipzig, 04013 Leipzig,<br />

2Löwen-Apotheke-Waldheim, 04736 Waldheim,<br />

3Wissenschaftliche Abteilung, Steigerwald Arzneimittelwerk GmbH, 64295 Darmstadt<br />

STW 5 (Iberogast ) is a fixed combination of nine plant extracts with Iberis amara (STW 6)<br />

as one of its components. It is successfully used for treatment of functional dyspepsia and<br />

irritable bowel syndrome (IBS). Because clinical data suggest inflammatory involvement in<br />

the etiology of IBS the influence of STW 5 and STW 6 on tone and acetylcholine (ACh)induced<br />

contractions of intact and inflamed intestinal preparations was examined. We used<br />

1 - 1.5 cm long ileum and colon preparations of male Wistar rats to analyze region specific<br />

differences. The inflammation was induced by intraluminal instillation of 2,4,6trinitrobenzene<br />

sulfonic acid (TNBS, 10 mM, 30 min). Incubation with STW 5 (64 – 512<br />

µg/ml, 30 min) concentration dependently reduced the tone and decreased ACh-induced<br />

contractions of untreated ileal and colonic preparations. STW 6 in equivalent concentrations<br />

(3 – 24.1 µg/ml) neither affected the tone nor the contractility. TNBS-induced inflammation<br />

was accompanied by a significant reduction of ACh-induced contractions and<br />

morphological disturbances. Co-incubation of TNBS with STW 5 (512 µg/ml) or STW 6<br />

(24.1 µg/ml) partially normalized the TNBS-induced attenuation of ACh-induced<br />

contractions and the morphological damage in ileum preparations, whereas in inflamed<br />

colon segments only the co-incubation of TNBS with STW 6 in a high concentration (24.1<br />

µg/ml) revealed similar effects.<br />

In conclusion, STW 5 influenced ACh-induced contractions and tone in untreated ileal and<br />

colonic preparations, whereas STW 6 did not contribute to these effects. In TNBS-inflamed<br />

ileum preparations STW 5 as well as STW 6 normalized morphological and contractile<br />

disturbances, while in colon preparations STW 6 but not STW 5 was effective. Our study<br />

confirms region specific effects of STW 5 and its fresh plant component STW 6.<br />

021<br />

The PI3/Akt signaling pathway is involved in endothelium-dependent relaxation<br />

by Salix spp. in porcine coronary arteries<br />

Kaufeld, A. M.; Pertz, H. H.; Kolodziej, H.<br />

Institut für Pharmazie, Freie <strong>Universität</strong> Berlin, Königin-Luise-Str. 2+4, 14195 Berlin<br />

Numerous lines of evidence have shown cardioprotective and cardiotonic effects of<br />

polyphenols [1]. Preliminary studies demonstrated that 2,3-cis procyanidins of Nelia meyeri<br />

Schwant. induced an endothelium-dependent relaxation of isolated porcine coronary<br />

arteries [2]. The aim of the present study was to gain insight into the molecular mechanism<br />

by which 2,3-trans procyanidins of willow bark elicit blood vessel relaxation.<br />

The willow bark procyanidin sample comprising a mixture of dimeric to hexameric 2,3-trans<br />

flavan-3-ols [3] induced a concentration-dependent relaxation in pre-contracted<br />

endothelium-intact coronary arterial rings. The effect was inhibited by NG-nitro-L-arginine methyl ester (L-NAME; inhibitor of eNOS) and rotenone (inhibitor of mitochondrial electron<br />

transport chain). Furthermore, the extract-induced relaxation was abolished by mechanical<br />

removal of the endothelium, addition of wortmannin (inhibitor of phosphoinositide 3-kinase;<br />

PI3K), as well as by manganese(III) tetrakis(1-methyl-4-pyridyl)porphyrin (MnTMPyP; cell<br />

permeable mimetic of superoxide dismutase). Similarly, the response was completely<br />

inhibited by the combination of charybdotoxin, apamin (Ca2+-activated K + channel blockers)<br />

and L-NAME. This showed that the endothelium-derived hyperpolarizing factor (EDHF) and<br />

NO (EDRF) were involved in the relaxant response to Salix spp.. In contrast, the addition of<br />

ICI 182,780 (estrogen receptor antagonist) or pertussis toxin (inhibitor of Gi proteins) did not<br />

affect the relaxation. These findings indicated that willow bark procyanidins triggered the<br />

activation of the PI3/Akt pathway. Western Blot analysis using human umbilical vein<br />

endothelial cells (HUVEC) demonstrated that Salix procyanidins concentration-dependently<br />

induced Akt and eNOS phosphorylation. The addition of wortmannin, MnTMPyP or<br />

rotenone significantly inhibited key phosphorylation sites.<br />

In conclusion, the present data confirm procyanidin-induced production of potent<br />

vasoprotective factors including NO and EDHF. The results support the view that<br />

polyphenols have beneficial effect on endothelial cell functions of blood vessels.<br />

References:<br />

1. Rasmussen, S.E. et al.: Mol. Nutr. Food Res. 2005, 49: 159–174.<br />

2. Kaufeld, A.M., Pertz, H.H., Kolodziej, H.: Planta Med. 2011, 77: 1403.<br />

3. Kolodziej, H.: Phytochemistry 1989, 29: 955–960.<br />

022<br />

Development of NR2B-selective NMDA receptor antagonists<br />

Lütnant I. 1; Wünsch B. 1<br />

1 Institut für Pharmazeutische und Medizinsche Chemie der Westfälischen Wilhelms-<strong>Universität</strong><br />

Münster, Hittorfstraße 58-62, D-48149 Münster, Germany<br />

The N-Methyl-D-Aspartate (NMDA) receptor is an excitatoric amino acid receptor belonging<br />

to the class of ionotropic glutamate receptors. The receptor is a ligand-gated ion channel<br />

highly permeable for Ca2+-ions consisting of four subunits. Seven subunits named NR1,<br />

NR2 A-D and NR3 A, B have been identified. A functional NMDA receptor contains at least<br />

one NR1-subunit and one NR2-subunit. The receptor is distributed all over the brain, but its<br />

composition differs depending on the region in the brain in which it is expressed. [1, 2]<br />

The NMDA receptor shows unique properties such as a co-agonism of (S)-glutamate and<br />

glycine, permeability for Na +-, K +-, Ca2+-ions and a voltage-dependent Mg2+-block. When the<br />

channel is opened Ca2+-ions enter the cell and cause activation of several pathways<br />

leading to different effects amongst others neuronal development. Overstimulation of the<br />

NMDA receptor causes cell death and therefore “neurodegeneration”. [3, 4]<br />

Ifenprodil (1) is a non-competitive NMDA receptor antagonist with a high affinity towards the<br />

ifenprodil binding site on the NR2B subunit. Due to the fact that ifenprodil also binds to 5-<br />

HT1A, 5-HT2 and σ- receptor, it causes many adverse effects.<br />

To reduce the adverse side effects on the one hand and to improve the neuroprotective<br />

effects on the other hand, compounds with increased selectivity have been developed<br />

derived from the lead structure ifenprodil (1). [4]<br />

NR2B-selective NMDA receptor antagonists were found to be of value because the NR2B<br />

subunit is not expressed in the cerebellum. [5] Therefore NR2B-selective antagonists lead<br />

to reduced motoric side effects. They seem to be useful compounds considering the<br />

therapy of Parkinson´s and Alzheimer´s disease because they are capable to inhibit the<br />

effects of an overstimulation of the NMDA receptor.<br />

We develop a new group of potential NR2B-selective NMDA receptor antagonists with a<br />

benzimidazolone bioisoster (2) of the phenol of ifenprodil (1).<br />

References:<br />

1. Dingledine, R. et al.: Pharmacol. Reviews 1999, 51(1).<br />

2. Goebel, D.J., Poosch, M.S.: Mol. Brain Res. 1999 69: 164-170.<br />

3. Stark, H, Graßmann, S., Reichert, U.: Pharm. In unsrere Zeit 2000, 29(3): 159-166.<br />

4. Stark, H, Graßmann, S., Reichert, U.: Pharm. In unsrere Zeit 2000, 29(4): 228-236.<br />

5. Monyer, H. et al.: Neuron. 1994, 12: 529-540.<br />

023<br />

Chain modified oxa-analogs of primaquine and some of its derivatives<br />

Leven, M. 1; Held, J. 2; Tschan, S. 2; Delves, M. 3; Plouffe, D. 4; Mordmüller, B. 2; Kurz, T. 1<br />

1 Institut für Pharmazeutische und Medizinische Chemie, Heinrich Heine <strong>Universität</strong> Düsseldorf,<br />

<strong>Universität</strong>sstr. 1, 40225 Düsseldorf, Germany<br />

2 Institut für Tropenmedizin, Eberhard Karls <strong>Universität</strong> Tübingen, Wilhelmstr. 27, 72074 Tübingen,<br />

Germany<br />

3 Department of Life Sciences, Imperial College London, South Kensington Campus, London SW7<br />

2AZ, United Kingdom<br />

4 Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San<br />

Diego, CA 92121, United States of America<br />

The 8-aminoquinoline antimalarial primaquine 1 affects multiple life cycle stages of<br />

Plasmodium species. It exhibits activity against developing and dormant liver stages of P.<br />

vivax and P. ovale. Thus it is used for the treatment of symptomatic patients with P. vivax or<br />

P. ovale malaria and for the presumptive anti-relapse therapy in persons heavily exposed to<br />

these plasmodia. As 1 also influences developing liver stages of P. falciparum, it is used as<br />

a primary prophylactic agent. Due to its activity against gametocytes of all species of<br />

human malaria, 1 is recommended as a transmission-blocking agent in endemic regions. Its<br />

weak inhibitory activity against asexual blood stages of all Plasmodium species calls for a<br />

combination therapy with an effective blood schizontocidal antimalarial agent. Serious<br />

adverse effects including hemolysis in persons with glucose-6-phosphate dehydrogenase<br />

(G6PD) deficiency and methemoglobinemia limit the therapeutic potential of 1. Its<br />

application requires a previous test for G6PD activity. 1 is contraindicated in pregnancy and<br />

in children under the age of four years [1,2].<br />

In contrast to 1, clinical phase III drug candidate tafenoquin 2 demonstrates potent<br />

inhibitory activity against blood stages of P. falciparum and P. vivax. Preliminary data<br />

suggest that 2 is less toxic than 1, but hemolysis in persons with G6PD deficiency has also<br />

been reported.<br />

The (-)-enantiomer NPC1161B 3, currently in preclinical trials, affects effectively blood<br />

stages of P. berghei in mice and is less hematotoxic in beagle dogs than 1. In addition, 3<br />

inhibits effectively sporogony in vitro [3,4].<br />

98 Poster


In order to improve activities and to reduce side effects numerous derivatives of 1 have<br />

been prepared and biologically evaluated. An established concept for the modification of<br />

lead structures is the bioisosteric replacement of methylene groups in alkyl chains by<br />

bivalent atoms like oxygen and sulfur. In an attempt to optimize structural features of 1 we<br />

now report on the synthesis and biological evaluation of side chain modified oxa-analogs of<br />

1 and some of its derivatives.<br />

References:<br />

1. Hill, D. R. et al.: Am. J. Trop. Med. Hyg. 2006, 75(3): 402–415.<br />

2. Vale, N., Moreira, R., Gomes, P.: Eur. J. Med. Chem. 2009, 44(12): 937-953.<br />

3. Nannayakara, N. P. D. et al.: Antimicrob. Agents Chemother. 2008, 52(6): 2130-2137.<br />

4. Delves, M. et al.: PLoS Med. <strong>2012</strong>, 9(2): e1001169. doi:10.1371/journal.pmed.1001169.<br />

024<br />

Enantioselective Synthesis of Spirocyclic σ1 Ligands<br />

Holl, K. 1; Wünsch, B. 1<br />

1 Institut für Pharmazeutische und Medizinische Chemie, Westf. Wilhelms-<strong>Universität</strong> Münster,<br />

Hittorfstr. 58-62, D-48149 Münster, Germany<br />

σ receptors represent a unique class of receptors, consisting of two subtypes, σ1 and σ2<br />

receptors. They are widely distributed in the human body, including the central nervous<br />

system, as well as in many tissues of the periphery, e.g. heart, lung, liver, kidney [1]. The σ1<br />

receptor has been associated with various neuronal diseases, such as schizophrenia,<br />

depression and Alzheimer’s disease. In addition to that, an overexpression of σ1 receptors<br />

in certain tumor cells was reported [2]. Therefore, high-affinity σ1 ligands have potential for<br />

the therapy and diagnosis of CNS diseases and cancer. Fluorinated ligands can be used as<br />

PET tracers [3].<br />

A variety of spirocyclic ligands with high affinity for the σ1 receptor has been synthesized,<br />

but so far, only racemic mixtures have been characterized. We report on the first<br />

enantioselective synthesis of this kind of ligands, in order to detect relationships between<br />

the absolute configuration and the σ1 affinity.<br />

Starting from 2-bromostyrene (1), a series of spirocyclic σ1 ligands with a benzopyrane<br />

structure was synthesized, using Sharpless asymmetric dihydroxylation as key step.<br />

Furthermore, several variations in position 4 were carried out, including fluoro derivatives<br />

Br<br />

OH<br />

N<br />

R<br />

OH<br />

OH<br />

1 2 3<br />

Acknowledgments: We thank Deutsche Forschungsgesellschaft for financial support.<br />

References:<br />

1. Matsumoto, R. R. et al.: Eur. J. Pharmacol. 2003, 469(1-3): 1-12.<br />

2. Maurice, T. Su, T.: Pharmacol. Ther. 2009, 124(2): 195-206.<br />

3. van Waarde, A. et al.: Curr. Pharm. Des. 2010, 16(31): 3519-3537.<br />

025<br />

Comparative effect of proanthocyanidin enriched extracts on the in vitro<br />

inhibition of bacterial and viral neuraminidase<br />

Quosdorf, S. J.; Kolodziej, H.<br />

Institut für Pharmazie, Freie <strong>Universität</strong> Berlin, Königin-Luise-Str. 2+4, 14195 Berlin<br />

Influenza infections are still a major threat to human health and emergence of resistance to<br />

anti-influenza drugs is a cause for concern. Expanding the range of antiviral drugs that<br />

effectively inhibit influenza viruses is therefore a matter of urgency. Neuraminidase (NA), a<br />

key enzyme in the influenza life cycle and pathogenesis, has been established as a primary<br />

drug target. Accordingly, NA inhibition assays represent a promising strategy for finding<br />

new antiviral compounds. However, the substrates used in such functional assays are<br />

recognized by both bacterial and viral NAs [1]. Due to limited comparative data, IC50 values<br />

obtained from either setup must be regarded with caution. The current study was carried<br />

out to examine the validity and significance of the origin of NAs in a functional NA inhibition<br />

test system.<br />

Using an established fluorescence based in vitro assay, the inhibitory potential of a series<br />

of polyphenolic samples (Pelargonium sidoides DC, Salix spp, Nelia meyeri Schwant. and<br />

Diospyros kaki L.) was measured for bacterial (Vibrio cholerae) and viral NA (Baculovirus).<br />

Zanamivir (Relenza ®) and oseltamivir carboxylic acid served as positive controls. Selection<br />

of the test substances is based on preliminary studies regarding prominent in vitro inhibition<br />

of bacterial NA and differences in their proanthocyanidin patterns [2]. In this assay the<br />

substrate 4-methylumbelliferyl-N-acetyl-α-D-neuraminic acid (MUNANA) is cleaved by NA<br />

to produce the fluorescent compound 4-methylumbelliferone (MU) [3]. The quantity of MU is<br />

inversely proportional to the inhibitory activity of the tested sample.<br />

The results demonstrate that the inhibitory potential of the proanthocyanidin enriched<br />

extracts differ significantly and depend strongly on the origin of NA under the same<br />

experimental conditions. All extracts were more effective against Vibrio cholerae NA (IC50<br />

ca. 1 µg/mL – ca. 19 µg/mL) compared to Baculovirus NA (IC50 ca. 19 µg/mL – ca. 58<br />

µg/mL). Interestingly and in contrast, zanamivir and oseltamivir carboxylic acid exhibited<br />

much stronger inhibitory activities against the viral NA (IC50 ca. 4 µg/mL and ca. 1 µg/mL,<br />

respectively) than against the bacterial NA (IC50 ca. 16 µg/mL and ca. 40 µg/mL,<br />

respectively). Therefore it appears reasonable to select preferably viral NAs when testing<br />

samples for anti-influenza potentials or searching for promising antiviral compounds. On the<br />

OR<br />

N<br />

Bn<br />

O<br />

other hand, the test system should be adapted to a given pathogenic condition that may<br />

well include bacterial NAs. The observed difference in susceptibility of the NAs is an<br />

important parameter affecting generated results in the applied test system.<br />

References:<br />

1. Grienke, U., et al.: Nat. Prod. Rep. <strong>2012</strong>, 29(1): 11–36.<br />

2. Janecki, A., Kiderlen, A.F., Kolodziej, H.: Planta Med. 2009, 75(9): 989.<br />

3. Potier, M., et al.: Analytical Biochemistry. 1979, 94(2): 287–296.<br />

026<br />

Spirocyclic isochromane-piperidines as new lead structures against<br />

atherosclerosis<br />

Strunz, A. K. 1; Wünsch, B. 1<br />

1Institut für Pharmazeutische und Medizinische Chemie der Westfälischen Wilhelms-<strong>Universität</strong><br />

Münster, Hittorfstraße 58-62, D-48149 Münster, Germany<br />

The chemokine receptor 2 (CCR2) is a G-protein coupled receptor (GPCR) and part of the<br />

chemokine system, a complex network consisting of soluble proteins and their receptors.<br />

Chemokines and their receptors mediate effects on the cellular signalling pathway. The<br />

CCR2 receptor is expressed on various immune cells and regulates cellular movement and<br />

activation.<br />

The interaction of the endogenous ligand MCP-1(monocyte chemoattractant protein 1) with<br />

the CCR2 receptor plays a crucial role in several inflammatory processes [1]. In the chronic<br />

inflammatory disease of atherosclerosis, this interaction leads to the mobilization of<br />

activated monocytes into the subendothelial tissue [2].<br />

On one hand CCR2 antagonists can stop the growth and progression of atherosclerotic<br />

plaques by inhibition of early key steps, on the other hand [ 18F]-labeled CCR2 antagonists<br />

can be applied in PET (positron emission tomography) studies to analyze atherosclerotic<br />

plaques.<br />

The synthesis of new CCR2 antagonists with a spirocyclic substructure will be presented on<br />

this poster. The main building block is a spirocyclic system which consists of an<br />

isochromane (A) with a piperidine ring (B), prepared by Oxa-Pictet-Spengler reaction [3].<br />

The cyclization is performed with various phenylethanol derivatives. Different benzylamines<br />

(D) lead to different test compounds. The introduction of an aromatic substituent R5 might<br />

lead to compounds with increased CCR2 affinity [4]. The four variable building blocks (A-D)<br />

can be combined in different ways.<br />

References:<br />

1. Gerard, C., Rollins, B. J.: Nat. Immunol. 2001, 2 (2): 108-115.<br />

2. Galkina, E., Ley, K.: Annu. Rev. Immunol. 2009, 27: 165-197.<br />

3. Larghi, E. L., Kaufman, T. S.: European Journal of Organic Chemistry 2011, 2011 (27): 5195-5231.<br />

4. Butora, G. et al.: Bioorg. Med. Chem. Lett. 2006, 16 (18): 4715-4722.<br />

027<br />

Reverse α-aryl substituted β-thia-analogs of fosmidomycin as 1-deoxy-Dxylulose<br />

5-phosphate reductoisomerase (Dxr) inhibitors<br />

Lienau, C. 1, Kunfermann, A. 2, Hähn, S. 1, Konzuch, S. 1, Behrendt, C. T. 1, Illarionov, B. 3,<br />

Held, J. 4, Mordmüller, B. 4, Fischer, M. 3, Groll, M. 2, Kurz, T. 1<br />

1 Institut für Pharmazeutische und Medizinische Chemie, Heinrich Heine <strong>Universität</strong> Düsseldorf,<br />

<strong>Universität</strong>sstr. 1, 40225 Düsseldorf, Germany<br />

2 Center for Integrated Protein Science Munich, Lehrstuhl für Biochemie, Technische <strong>Universität</strong><br />

München, Lichtenbergstr. 4, 85747 Garching, Germany<br />

3 Hamburg School of Food Science, <strong>Universität</strong> Hamburg, Grindelallee 117, 20146 Hamburg,<br />

Germany<br />

4 Institut für Tropenmedizin, Eberhard Karls <strong>Universität</strong> Tübingen, Wilhelmstr. 27, 72074 Tübingen,<br />

Germany<br />

Inhibition of enzymes of the non-mevalonate pathway (MEP pathway) is a promising<br />

concept for the development of novel anti-infective drugs [1,2]. The non-mevalonate<br />

pathway of isoprenoid biosynthesis is the only source of the isoprenoid precursors<br />

isopentenyl diphosphate (IPP) and dimethylallyl diphosphate (DMAP) in several pathogenic<br />

bacteria and apicomplexan protozoa including the causative agents of malaria, Plasmodium<br />

spp.. The MEP pathway is absent in mammalians but essential for various pathogenic<br />

bacteria and Plasmodium spp.. A validated target for the development of novel<br />

antimalarials within the MEP pathway is the 1-deoxy-D-xylulose 5-phosphate<br />

reductoisomerase (Dxr) [3,4]. Reverse α-aryl substituted carba- and oxa-analogs (1,2) of<br />

the natural product fosmidomycin are highly active Dxr inhibitors and show potent<br />

antiplasmodial activity [3,4]. Further structural modifications of analogs (1,2) led to<br />

previously unreported reverse α-aryl substituted β-thia-isosters of fosmidomycin.<br />

References:<br />

1. Jomaa, H. et al.: Science 1999, 285: 1573-1576.<br />

2. Rohmer, M. et al.: Curr. Opin. Investig. Drugs 2004, 5: 154-162.<br />

3. Behrendt, C. T. et al.: J. Med. Chem. 2011, 54: 6796-6802.<br />

Poster 99


4. Behrendt, C. T. et al.: ChemMedChem 2010, 5(10):1673-1676.<br />

028<br />

Influence of the indirubin derivative 6BIO on proliferation and metastasis of<br />

breast cancer cells<br />

Braig S. 1, Kressirer C. 1, Bischoff F. 1, Meijer L. 2, Zahler S. 1, Vollmar A.M. 1<br />

1Department of Pharmacy, Center for Drug Research, Butenandtstr.5-13, 81377 Munich, Germany<br />

2CNRS, Station Biologique, Roscoff, France<br />

Initially, indirubins were discovered as the active component of a well known traditional<br />

Chinese medicine. Since then, it was shown that natural indirubins and also its synthetic,<br />

cell permeable derivative 6BIO (6-bromo-indirubin-3-oxime) display a remarkable potential<br />

to inhibit glycogen synthase kinase 3 (GSK-3) and cyclin-dependent kinases (CDKs) by<br />

interacting with the ATP-binding pocket of these kinases.<br />

By an inverse virtual screening approach phosphoinositide-dependent kinase 1 (PDK1) was<br />

proposed by us as a yet unknown target of 6BIO. Western blot analysis and confocal<br />

microscopy revealed that treatment of breast cancer cells with 6BIO resulted in a reduced<br />

phosphorylation of AKT, an important downstream target of PDK1. Since about 70% of all<br />

breast cancer carcinomas express moderate to high levels of activated PDK1, we<br />

investigated the impact of 6BIO treatment on different breast cancer cell lines by analyzing<br />

the anti-proliferative and cytotoxic effects as well as the influence on reduction of metastatic<br />

capabilities of these cells.<br />

As determined by crystal violet staining and xCELLigence experiments, incubation of<br />

SkBr3, MDA-MB-231, MCF7 and AKT overexpressing SkBr3 breast cancer cell lines with<br />

increasing concentrations of 6BIO led to a dose-dependent inhibition of proliferation. In<br />

addition, 6BIO treated breast cancer cells showed a strongly diminished long-term<br />

clonogenic survival rate.<br />

Furthermore, Nicoletti assays and Hoechst staining indicated that stimulation of breast<br />

cancer cell lines with high doses of 6BIO led to an induction of apoptosis.<br />

Cells treated with sub-toxic doses of 6BIO displayed a strongly reduced attachment<br />

capacity, as shown by adhesion assays on fibronectin and xCELLigence experiments.<br />

Scratch assays as well as chemotaxis assays revealed a significantly diminished migratory<br />

potential of 6BIO treated breast cancer cells compared to control cells. Furthermore, 6BIO<br />

significantly disrupted the invasion capacity of the cells. The impact of 6BIO treatment on<br />

the migration and invasion inhibition might be due to altered localization of the well-known<br />

migration associated molecules FAK and Rac1 within the cells, as shown by confocal<br />

microscopy.<br />

In summary, we were able to show that 6BIO inhibits PDK1 signal transduction in different<br />

breast cancer cell lines and reduces proliferation, clonogenic survival, attachment,<br />

migration and invasion of the cells. Thus, the indirubin derivative 6BIO might serve as a<br />

highly promising potential therapeutic agent for metastatic breast cancer cells.<br />

029<br />

Discovery of the first antagonists of PqsR to interrupt Pseudomonas<br />

aeruginosa cell-to-cell communication<br />

Steinbach, A. 1; Lu C1; Kirsch, B. 1; Zimmer, C. 1; de Jong, J.C. 1; Henn, C. 1; Maurer, C.K. 1;<br />

Müsken, M. 2; Häussler, S. 2 3; Hartmann, R.W. 1<br />

1 Helmholtz-Institute for Pharmaceutical Research Saarland, Campus C2.3, 66123 Saarbrücken,<br />

Germany<br />

2 Twincore, Department of Pathophysiology of Bacterial Biofilms, Feodor-Lynen-Str. 7, 30625<br />

Hannover, Germany<br />

3 Helmholtz Centre for Infection Research, Chronic Pseudomonas Infection Research Group,<br />

Inhoffenstr. 7, 38124 Braunschweig, Germany<br />

P. aeruginosa coordinates group behaviors such as virulence factor expression and biofilm<br />

formation via a cell-to-cell communication system known as quorum sensing (QS). The<br />

transcription of target genes are regulated via a pqs QS system that functions via the signal<br />

molecules PQS and its precursor HHQ that interact with their receptor PqsR. Targeting<br />

PqsR to develop quorum sensing inhibitors is considered as novel strategy to attenuate P.<br />

aeruginosa pathogenicity [1]. In order to discover PqsR antagonists, a ligand-based drug<br />

design approach was followed to synthesize a set of HHQ and PQS derived compounds.<br />

HHQ (R = H) R 1 = ED or EW groups<br />

PQS (R = OH) R 2 = alkyl or alkylether<br />

Agonistic or antagonistic properties were determined in a heterologous β-galactosidase<br />

reporter gene assay in E. coli to examine effects of side chain modifications and<br />

substitutions at the benzene moiety. An n-heptyl chain in 2-position was found to be<br />

optimal. Most importantly, the introduction of strong electron withdrawing functions like CN,<br />

NO2 or CF3 in 6-position of HHQ led to the discovery of the first antagonists (IC50 values of<br />

259 nM, 51 nM and 54 nM), while HHQ analogues with the same substituents in 7- or 8position<br />

or other substituents and all PQS derivatives were moderate to weak agonists.<br />

Direct evidence for the binding of a selected antagonist (6-CN HHQ) to PqsR was provided<br />

by surface plasmon resonance (SPR) biosensor experiments (KD=57 nM). Moreover, 6-CF3<br />

HHQ reduced pyocyanin production by 74 % (3 µM) in the target organism P. aeruginosa<br />

[2]. The aqueous solubility of the antagonists could be improved by the introduction of O<br />

into the alkyl side chain or CONH2 into 3-position.<br />

The first antagonists of PqsR are highly valuable scientific tools for in-depth study of<br />

protein-ligand interactions and provide a promising starting point for the development of a<br />

new anti-infective strategy.<br />

References:<br />

Deziel E et al., Mol. Microbiol. 2005, 55: 998–1014.<br />

Lu, C. et al.; Chem. Biol. <strong>2012</strong>, 19: 381–390.<br />

030<br />

Diarylpyrrolizines as Inhibitors of Microsomal Prostaglandin E2 Synthase-1<br />

(mPGES-1) or as Dual Inhibitors of mPGES-1 and 5-Lipoxygenase (5-LO)<br />

Keck, P. R. W. E. F. 1; Liedtke, A. J. 2; Koeberle, A. 3; Werz, O. 3; Schollmeyer, D. 4; Laufer, S.<br />

A. 1<br />

1 Department of Pharmaceutical and Medicinal Chemistry, Pharmaceutical Institute, University of<br />

Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, Germany<br />

2 Pharmaceutical Institute, Pharmaceutical Chemistry I, University of Bonn, An der Immenburg 4,<br />

53121 Bonn, Germany<br />

3 Department of Pharmaceutical and Medicinal Chemistry, Pharmaceutical Institute, University of<br />

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

4 Institute Organic Chemistry, University Mainz, Duesbergweg 10-14, 55099 Mainz, Germany<br />

Prostaglandin E2 (PGE2) is one of the most abundant and powerful prostanoids with diverse<br />

biological activities. As a key cyclic lipid mediator derived by the cyclooxygenase (COX)<br />

from arachidonic acid (AA), it is involved in the development and perpetuation of<br />

inflammation seen in diseases such as rheumatoid arthritis (RA). PGE2 has been implicated<br />

in the development of peripheral and central sensitization during nociceptive processing<br />

(e.g., hyperalgesia, allodynia) and in tumorigenesis[1]. Nonsteroidal anti-inflammatory<br />

drugs (NSAIDs), including selective COX-2 inhibitors, have analgesic, antipyretic and antiinflammatory<br />

properties. The main mechanism of action of NSAIDs is the inhibition of the<br />

COX isoenzymes. However, inhibition of the COX pathway by NSAIDs also reduces gastro<br />

protective PG synthesis, resulting in the well-known gastrointestinal side effects. In<br />

addition, the resulting increase in leukotriene (LT) biosynthesis due to shunting of AA to the<br />

5-lipoxygenase (5LO) pathway activated by the 5-lipoxygenase activating protein (FLAP)<br />

causes additional gastrointestinal injury[2,3].<br />

A dual COX/FLAP inhibition by Licofelone (ML3000) is an approach to minimize the side<br />

effects[2]. Licofelone also shows an inhibitory activity on the mPGES-1[4]. Going<br />

downstream, we synthesized and evaluated inhibitors for the mPGES-1, based on the<br />

diarylpyrrolizine scaffold of Licofelone while conserving the activity on the FLAP respective<br />

5-LO[1].<br />

Additionally, two crystal structure have been obtained to determine the angle of the aryl<br />

substituents[5,6].<br />

References:<br />

1. Liedtke, A. J. et al. J. Med. Chem. 2009, 52(15): 4968-4972.<br />

2. Laufer, S. A. et al. J. Med. Chem. 1994, 37(12): 1894-1897.<br />

3. Laufer, S. A.; Gay, S.; Brune, K.: Inflammation and Rheumatic Diseases (Thieme) 2003.<br />

4. Koeberle, A. et al. J. Pharmacol. Exp. Ther. 2008. 326(3): 975-982.<br />

5. Keck, P. R. W. E. F.; Schollmeyer, D.; Laufer, S. Acta. Cryst. E 2011, E67(9): 2292.<br />

6. Keck, P. R. W. E. F.; Schollmeyer, D.; Laufer, S. Acta. Cryst. E 2011, E67(9): 2417.<br />

031<br />

The actin cytoskeleton as potential antitumor target: Chondramide and its<br />

effects on breast cancer cells<br />

Förster F1; von Schwarzenberg K; Müller R2; Vollmar A<br />

1 Department of Pharmacy - Center for Drug Research, Pharmaceutical Biology, LMU Munich,<br />

Butenandtstaße 5-13, 81377, München<br />

2 Institut für Pharmazeutische Biotechnologie, <strong>Universität</strong> des Saarlandes, Postfach 15 11 50,<br />

66041, Saarbrücken”<br />

The actin cytoskeleton is a crucial component to maintain cellular homeostasis. It is<br />

important in processes like muscle contraction, trafficking of cellular compartments, mitosis,<br />

invasion and migration. Thus, actin-binding compounds like Chondramide A (Chon A), a<br />

cyclic depsipeptide of myxobacterial origin could be developed as potential anticancer<br />

therapeutics. Aim of this study was to characterize antitumoral effects of Chon A in<br />

mammary cancer cells in vitro.<br />

In fact we could show that Chon A inhibits proliferation of two breast tumor cell lines MCF-7<br />

and MDA-MB-231 in a low nanomolar range. The long term survival of these cell lines was<br />

impaired by Chon A as the clonogenic survival was also decreased by about 60 % (MDA-<br />

MB-231) and 80 % (MCF-7), respectively. To proof if the observed effects are due to actin<br />

overpolymerization, the Triton-X 100 soluble fraction was analyzed for its actin content and<br />

showed a time (24h – 72h) and dose dependent decrease, which suggested that Chon A<br />

overpolymerized actin to its filamentous form that is not soluble in the Triton-X-100 fraction.<br />

Clarifying the mechanisms underlying the antiproliferative effects of Chon A, we examined<br />

whether Chon A induces apoptosis and/or premature cellular senescence. Annexin V/PI<br />

costaining revealed a concentration dependent apoptotic cell death induced by 500 nM<br />

Chon A in both cell lines. Cleavage of PARP, a known substrate of caspase-3, detected by<br />

western blotting, supported the induction of apoptosis by Chon A, too. The induction of<br />

cellular senescence was cell line specific, as MCF-7 showed an increased β-galactosidase<br />

activity, compared to MDA-MB-231 which did not display this hallmark of senescence. The<br />

cdk-inhibitor p21, which is important for senescence induction, was also increased with<br />

Chon A 500 nM treatment in MCF-7 cells. These results suggest, that the inhibition of<br />

growth in MDA-MB-231 is only due to apoptosis induction, whereas in MCF-7 cells both<br />

processes apoptosis and senescence are involved. Furthermore, phosphorylation of<br />

histone γH2A.X, an established sign for DNA damage, treated with Chon A was observed<br />

in both cell lines. To this end, Chon A induces pronounced cell cycle arrest in G2-phase (40<br />

% versus 20 % in untreated cells).<br />

100 Poster


Our investigations reveal that Chon A is a potent inhibitor of breast cancer cell proliferation<br />

via induction of apoptosis and premature cellular senescence. Induction of senescence has<br />

not yet been reported for actin – binding drugs. Thus, further experiments will focus on the<br />

mechanism underlying senescence induction. One major tool will be PCR and micro-array<br />

technologies to possibly find new and valuable players and targets in senescence and<br />

growth arrest signaling.<br />

Acknowledgments: Supported by the DFG: FOR 1406 Vo 376/15-1<br />

032<br />

Phenylamino-substituted 5,11-dihydrodibenzo[a,d]cyclohepten-10-ones and<br />

11H-dibenzo[b,f]-oxepin-10-ones as novel p38 MAP kinase inhibitors<br />

Dorn, A. 1; Martz, K. E. 1; Laufer, S. A. 1<br />

1 Department of Pharmaceutical and Medicinal Chemistry, Eberhard-Karls-<strong>Universität</strong>, Auf der<br />

Morgenstelle 8, 72076 Tübingen<br />

The p38 MAP kinase is a key player in signalling pathways regulating the biosynthesis of<br />

pro-inflammatory cytokines like TNF-α and IL-1β. Small molecule p38 inhibitors suppress<br />

the production of these cytokines making p38 a promising drug target for novel antiinflammatory<br />

drugs.<br />

We recently reported a novel series of dibenzepinone inhibitors belonging to the class of so<br />

called linear binders [1]. Upon binding of the dibenzepinone inhibitor, the p38 MAP kinase<br />

undergoes a Gly110 flip. The glycine flip is a small conformational rearrangement in the<br />

hinge region of the p38 MAP kinase induced by the inhibitor and it provides selectivity for<br />

p38 over other kinases with less flexible, non-glycine residues at this position. Hence the<br />

carbonyl functionality of the dibenzepinone inhibitor is essential for the inhibitory activity<br />

and selectivity as it forms two hydrogen bonds towards Met109 and Gly110 in the hinge<br />

region.<br />

In this study, we report the design, synthesis, and SAR of novel N-substituted 11Hdibenzo[b,f]oxepin-10-ones<br />

and 5,11-dihydro-dibenzo[a,d]cyclohepten-10-ones as p38α<br />

inhibitors. The main aim was to conserve the key interaction: the bidentate hydrogen bond<br />

of the carbonyl oxygen of the inhibitor to the backbone NH of Met109 and the backbone NH<br />

of Gly110. Docking studies predicted alternative positions for the carbonyl oxygen within<br />

the scaffold of the inhibitor. Within these inhibitor structures, the carbonyl functionality was<br />

moved on the bridge side and the linker atoms X and Y were varied to obtain distinctive<br />

molecular geometries and additional interaction opportunities.<br />

Initial investigations of the inhibitory activities and structure-activity-relationships of<br />

phenylamino-substituted dibenzo[b,f]oxepin-10(11H)-one, 5,11-dihydro-(10H)-dibenzo[a,d]cyclohepten-10-one<br />

and dibenzo[b,f][1,4] oxazepin-11(10H)-one inhibitors for p38 MAP<br />

kinase were accomplished. The promising structural variations suggested by our docking<br />

experiments did not result in any novel compounds as active as the lead compound. While<br />

our initial hypothesis that substitution of a tricyclic scaffold would result in a favorable<br />

position for the carbonyl functionality was not supported by our data, we did identify some<br />

structural determinants that may be useful for the development of future p38 MAPK<br />

inhibitors.<br />

References:<br />

1. Laufer, S. A. et al.: J. Med. Chem. 2006, 49: 7912-7915.<br />

033<br />

Long-Range Intramolecular S → N Acyl Migration: A Study of the Formation of<br />

Native Peptide Analogues via 8-, 11-, 13-, 14-, 15-, and 16-Membered Cyclic<br />

Transition States<br />

Hansen, F. K. 1 2; Ha, K. 2; Chahar, M. 2; Monbaliu, J.-C. M. 2 3; Oliferenko, A. A. 2; Katritzky, A.<br />

R. 2<br />

1 Institute of Pharmaceutical and Medicinal Chemistry, Heinrich-Heine-University of Düsseldorf,<br />

<strong>Universität</strong>sstr. 1, 40225 Düsseldorf, Germany. Email: finn.hansen@uni-duesseldorf.de<br />

2 Center for Heterocyclic Compounds, Department of Chemistry, University of Florida, Gainesville,<br />

FL 32611-7200, USA.<br />

3 Department of Sustainable Organic Chemistry and Technology, Faculty of Bioscience<br />

Engineering, Ghent University, B-9000 Ghent, Belgium.<br />

Native chemical ligation (NCL) is the most widely used chemoselective ligation technique<br />

based on a rearrangement concept nowadays. NCL involves the covalent condensation of<br />

a peptide-thioester with N-terminal cystein peptide, followed by S to N acyl shift to form a<br />

native amide linkage.[1] In recent years, various chemical ligation methodologies such as<br />

sugar-assisted ligation, nucleotide-based ligation and chemical ligation using click<br />

chemistry have been developed. The classical NCL method is limited to peptides<br />

possessing an N-terminal cysteine residue. Unfortunately, cysteine is a relatively rare<br />

amino acid (1.3% average content) and is not always available in a terminal position.<br />

Moreover, some amino acid-cysteine bonds such as Pro-Cys, Asp-Cys, Glu-Cys and Lys-<br />

Cys can be difficult to access by chemical ligation.[2]<br />

To overcome this requirement of a specifically placed cysteine residue one approach is the<br />

use of thiol ligation auxiliaries, but removable cysteine mimics can sterically hinder ligation<br />

and difficulties can arise at the stage of auxiliary removal.[3]<br />

A long-range S to N acyl migration methodology would avoid the requirement of a Nterminal<br />

cysteine residue. We have now studied the feasibility of chemical ligation of Sacylated<br />

cysteine peptides via 8-, 11-, 13-, 14-, 15-, and 16-membered cyclic transition<br />

states.[4,5] Our microwave-assisted methodology allows the synthesis of native peptides<br />

from non-terminal cysteine residues without the need of ligation auxiliaries.[4,5]<br />

Computational studies provide mechanistic evidence for the ligation process.[5] The impact<br />

of pH on the long-range S to N acyl migration is exemplified using a S-acyl tetrapeptide.[5]<br />

Acknowledgments: The authors gratefully acknowledge the Kenan foundation for financial support.<br />

References:<br />

1. Dawson, P. E. et al.: Science 1994, 266(5186): 776-779.<br />

2. Haase, C. and Seitz, O.: Eur. J. Org. Chem., 2009, (13): 2096-2101.<br />

3. Marinzi, C. et al.: Bioorg. Med. Chem. 2001, 9(9): 2323-2328.<br />

4. Hansen, F. K. et al.: Org. Biomol. Chem. 2011, 9(20): 7162-7167.<br />

5. Ha, K. et al.: J. Org. Chem., <strong>2012</strong>, 77(6): 2637-2648.<br />

034<br />

Synthesis and pharmacological evaluation of thiazole-featured pirinixic acid<br />

derivatives acting as dual 5-lipoxygenase and microsomal prostaglandin E2<br />

synthase-1 inhibitors<br />

Hanke, T. 1; Popella, S.-D. 2; Liening, S. 2; Dehm, F. 2; Lämmerhofer, M. 3; Werz, O. 2;<br />

Schubert-Zsilavecz, M. 1<br />

1Goethe University of Frankfurt am Main, Max-von-Laue Str. 9, 60438 Frankfurt am Main, Germany<br />

2Friedrich Schiller University of Jena, Philosophenweg 14, 07743 Jena, Germany<br />

3Eberhard Karls <strong>Universität</strong> Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, Germany<br />

Imbalances in the lipid signaling network contribute to the pathogenesis of a large number<br />

of human diseases. The metabolic fate of arachidonic acid (AA) plays a crucial role within<br />

this network and is associated with pathophysiological conditions such as inflammation,<br />

analgesia, asthma and cancer.<br />

The metabolic pathway of AA can be divided into two different ways: The formation of<br />

prostaglandins (PGs) catalyzed by cyclooxygenases (COXs) and the biosynthesis of<br />

leukotrienes (LTs) initialized by 5-lipoxygenase (5-LO). Non steroidal anti-inflammatory<br />

drugs (NSAIDs) and COX-2 selective inhibitors (coxibs) are the most wide-spread drugs in<br />

the therapy of inflammation. However, especially long-term use of these drugs is closely<br />

related to severe side effects such as gastrointestinal and renal complications (NSAIDs) or<br />

an increased cardiovascular risk (coxibs) due to the suppression of physiological relevant<br />

prostaglandins [1]. Consequently, new pharmacological strategies for anti-inflammatory<br />

therapy are urgently needed. One promising approach is the selective inhibition of<br />

downstream-acting enzymes such as the microsomal prostaglandin E2 synthase-1<br />

(mPGES-1), which catalyzes the formation of PGE2 from PGH2. PGE2 is the most<br />

prominent mediator in inflammatory pain. On the other hand, LTs produced by 5-LO are<br />

important inflammatory mediators which act as bronchoconstrictors and increase vascular<br />

permeability. The dual inhibition of 5-LO and mPGES-1 is considered as a novel strategy to<br />

avoid COX-related side effects such as the analgesic asthma syndrome and to maintain the<br />

physiological prostaglandin levels.<br />

The structural basis of the presented compounds is pirinixic acid, which is inactive on both,<br />

mPGES-1 and 5-LO. Especially the introduction of n-alkyl chains in α-position led to potent<br />

dual 5-LO / mPGES-1 inhibitors. Furthermore, a broad modification of the lipophilic<br />

backbone is tolerated with an equal or increased activity [2]. Herein we present a novel class<br />

of pirinixic acid derivatives featuring a thiazole-scaffold at the lipophilic backbone. The<br />

resulting thiazole-substituted derivatives show balanced dual inhibition of mPGES-1 and 5-<br />

LO with IC50 values mainly in the submicromolar range.<br />

References:<br />

1. Koeberle, A. et al.: Curr. Med. Chem. 2009, 16: 4274–4296.<br />

2. Koeberle, A. et al.: J. Med. Chem. 2008, 51: 8068–8076.<br />

035<br />

Optimization of transport conditions for the shipping of 3D human cornea<br />

constructs used for in vitro drug absorption studies<br />

Beißner N1; Reichl S1 1 Institut für Pharmazeutische Technologie, Technische <strong>Universität</strong> Carolo-Wilhelmina zu<br />

Braunschweig, Mendelssohnstraße 1, 38106 Braunschweig<br />

In vitro data of transcorneal absorption behaviour of ophthalmic drugs may be obtained by<br />

permeation studies using either excised cornea taken from experimental animals or corneal<br />

cell culture models. In recent years we developed serum-free cultivated 3D cornea<br />

constructs (HC) based on immortalized human corneal cells exhibiting similar barrier<br />

properties as excised porcine and rabbit cornea [1]. In a prevalidation study we generated<br />

SOPs for HC cultivation and permeation procedures and transferred SOPs in two other cell<br />

Poster 101


culture labs. Furthermore, in this study using seven ophthalmic drugs we demonstrated that<br />

our cornea model exhibits lower intra- and inter-laboratory variances than excised animal<br />

cornea [2]. The wide use of validated cell culture models normally requires valid cultivation<br />

in few labs and standardized shipment without loss of their functionality. In the present<br />

study we evaluated optimal transport conditions for the shipping of our cornea construct.<br />

Human cornea constructs were cultivated under serum-free conditions on permeable<br />

polycarbonate filters (Transwells ®) using SV40 immortalized human keratocytes (HCK-Ca)<br />

and human corneal epithelial cells (HCE-T) as described before [1]. After finishing of<br />

cultivation HC were kept in medium under submerged conditions and stored in different<br />

transport containers for various time periods changing shipment parameters as temperature<br />

and CO2 content. Cell viability after simulated transport was evaluated via MTT testing.<br />

Barrier properties of HC expressed as epithelial integrity was estimated by transepithelial<br />

electrical resistance (TEER) measurements.<br />

Both storage temperature and CO2 content had great impact on viability and TEER after 24<br />

and 48 h of transport simulation. Storage at 4 °C and 20 °C without additional CO2 support<br />

resulted in decreased TEER values. In addition, the viability was diminished at 4 °C. In<br />

contrast, keeping cornea constructs at lowered temperature in 5% CO2 atmosphere yielded<br />

viable HC exhibiting constant TEER values. However, lowering CO2 content resulted in<br />

strongly decreased TEER values, which couldn’t be recovered after cultivation for additional<br />

24 h in incubator (37 °C, 5% CO2, humidified atmosphere). Sealing of storage container led<br />

to TEER values similar to those obtained for HC stored under incubator conditions at same<br />

temperature. The combination of sealed transport containers and lowered temperature (22<br />

– 24 °C) caused only marginal reduction of TEER values after 24 h storage. Various<br />

preparatory treatments of cell culture medium as pre-warming to 37 °C or 24 h equilibration<br />

in incubator atmosphere (5% CO2) before use seemed to have no significant influence on<br />

the resulting corneal barrier properties after shipment simulation. Additionally, the container<br />

material (polystyrene, polyamide) didn’t show significant impact.<br />

Our results suggest CO2 atmosphere and medium pH, respectively, as critical parameters<br />

during transport of HC. Thus, an optimized transport system should ensure constant CO2<br />

atmosphere during shipment. In the next step appropriate transport container will be<br />

developed to ensure these requirements. Furthermore, real time transport experiments<br />

have to prove the usefulness of our developed shipment SOP.<br />

Acknowledgments: We are grateful to the German Federal Institute for Risk Assessment (BfR), which<br />

funded this work under grant no. 3-1328-488.1.<br />

References:<br />

1. Hahne, M., Reichl, S., International Journal of Pharmaceutics, 2011, 416, 268-279<br />

2. Hahne, M. et al., Journal of Pharmaceutical Science (accepted and in press,) <strong>2012</strong>, Prevalidation of a<br />

human construct as an alternative to animal corneas for vitro drug absorption studies<br />

036<br />

Type-I-allergic diseases and a possible way to treat them: new clozapinederived<br />

Histamine H1-/H4-receptor-ligands and their pharmacological<br />

characterization<br />

Gobleder S. 1, Wittmann H.-J. 2, Elz S. 1 Straßer A. 3<br />

1 Department of Pharm./Med. Chemistry I, <strong>Universität</strong>sstr. 31, 93040 Regensburg<br />

2 Faculty of Chemistry and Pharmacy, <strong>Universität</strong>sstr. 31, 93040 Regensburg<br />

3 Department of Pharm./Med. Chemistry II, <strong>Universität</strong>sstr. 31, 93040 Regensburg<br />

The number of patients that suffer from allergic rhinitis and conjunctivitis (the so called<br />

“hay-fever”) increases every year, a trend that indicates the need of good pharmaceutical<br />

treatment. A very common and well-tried therapy is the combined p.o. and local application<br />

of “antihistamines”, drugs with antagonistic effects at H1R, such as cetirizine and azelastine<br />

to name only a few. Recent research results pointed out, that not only the H1R but also the<br />

H4R is involved in type-I-allergic diseases. [1] Good therapeutic response could be<br />

achieved in the acute murine asthma model with a combined application of mepyramine<br />

(H1-antagonist) and JNJ7777120 (H4-antagonist). [2] These results suggest that addressing<br />

both, H1R and H4R, could be a new therapy-option of hay-fever and other type-I-allergic<br />

diseases like histamine induced itch or inflammatory asthma.<br />

Screening compounds with good affinity to H1R and H4R, we choose the antipsychotic drug<br />

clozapine (1) which has high antagonistic affinity to H1R and moderate agonistic affinity to<br />

H4R. [3] Soft structure modifications, e.g. exchanging the 5-nitrogen by a 5-oxygen and<br />

shifting the 8-chlorine to position 7 lead to an optimized clozapine-derivative (2) with<br />

increased agonistic affinity at H4R and constant high antagonistic affinity to H1R. [4] The<br />

aim of the project was to get a closer insight into structure-activity relationships at H1R and<br />

H4R and, according to our results, to convert the agonism at H4R into an antagonism or<br />

inverse agonism by modifying the clozapine-molecule in different ways.<br />

In a first part of the project we reduced the 5H-dibenzo[b,e][1,4]diazepine-structure of<br />

clozapine to an “open” (E)-N-((phenyl)methylene)aniline structure and varied the kind and<br />

position of substituents on both aromatic rings. Examples of the synthesized compounds<br />

are shown in (3), (4) and (5). Since our compounds showed, compared to clozapine, a<br />

decreased affinity to hH1R and hH4R, we continued synthesizing compounds with<br />

dibenzo[b,f][1,4]oxazepine-structure and added new non-halogenide substituents to this<br />

optimized clozapine derivatives (6), (7) and (8). All compounds were tested at the guinea<br />

pig ileum in presence of histamine and in a competition binding assay at hH1R and hH4R.<br />

For selected compounds ((6), (7) and (8)) we performed a GTPγS assay for hH2R, hH3R<br />

and hH4R to get information about receptor-selectivity and potency. First data revealed<br />

selectivity towards hH1R and/or hH4R for all tested compounds. To explain structure-activity<br />

relationships on a molecular level, molecular modelling studies at appropriate models of<br />

hH1R and hH4R were performed.<br />

R 1<br />

R 2<br />

N N<br />

X<br />

N<br />

R1 Cl,R2 H,X NH 1<br />

R 1 H,R 2 Cl,X O 2<br />

R 1<br />

R 2<br />

N N<br />

N<br />

R1 R2 H,R3<br />

R3<br />

Cl 3<br />

R1 Cl,R2 R3 H 4<br />

R1 H,R2 Cl,R3 NH2 5<br />

R 1<br />

R 2<br />

pA2 pKi pKi<br />

gpH1R hH1R hH4R<br />

1 n.d. 8,6 5,9<br />

2 7,6 7,8 7,0<br />

N N<br />

O<br />

N<br />

NH2<br />

R1 Cl,R2 H 6<br />

R 1 H,R 2 Cl 7<br />

R1 R2 H 8<br />

3 6,6 6,1 4,7<br />

4 6,9 6,0 4,9<br />

5 5,0 4,1 6,6<br />

6 8,9 8,2 6,1<br />

7 7,9 7,7 6,5<br />

8 n.d. 8,0 5,1<br />

References:<br />

1. Thurmond, R.L. et al.: Nature Reviews Drug Discovery 2008, 7 : 41-53.<br />

2. Deml, K.-F. et al.: Mol. Pharmacol. 2009, 76 : 1019-1030.<br />

3. Appl, H. et al.: Nauyn-Schmiedeberg’s Arch. Pharmacol. <strong>2012</strong>, 385(2) : 145-170.<br />

4. Smits, R. A. et al.: J. Med. Chem. 2006, 49 : 4512-4516.<br />

037<br />

Optimization of capillary electrophoresis-based analysis of 5-lipoxygenase<br />

metabolites.<br />

Abromeit H; Schaible A M; Werz O, Scriba G K E<br />

Department of Pharmaceutical/Medical Chemistry, Friedrich-Schiller-<strong>Universität</strong>, Philosophenweg<br />

14, 07743 Jena, Germany<br />

A new sensitive method using α-cyclodextrin-modified micellar electrokinetic<br />

chromatography has been developed to separate and quantify arachidonic acid metabolites<br />

of the lipoxygenase pathways in human polymorphonuclear leukocytes, i.e. leukotriene B4,<br />

6-trans-leukotriene B4, 6-trans-12-epi-leukotriene B4, 5(S)-hydroxy-6-trans-8,11,14-ciseicosatetraenoic<br />

acid, 12(S)-hydroxy-6-trans-8,11,14-cis-eicosatetraenoic acid, and 15(S)hydroxy-6-trans-8,11,14-cis-eicosatetraenoic<br />

acid. The electrophoresis system was<br />

optimized with regard to the pH, boric acid, SDS and α-cyclodextrin concentration as well<br />

as separation voltage and temperature using a three level resolution IV fractional factorial<br />

design and a five level circumscribed central composite design. The optimized conditions<br />

included 80 mM sodium borate buffer, pH 10.07, containing 16.6 mM sodium dodecyl<br />

sulfate and 15 mM α-cyclodextrin, using a separation voltage of 12.5 kV at 23 °C.<br />

Sensitivity was enhanced employing head-column field amplified sample stacking which<br />

resulted in limits of quantification between 30 and 50 ng/mL and limits of detection between<br />

10 and 17 ng/mL after solid phase extraction of the lipoxygenase products. The method<br />

was validated according to the recommendations of the International Conference on<br />

Harmonization and applied to the determination of the lipoxygenase metabolites in<br />

polymorphonuclear leukocytes upon stimulation with Ca2+-ionophore A23187 and<br />

arachidonic acid. Robustness was confirmed using a three level resolution IV fractional<br />

factorial design. In conclusion, the novel method is suitable for the analysis of various<br />

arachidonic acid metabolites produced by living cells and may be used for evaluation of<br />

lipoxygenase inhibitors.<br />

038<br />

Carnosol and Carnosic acids from Salvia officinalis inhibit microsomal<br />

Prostaglandin E2 Synthase-1<br />

Bauer, J1., Kuehnl, S. 2, Rollinger, J. M. 2, Scherer, O. 4, Northoff, H. 3, Stuppner, H. 2, Werz,<br />

O. 4, Koeberle, A. 4<br />

1 Department for Pharmaceutical Analytics, Pharmaceutical Institute, University of Tuebingen,<br />

Tuebingen, Germany);<br />

2 Institute of Pharmacy / Pharmacognosy and Center for Molecular Biosciences Innsbruck,<br />

University of Innsbruck, Innsbruck, Austria<br />

3 Institute for Clinical and Experimental Transfusion Medicine, University Medical Center,<br />

Tuebingen, Germany<br />

4 Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, University of Jena, Jena,<br />

Germany<br />

Prostaglandin E2 (PGE2), the most relevant eicosanoid promoting inflammation and<br />

tumorigenesis, is formed by cyclooxygenases (COXs) and PGE2 synthases from free<br />

arachidonic acid. Preparations of the leaves of Salvia officinalis are commonly used in folk<br />

medicine as an effective antiseptic and anti-inflammatory remedy and possess anticancer<br />

activity. Here, we demonstrate that a standard ethyl acetate extract of S. officinalis<br />

efficiently suppresses the formation of PGE2 in a cell-free assay by direct interference with<br />

microsomal PGE2 synthase (mPGES)-1. Bioactivityguided fractionation of the extract<br />

yielded closely related fractions that potently suppressed mPGES-1 with IC50 values<br />

between 1.9 and 3.5 µg/ml. Component analysis of these fractions revealed the diterpenes<br />

carnosol and carnosic acid as potential bioactive principles inhibiting mPGES-1 activity with<br />

IC50 values of 5.0 µM. Using a human whole-blood assay as a robust cell-based model,<br />

carnosic acid, but not carnosol, blocked PGE2 generation upon stimulation with<br />

lipopolysaccharide (IC50 9.3 µM). Carnosic acid neither inhibited the concomitant<br />

biosynthesis of other prostanoids [6-keto PGF1a, 12(S)-hydroxy-5-cis-8,10-transheptadecatrienoic<br />

acid, and thromboxane B2] in human whole blood nor affected the<br />

activities of COX-1/2 in a cell-free assay. Together, S. officinalis extracts and its ingredients<br />

carnosol and carnosic acid inhibit PGE2 formation by selectively targeting mPGES-1. We<br />

conclude that the inhibitory effect of carnosic acid on PGE2 formation, observed in the<br />

physiologically relevant whole-blood model, may critically contribute to the antiinflammatory<br />

and anticarcinogenic properties of S. officinalis.<br />

039<br />

Investigation of the degradation of the model tetrapeptide Gly-Phe-Asp-GlyOH<br />

at alkaline pH by capillary electrophoresis including identification of the<br />

degradation products by CE-ESI-MS<br />

Brückner, C. 1; Scriba, G. K. E. 1<br />

1 Friedrich Schiller University, Department of Pharmaceutical and Medicinal Chemistry, School of<br />

Pharmacy, Philosophenweg 14, 07743 Jena, Germany<br />

Asparagine and aspartic acid are prone to chemical degradation such as deamidation,<br />

isomerization, and enantiomerization. [1-3] Enantiomerization and isomerization proceed<br />

via the formation of an aminosuccinimidyl intermediate (Asu). Hydrolysis of the Asu peptide<br />

yields native Asp and β-Asp (iso-Asp) peptides favouring the β-Asp peptide over the Asp<br />

peptide by a ratio of 3-5:1. [1,2] Differentiation of the Asp isomers by mass spectrometry<br />

can be achieved by the ratio of the complementary b- and y-ions generated during peptide<br />

fragmentation. [4] In contrast, diastereomeric Asp peptides, i.e. peptides containing an L-<br />

Asp or D-Asp moiety could not be distinguished based on their MS spectra.<br />

102 Poster


The degradation of the model tetrapeptide Gly-L-Phe-α-L-Asp-GlyOH was investigated at<br />

80°C and 25°C in sodium borate buffer pH 10. Separation and quantification of the<br />

degradation products was achieved using a validated capillary electrophoresis assay. Peak<br />

identification was obtained by CE-ESI-MS experiments and by co-injection of synthetic<br />

reference compounds. Due to the differences in the MS spectra the degradation products<br />

could be clearly assigned to the α-Asp or β-Asp linkage. While for α-Asp peptides b2-ion<br />

formation competes with y2-ion formation, in case of β-Asp peptides the intensity of the y2ion<br />

considerably exceeded that of the b2-ion. At 80°C a rapid decline of the starting peptide<br />

was observed yielding a mixture of α-Asp and β-Asp as well as L-Asp and D-Asp containing<br />

peptides. The β-aspartyl peptides exceeded the α-Asp linkage after 288 h by a ratio of<br />

4.0:1. Furthermore, the peptides containing the D-Asp moiety were favored over the L-Asp<br />

residues. At 25°C the tetrapeptide Gly-L-Phe-α-L-Asp-GlyOH proved to be relatively stable<br />

compared to 80°C. After an incubation interval of 2350 h 88 % of the starting peptide was<br />

still present in the incubation solution. Nevertheless, formation of the enantiomerization and<br />

isomerization products could be demonstrated yielding β-Asp residues in higher amounts<br />

as the respective α-D-Asp peptide.<br />

References:<br />

1. Aswad DW, CRC Press. 1995, 279.<br />

2. Geiger T, Clarke S, J. Biol. Chem. 1987, 262(2):785-794.<br />

3. Manning MC, Patel K, Borchardt RT, Pharm. Res. 1989, 6(11):903-918.<br />

4. Lehmann, WD. et al., Protein Sci. 2000, 9(11):2260-2268.<br />

040<br />

Evaluation of the cytoprotective effects of individual and combined<br />

constituents of the combination drug STW 5<br />

Hoser S1; Winkelmann V1; Kelber O2, Abdel-Aziz H2, Weiser D2, Nieber K1 1 University of Leipzig, Institute of Pharmacy, Talstr. 33, 04103 Leipzig, Germany<br />

2 Steigerwald Arzneimittelwerk GmbH, Scientific Department, Havelstr. 5, 64295 Darmstadt,<br />

Germany<br />

STW 5 (Iberogast ® ), a multi-component herbal drug, is successfully used in the therapy of<br />

functional dyspepsia and irritable bowel syndrome (IBS). We investigated the cytoprotective<br />

action of individual herbal extracts of STW 5 in CaCo-2 cells. The CaCo-2 cells were<br />

incubated with the herbal extracts and stimulated with LPS (10 ng/ml) for 2 hours. Possible<br />

synergistic effects were determined by isobologram analysis of LPS-induced LDH release<br />

as a measure for cell toxicity using a commercially available kit.<br />

STW 5 (500.5 µg/ml) significantly inhibited LPS-induced LDH release by 74 %.<br />

Comparable effects were found with lemon balm, whereas in equivalent concentrations to<br />

STW 5 the effects of Iberis amara, peppermint, chamomile, angelica and milk thistle were<br />

less potent. Caraway and celandine had stimulatory effects. Isobolgram analysis indicated<br />

that a 2 h exposure to peppermint and Iberis amara (ratio 0.5/0.5) resulted in a synergistic<br />

effect of 28 %. Less pronounced synergy was observed using the component ratio<br />

0.75/0.25. An additive effect was present in the ratio 0.25/0.75. Application of peppermint<br />

together with milk thistle (ratio 0.5/0.5) showed ratio-independent synergistic effects of 13-<br />

16 % whereas the combined application of chamomile and Iberis amara had ratioindependent<br />

additive effects. The combination of chamomile and angelica (ratio 0.5/0.5)<br />

exerted an additive effect, the ratio 0.75/0.25 showed an antagonistic effect which<br />

increased using the ratio 0.25/0.75.<br />

Our observations confirm that the components of STW 5 contribute to different extents to its<br />

cytoprotective effect in CaCo-2 cells and clearly indicate that the individual herbal extracts<br />

can contribute synergistically, additive and antagonistically to the overall effects of STW 5.<br />

The type of effect was determined in some cases by the ratio of the constituents.<br />

041<br />

Novel synthetic analogues of toxiferine I and their pharmacological<br />

characterization at α7 nAChRs, muscle-type nAChRs, and the allosteric<br />

binding site of muscarinic M2 receptors<br />

Zlotos, D.P1; Gündisch, D2;Tränkle, C3; Holzgrabe, U4; Jensen, A.A5 1 The German University in Cairo, Dept. of Pharmaceutical Chemistry, New Cairo City, 11835<br />

Cairo, Egypt<br />

2 Department of Pharmaceutical Sciences, College of Pharmacy, University of Hawai'i at Hilo, Hilo,<br />

HI 96720, USA<br />

3 Pharmacology and Toxicology Section, Institute of Pharmacy, Rheinische Friedrich-Wilhelms-<br />

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

4Institut für Pharmazie and Lebensmittelchemie, <strong>Universität</strong> Würzburg, Am Hubland, 97074<br />

Würzburg, Germany<br />

5 Department of Medicinal Chemistry, University of Copenhagen, Universitetsparken 2, DK-2100<br />

Copenhagen, Denmark<br />

Toxiferine I is a major bisquaternary alkaloid of calabash-curare and Strychnos species. [1]<br />

Its semi-synthetic diallyl analogue alcuronium (Alloferin ®) is in clinical use as muscle<br />

relaxant. Both compounds display high binding affinities for the muscle-type nAChRs from<br />

the electric organ of Torpedo californica. [2] In addition to their neuromuscular-blocking<br />

activity, toxiferine I and alcuronium have been reported to be moderately potent antagonists<br />

at α7-nAChRs [3], and to possess moderate to high affinity for the allosteric binding site of<br />

M2-mAChRs, respectively. [4] Interestingly, the structurally related caracurine V analogues<br />

show a different pharmacological profile displaying considerably lower affinity for the<br />

muscle-type nAChRs, [2] higher antagonistic activity at α7-nAChRs, [3] and similar<br />

allosteric potency at M2-mAChRs, [4] when compared to the correspondingly substituted<br />

bisnortoxiferine analogues. Here, we describe the synthesis and pharmacological<br />

evaluation of novel bisquaternary toxiferine I analogues with structurally modified side<br />

chains. In particular, a series of non-symmetrical derivatives lacking only one alcohol group<br />

(1a-c), and of symmetrical compounds lacking both alcohol functions (2a-c) have been<br />

examined. The findings help delineate the structural requirements for the action at different<br />

AChRs and design ligands selective for one of the subtypes.<br />

Poster 103<br />

OH<br />

H<br />

N<br />

R<br />

N H<br />

H<br />

H<br />

H N<br />

R<br />

N<br />

H<br />

toxiferine I, R Me<br />

alcuronium, R allyl<br />

OH<br />

2Cl -<br />

H<br />

N H H<br />

H<br />

O<br />

O<br />

H<br />

H<br />

H N<br />

H<br />

N<br />

R +<br />

+ R<br />

N<br />

caracurinium V analogues<br />

R Me, allyl, pNO 2 -benzyl<br />

2X -<br />

1<br />

R<br />

H<br />

2 N<br />

R<br />

N H<br />

H<br />

H<br />

H N<br />

2<br />

R<br />

N<br />

H<br />

CH 3<br />

2X -<br />

1a c R 1 -CH 2 OH, R 2 Me, allyl, pNO 2 -benzyl<br />

2a c R 1 -CH 3 , R 2 Me, allyl, pNO 2 -benzyl<br />

Acknowledgments: Bonn University, Mechthild Kepe, Würzburg University, Elfriede Ruckdeschel, Matthias<br />

Grüne<br />

References:<br />

1. Battersby, A.R.; Hodson, H.F. Q. Rev. Chem. Soc. 1960, 14, 77-103.<br />

2. Zlotos, D.P. et al.: Bioorg. Med. Chem., 2004, 12, 6277-6285.<br />

3. Jensen, A.A.; Zlotos, D.P.; Liljefors, T. J. Med. Chem. 2007, 50, 4616-4629.<br />

4. Zlotos, D. P. et al.: J. Med. Chem. 2004, 47, 3561-3571.<br />

042<br />

Ligand based approaches to reveal the active conformation of highly flexible<br />

azecine derivative<br />

Enzensperger C1, Robaa D2, Schulze M3, Lehmann J1 1 Institute for Pharmacy, Department of Medicinal Chemistry, Philosophenweg 14, D-07743 Jena,<br />

Germany<br />

2 Institute for Pharmacy, Department of Medicinal Chemistry, Wolfgang-Langenbeck Str. 4 D-06120<br />

Halle-Wittenberg, Germany<br />

3 Temporarily at ‘Institute de Genomique Fonctionnelle-CNRS’, Montpellier cedex 05, France<br />

Annulated azecine derivatives are highly flexible compounds and some of them exert very<br />

high affinity for dopamine and serotonin receptors. In the crystal structure there are<br />

obviously two enantiomers present, which give rise to the question about the eutomer,<br />

dystomer and the active conformation (in figure 1 LE300 (1) is shown). We succeeded to<br />

synthesize, screen and X-ray an enantiopure chiral benzindolo azecine (2).[1]<br />

Figure 1: Both enantiomers of LE300 (1) present 1:1 in the crystal (left). The absolute<br />

configuration and conformation of the chiral eutomer (2) (right)<br />

This crystal structure is the only conformation that allows a rigid alignment with<br />

conformationally constricted and stereochemically defined templates like asenapine (3) and<br />

ecopipam (4). In that case, the indole moiety can only be superimposed with the aromatic<br />

ring not containing a chloro subtitutent.<br />

O<br />

Cl<br />

1<br />

H<br />

N<br />

H<br />

N CH3<br />

3<br />

H<br />

N CH3<br />

S<br />

CH3<br />

Cl<br />

HO<br />

N<br />

H<br />

N CH3<br />

Figure 2: Rigid alignment of the X-ray structure of the chiral eutomer (2) with asenapine (3)<br />

and ecopipam (4)<br />

This hypothesis is also supported by observed SARs in the class of benzazepine- and<br />

benzindolo- azecine derivatives. The most important structural features of these<br />

compounds seem to be the shape and the site of protonation of the basic nitrogen. We are<br />

now also able to explain, why azecine derivatives with a different annulation pattern are<br />

inactive.[2]<br />

References:<br />

1. Robaa, D.et al., J Med Chem 2011, 54: 7422-6.<br />

2. Schulze, M. et al., Bioorg Med Chem 2009, 17: 6898-907.<br />

2<br />

1<br />

N CH3<br />

CH3<br />

S<br />

043<br />

Design, synthesis, biological testing and SAR of 5-Pyridinyl-2-thioimidazole<br />

derivatives as selective JNK3-inhibitors over p38α<br />

S. Klos1, F. M. Muth1, M. Goettert / S. Bauer / K. Bauer1, S. Laufer1 1 Institute of Pharmacy, Department of Pharmaceutical and Medicinal Chemistry, Eberhard-Karls-<br />

University, Auf der Morgenstelle 8, 72076 Tuebingen, Germany<br />

Many inflammatory conditions are driven by both p38α and JNK3 MAPK.[1] To differentiate<br />

the contribution of each kinase, selective inhibitors are necessary. For p38α this might be<br />

the case[2], but as of today only few selective JNK3 inhibitors with good ADME (Absorption,<br />

Distribution, Metabolism, Excretion) properties are available.<br />

JNK-kinases are c-jun NH2-terminal serine/threonine mitogen activated kinases which are<br />

Cl<br />

HO<br />

4<br />

N CH3<br />

Cl<br />

H<br />

O<br />

2<br />

H<br />

N CH3


mainly activated by cytokines and environmental influences.[3][4] JNK3-kinases are<br />

believed to play a central role in the pathology of neurologic diseases such as<br />

cerebrovascular accidents, Parkinson's and Alzheimer's disease.[5] Therefore it has<br />

become an attractive and valid drug target.<br />

5-Pyridinyl-2-thioimidazole derivatives are known as p38α-inhibitores.[6] Due to the<br />

sequential and steric similarity of p38α- and JNK3-kinase, we assumed to gain active and<br />

selective JNK3-inhibitores by introducing different substitution patterns for R1, R2, and R3.<br />

At the edge of the hydrophobic region II in JNK3-kinase, there are Asn152 and Gln155,<br />

whereas Asp112 and Asn152 are shown in p38α-kinase.[3] In order to create repulsion<br />

between the Asp112 and p38α, we introduced anionic substituents at the aminopyridine<br />

scaffold.<br />

Furthermore, we tried to hit the Asp112[3] by introducing carboxylic moieties at the<br />

imidazole nitrogen. In order to target the conserved but steric diverse Arg107 and<br />

Asn194[3], we synthesised carboxylic substituents linked by a sulfide at R1 resulting in 50<br />

nM inhibition of JNK3 with about 10 fold selectivity against p38α.<br />

References:<br />

1. Kamenecka T.,: J. Biol. Chem. 2009, 284(19): 12852-61.<br />

2. Koebler S. C., et al., Nat. Chem. Biol. <strong>2012</strong>, 8(2): 141-3.<br />

3. Scapin G. L., et al., Chem. Biol. 2003, 10: 705-712.<br />

4. Johnson G. L., et al., Science 2002, 298: 1911-1912.<br />

5. Bogoyevitch M. A., Trends Mol. Med. 2005, 11: 232-239.<br />

6. Laufer S. A., et al., J. Med. Chem. 2008, 51(14), 4122-4149.<br />

044<br />

8-Substitued Indolo[3,2-f][3]Benzazecines Receptor Antagonists: Synthesis<br />

and Activity of Racemic and Enantiopure Derivatives<br />

Robaa, D. 1; Enzensperger, C. 2; AbulAzm, S.; 3 Lehmann, J. 2<br />

1Institute for Pharmacy, Department of Medicinal Chemistry, Wolfgang-Langenbeck Str. 4, 06120<br />

Halle<br />

2Institute for Pharmacy, Department of Medicinal Chemistry, Philosophenweg 14, 07743 Jena,<br />

Germany<br />

3Department of Medicinal Chemistry, Faculty of Pharmacy, El Khartoum Square 1, 21521<br />

Alexandria, Egypt<br />

Partially hydrogenated indolo[3,2-f][3]benzazecines represent a structurally novel class of<br />

dopamine receptor antagonists, distinguished by nanomolar affinities as well as their<br />

preference for the receptors of the D1 family [1,2].<br />

Derivatives of the lead indolo[3,2-f][3]benzazecine derivative LE 300 (1), substituted at<br />

position 8 with three different residues, namely methyl, hydroxymethyl and carboxylic acid,<br />

were prepared. The 8-methyl and 8-hydroxymethyl derivatives were obtained as the<br />

separated R- and S- enantiomer as well as the racemic mixture, while the amino acid<br />

derivative could only be obtained in a racemized form. The separate enantiomers showed<br />

significantly different affinities; the (8S)-methyl and (8R)-hyroxymethyl derivatives where the<br />

substituents point below the reference plane of the indolo[3,2-f][3]benzazecine scaffold<br />

were markedly more active than their enantiomeric counterparts. The racemic 8-carboxy<br />

derivative was highly interesting since it showed a pronounced selectivity for the D5receptor,<br />

even against D1.<br />

11<br />

10<br />

12<br />

13 N<br />

H<br />

1<br />

1<br />

(LE300)<br />

9<br />

8<br />

CH3 N 6<br />

7<br />

15 5<br />

1<br />

2<br />

3<br />

N<br />

H<br />

R<br />

CH3 N<br />

Derivatives with substituents<br />

pointing below the projection<br />

place<br />

References:<br />

1. Hoefgen, B. et al.: J. Med. Chem. 2006, 49, 760–769.<br />

2. Witt, T.; Hock, F. J.; Lehmann, J. J. Med. Chem. 2000, 43, 2079–2081.<br />

N<br />

H<br />

R<br />

CH3 N<br />

Derivatives with substituents<br />

pointing above the projection<br />

place<br />

045<br />

Influence of API and Surfactant Selection on Nanosuspensions Properties<br />

Comparing Two Different Top Down Methods<br />

Lestari, Maria L.A.D1; Müller, R.H1; Möschwitzer, J.P 1 2 *<br />

1 Freie <strong>Universität</strong> Berlin, Institute of Pharmacy, Biopharmaceutics & NutriCosmetics, Kelchstrasse<br />

31, 12169 Berlin, Germany<br />

2 Pharmaceutical Development, Abbott GmbH &Co.KG, Ludwigshafen, Germany<br />

* Corresponding author: Telephone: 030-838-50703, email: info@janmoeschwitzer.de<br />

High Pressure Homogenization (HPH) and wet ball milling (WBM) are two well known top<br />

down methods for producing nanosuspension. Both methods are based on different<br />

diminution principles. These different principles may affect the stabilization of the<br />

nanosuspension as well as process efficiency. Besides these two factors also the active<br />

pharmaceutical ingredients (API) and surfactants are playing an important role for the<br />

particle size reduction process. Amphotericine B and resveratrol were used as model drugs<br />

in this study. Various surfactants (anionic, cationic, nonionic, polymeric and combination)<br />

were tested at specified concentration level. Nanosuspensions were produced with low<br />

energy wet ball milling using yttrium stabilized milling beads (diameter 0.5 mm) with electric<br />

stirring system at 900 rpm for 72h. HPH was done by two pre-milling cycles at 150 and 500<br />

bar and 20 cycles at high pressure of 1500 bar. The nanosuspensions were characterized<br />

using photon correlation spectroscopy (PCS), laser diffractometry (LD), light polarized<br />

microscopy and laser doppler anemometry (zeta potential). Results showed that LE-WBM<br />

of resveratrol produced nanosuspensions with mean particle sizes below 500 nm and a<br />

narrow size distribution. In case of WBM only TPGS led to a mean particle size larger than<br />

1000nm. In contrast, HPH of resveratrol resulted in general in larger particle sizes above<br />

1000 nm. Only nanosuspensions stabilized with sodium cholate had particle sizes below<br />

1000 nm. For amphotericine B, some nanosuspensions had similar particle sizes<br />

irrespectively of the employed technique. Sodium cholate and a combination of sodium<br />

cholate-poloxamer were identified as most suitable stabilizers for both techniques. In<br />

conclusion, both models behaved differently regarding the optimal stabilizer applied. When<br />

LE-WBM technique was used, stabilizer selection was less important since most of the<br />

nanosuspension produced had small mean particle sizes. For the HPH method, the<br />

selection of the right stabilizer system in combination with the API was more important.<br />

046<br />

inSARa: SAR interpretation by network navigation<br />

Wollenhaupt, S. 1; Baumann, K. 1<br />

1 Institut für Medizinische und Pharmazeutische Chemie, Technische <strong>Universität</strong> Braunschweig,<br />

Beethovenstr. 55, D-38106 Braunschweig, Germany<br />

Structure-Activity-Relationship (SAR) analysis of small molecules is a fundamental and<br />

challenging task in drug discovery. The knowledge of this relationship between structural<br />

determinants and bioactivity is of high value for the medicinal chemist e.g. in the leadoptimization<br />

process or de-novo-design. For the elucidation of SARs the recognition of<br />

molecular similarities is a crucial step due to the assumption that molecules with similar<br />

properties or features exhibit similar biological activities [1].<br />

The question arising from this is how to define molecular similarity ideally. Fingerprintbased<br />

similarity is an established and widely used concept [2]. Fingerprints represent<br />

molecules in the form of binary vectors which show the presence or absence of<br />

substructures or other structural features. Although fingerprints are fast to compute and well<br />

suited for processing large data sets, the interpretation of the results is difficult and the<br />

concept appears less intuitive to a medicinal chemist. A promising and more intuitive<br />

alternative is the maximum common substructure (MCS), the largest common substructure<br />

of a pair of molecules. One drawback is the high computational complexity which prevents<br />

large-scale applications such as the analysis of high-throughput-screening data. Moreover,<br />

exact atom matching is a limiting factor which decreases the scaffold hopping potential and<br />

hinders the identification of structurally less similar bioisosteric substructures.<br />

To tackle these limitations our in-house approach inSARa (intuitive networks for Structure-<br />

Activity-Relationships analysis) combines MCS similarity with the reduced graph (RG)<br />

concept [3]. Reduced graphs promise a higher degree of abstraction as they provide a<br />

conceptual representation of physicochemical properties and pharmacophoric features of a<br />

molecule by reducing groups of atoms to single pseudoatoms while retaining the topology<br />

between these functional units.<br />

inSARa networks are derived by combining RG-MCS-calculations and iterative super- and<br />

substructure searches. By linking the molecules with bioactivity data the networks can be<br />

used for SAR interpretation. This opens up the possibility to display molecules sharing<br />

common features as a function of bioactivity. Furthermore, inSARa networks are of great<br />

value for the detection of so called activity cliffs [4], where minor structural changes lead to<br />

a major change in bioactivity. Even though no bioactivity information is used to generate the<br />

inSARa networks, they can also be used for compound classification and bioactivity<br />

prediction.<br />

References:<br />

1. Johnson, M. A., Maggiora, G. M.: Concepts and applications of molecular similarity (Wiley) 1990.<br />

2. Wawer, M. et al.: J. Med. Chem. 2008, 51(19): 6075-6084.<br />

3. Gardiner, E. J. et al.: J. Chem. Inf. Model. 2007, 47(2): 354-366.<br />

4. Stumpfe, D., Bajorath, J.: J. Med. Chem. <strong>2012</strong>, 55(7): 2932-2942.<br />

047<br />

Dibenzosuberones as highly potent p38α MAP kinase inhibitors: Efficient use<br />

of a hydrophobic pocket to improve activity<br />

Fischer S1, Wentsch H1 , Karcher S1, Dorn A1, Laufer S1 1 Institute of Pharmacy, Department of Pharmaceutical Chemistry, University of Tuebingen, Auf der<br />

Morgenstelle 8, 72076, Germany<br />

The dysregulation of inflammatory cellular signal transduction cascades is known to be<br />

involved in the development of many diseases including chronic inflammatory and<br />

autoimmune diseases like rheumatoid arthritis (RA) and inflammatory bowel disease (IBD).<br />

p38α MAP kinase is a key enzyme regarding the regulation of the pro-inflammatory<br />

cytokines TNF-α and IL-1β and is therefore a promising target for the treatment of many<br />

inflammatory diseases. 1 2<br />

By now, successful development of p38α MAP kinase inhibitors as drugs has been<br />

compromised by poor selectivity and / or high toxicity. 3 The ATP-binding pocket of kinases<br />

is highly conserved, offering almost no possibilities to achieve selectivity. Therefore our<br />

efforts concentrated on exploiting unique features of the enzyme. One of them is the ability<br />

to perform the so called “glycine-flip”, which is already enforced by the suberone template.<br />

Another one is the “selectivity-pocket”. This region is located adjacent to the ATP-bindingpocket.<br />

Figure 1: Proposed binding mode of dibenzosuberones and essential interaction sites<br />

(Hinge Region, Selectivity Pocket, Deep Pocket)<br />

In contrast to other kinases, the “selectivity-pocket” is accessible because of the small<br />

gatekeeper residue Thr106. As it is mostly surrounded by lipophilic residues, we had to<br />

104 Poster


design moieties fitting exactly in this location. By extending these moieties towards the<br />

deep pocket, we were able to achieve high potency in the p38α MAP kinase enzyme assay<br />

down to an IC50 of 1nM.<br />

References:<br />

1. Badger et al., J. Pharmacol. Exp. Ther. 1996, 279, 1453-1461.<br />

2. Boehm et al., J. Med. Chem. 1996, 39, 3929-3937.<br />

3. Dominguez et al., Curr. Opin. Drug Discov. Devel. 2005, 8, 421-430.<br />

048<br />

Different column classification systems applied to calixarene- and<br />

resorcinarene-bonded stationary phases<br />

Chamseddin, C.; Jira, T.<br />

Institute of Pharmacy, Pharmaceutical/Medicinal Chemistry, <strong>Ernst</strong>-<strong>Moritz</strong>-<strong>Arndt</strong>-University of<br />

<strong>Greifswald</strong>, Friedrich-Ludwig-Jahn-Strasse 17, <strong>Greifswald</strong> D-17487, Germany.<br />

The recently dramatic increase in the number of reversed phase columns could be an<br />

advantage of this liquid chromatographic technique. However, due to the insufficiency of<br />

information that is available from manufacturers in terms of the exact functionality of their<br />

phases, it is problematic to determine which could be the best column for a given situation.<br />

There is not a “super column” that will work best for all applications. As a result, there have<br />

been several attempts to develop testing strategies to characterize column chemistries.<br />

These column classification systems are typically based on one or more test mixtures to<br />

identify the so called “column parameters”. Three different systems, originally developed for<br />

reversed phases, were applied to evaluate the chromatographic properties of the<br />

calixarene- and resorcinarene-bonded stationary phases. These are: Tanaka, USP, and<br />

Snyder/Dolan approaches [1,2,3]. Although all column classification systems aim to<br />

evaluate the “more or less” same characteristics, each system uses different test mixtures<br />

in different chromatographic conditions. All of that make the resulted “column parameters”<br />

using these systems deviate from one to another. Calixarene- and resorcinarene-bonded<br />

stationary phases are reversed phases; however they show, depending on the analytes,<br />

some additional interactions, since there steric, polar and ionic properties are different<br />

compared to those of conventional alkyl-bonded phases.<br />

The following table summarizes the parameters used by each system.<br />

System<br />

Tanaka<br />

USP Snyder/Dolan<br />

Parameter<br />

Hydrophobicity<br />

Hydrophobicity<br />

Hydrophobicity<br />

KPB<br />

KEB<br />

H<br />

Hydrophobic<br />

interactions<br />

Hydrophobic selectivity<br />

αCH2 = KPB/ KBB<br />

Hydrophobic selectivity<br />

αCH2 = KEB/ KT<br />

Steric<br />

Shape selectivity<br />

___ Steric resistance<br />

interactions αT/O = KT/ KO<br />

S*<br />

Hydrogen bonding Activity toward bases Hydrogen-bond<br />

Hydrogen<br />

capacity<br />

KAm and TAm<br />

acidity<br />

bonding<br />

αC/P = KC/ KP<br />

A<br />

and<br />

ionic interactions<br />

Acidic ion-exchange<br />

αB/P = KB/ KP (pH 2.7)<br />

Hydrogen-bond<br />

basicity<br />

Chelation<br />

B<br />

Total ion-exchange<br />

TQ<br />

Cation-exchange<br />

αB/P = KB/ KP (pH 7.6)<br />

activity<br />

C<br />

Am: Amitriptyline; B: Benzylamine; BB: n-butylbenzene; C: Caffeine; EB: Ethylbenzene; O: oterphenyl;<br />

P: Phenol; PB: n-pentylbenzene; Q: Quinizarin (1,4-dihydroxyanthraquinone); T:<br />

Triphenylene.<br />

Acknowledgments: Chamseddin, C. has a PhD scholarship from the DAAD and the Syrian ministry of high<br />

education.<br />

References:<br />

1. Kimata, K. et al.: J. Chromatogr. Sci.1989, 27: 721-728.<br />

2. Bidlingmeyer, B. et al.: Pharmacopeial Forum. 2005, 31(2): 637-645.<br />

3. Snyder, L.R. et al.: J. Chromatogr. A. 2004, 1057: 49-57.<br />

049<br />

Structure-based discovery of novel allosteric modulators addressing the<br />

chemokine receptor CXCR3<br />

Tschammer, N. 1; Bernat, V. 1; Schmidt, D. 2; Kolb, P. 2<br />

1 Friedrich Alexander-University Erlangen/Nürnberg, Department of Chemistry and Pharmacy,<br />

Chair of Medicinal Chemistry, Schuhstr. 19, 91052 Erlangen, Germany<br />

2 Philipps University Marburg, Pharmaceutical Chemistry, Wilhelm-Roser-Str. 2, 35032 Marburg,<br />

Germany<br />

Small soluble proteins chemokines and their G protein-coupled receptors are the key<br />

mediators of immune system function in health and disease. Irregularities in the regulation<br />

and response of the chemokine receptor CXCR3 are associated with numerous pathologies<br />

including autoimmune diseases (e.g. multiple sclerosis, rheumatoid arthritis, and psoriasis),<br />

cancer, vascular disease and transplant rejection. Allosteric modulators that inhibit this<br />

attractive pharmacological target could be thus used to treat or prevent a range of<br />

pathologies.<br />

We began the search for novel allosteric modulators addressing the chemokine receptor<br />

CXCR3 with a structure-based virtual screen [1]. Recently resolved X-ray structure of<br />

CXCR4 [2] served us as a template on which a homology model of CXCR3 was build. The<br />

homology model of CXCR3 was further refined using a large library of known CXCR3<br />

ligands in the combination with a set of non-binders. The homology model of CXCR3<br />

characterized by the best docking score for recent clinical candidate AMG487 was chosen<br />

as a basis for virtual screening. Nearly one million of “lead-like” compounds from ZINK, a<br />

free database of commercially available compound for virtual screening, were docked.<br />

From the 500 best ranked after visual inspection, a set of eight compounds was subjected<br />

to biological testing measuring [ 35S]GTPγS incorporation. To facilitate the identification of<br />

novel CXCR3 ligands we synthesized also a tritium labelled allosteric modulator of CXCR3<br />

RAMX3, which was based on an 8-azaquinazolinone core of AMG487 [3]. The study of<br />

allosteric modulators namely needs to differentiate between the ligand affinity toward the<br />

receptor from the cooperativity exhibited towards the endogenous agonist like chemokine,<br />

as the two properties are not correlated [4]. We discovered six novel chemical scaffolds<br />

modulating the function of CXCR3. Interestingly, although we refined the homology model<br />

with negative allosteric modulators of CXCR3, we identified also positive (PAM) and silent<br />

(SAM) besides negative (NAM) allosteric modulators, indicating the delicate balance<br />

between a molecules' function as SAM, NAM or PAM.<br />

Our investigations demonstrate that a carefully refined homology model can provide a<br />

valuable template for the discovery of novel pharmacophores for allosteric modulators even<br />

for such challenging targets as chemokine receptors.<br />

Acknowledgments: N.T. was supported by the German Research Foundation (DFG, TS 287/2-1).<br />

References:<br />

1. Kolb, P. et al.: P. Natl. Acad. Sci. USA 2009, 106(16): 6843-6848.<br />

2. Wu, B. Et al.: Science 2010, 330(6007): 1066-1071.<br />

3. Bernat, V. et al.: ChemMedChem <strong>2012</strong>, Early View, doi: 10.1002/cmdc.<strong>2012</strong>00184.<br />

4. Keov, P. et al.: Neuropharmacology 2011, 60 (1) : 24-35.<br />

050<br />

Structural variations on the thiazolone derivative C06 as potent 5-lipoxygenase<br />

inhibitor<br />

Lill A1, Barzen S1, Rödl C B1, Steinhilber D1, Hofmann B1, Stark H1 1Goethe University, Institute of Pharmaceutical Chemistry, Frankfurt am Main, Germany<br />

(h.stark@zafes.de)<br />

Arachidonic acid released from cytoplasmic membrane upon external stimuli is the source<br />

of one important group of inflammatory mediators: the leukotrienes (LTs). The key enzyme<br />

of their biosynthesis is the iron-containing, heme-free 5-lipoxygenase (5-LO). LTs play a<br />

pivotal role in inflammation, allergic disorders, asthma, cardiovascular diseases and cancer.<br />

Up to now, only two LT-interfering drugs are marketed with limited success: the LT-receptor<br />

antagonist Montelukast (Singulair®) and the iron chelating 5-LO inhibitor Zileuton (Zyflo®).<br />

The need to discover new active ligands for anti-leukotriene therapy is still urgent [1,2].<br />

Based on early virtual screening [3] and medium-throughput screening we previously<br />

identified 5-(4-methoxybenzylidene)-2-p-tolylthiazol-4(5H)-one, C06, as a promising new<br />

direct inhibitor of 5-LO [4,5].<br />

S<br />

N<br />

O<br />

C06 IC = 300nM<br />

50<br />

Poster 105<br />

H3C O<br />

CH 3<br />

R<br />

S<br />

N<br />

O<br />

Our previous studies pointed out that 5-benzylidene-2-phenyl-thiazolinones are a new class<br />

of 5-LO inhibitors. By variation of the substitution pattern we designed and synthesized<br />

novel derivatives of C06 leading to a promising new inhibitors.<br />

The compounds were either prepared by one-pot domino reaction using 2-sulfanylacetic<br />

acid and the corresponding benzaldehyd and benzonitrile or in a two-step synthetic<br />

procedure. All compounds were tested for their 5-LO inhibitory efficiency using a cell-free<br />

and whole-cell (PMNL cells) based assay systems [4,5].<br />

Kindly supported by LOEWE Schwerpunkte LIFF, OSF, NeFF, Anwendungsorientierte<br />

Arzneimittelforschung (Fraunhofer IME) and EU COST Actions CM1103, BM1107, BM0806.<br />

References:<br />

1 M. Peters-Golden and W. Henderson, N. Engl. J. Med. 2007, 357: 1841-1854<br />

2 O. Werz, O and D. Steinhilber, Pharmacol. Ther. 2006, 112: 701-718<br />

3 G. Schneider et al., ChemMedChem 2008, 3: 1535-1538<br />

4 B. Hofmann et al., J. Med. Chem. 2011, 54: 1943-1947<br />

5 S. Barzen et al., Bioorg. Med. Chem. <strong>2012</strong>, 20: 3575-3583<br />

051<br />

Myrrhinil-Intest � chamomile component affects contraction and morphology of<br />

rat ileum/jejunum ex-vivo preparations in a concentration dependent manner<br />

Vissiennon, C. 1; Raszek, M. 1; Goos, K.-H. 2; Nieber, K. 1<br />

1 University of Leipzig, Institute of Pharmacy, Talstraße 33, 04103 Leipzig, Germany<br />

2 REPHA GmbH Biologische Arzneimittel, Alt-Godshorn 87, 30855 Langenhagen, Germany<br />

Myrrhinil-Intest � is a prescribed phytotherapy for the treatment of inflammatory intestinal<br />

diseases. However, how its functional effects are exerted has not yet been fully elucidated.<br />

One of its active ingredients, lyophilised extract of chamomile flowers, was tested on the<br />

acute inflammation of rat ileum/jejunum preparations, experimentally induced by 2,4,6trinitrobenzene<br />

sulfonic acid (TNBS, 10 mM, 30 min). At low concentrations tested (8.75-70<br />

µg/mL) chamomile exhibited a basal tone enhancement of both the untreated controls and<br />

TNBS-treated preparations. At higher concentrations this trend subsided at first with the<br />

inflamed segments only (140 µg/mL), with subsequent inhibition of basal tone for both<br />

untreated and TNBS-treated preparations (210-280 µg/mL).<br />

Histological analysis revealed that the concentration-dependent effect correlated with a<br />

protective function of chamomile from TNBS-induced morphological damage (100 mM, 30<br />

min). Thus whilst pre-treatment with 70 µg/mL exhibited protective function, higher levels of<br />

chamomile (280 µg/mL) did not. 10 mM TNBS also appeared to have 1.6-fold reduction<br />

effect on acetylcholine (ACh)-induced contraction (EC50 0.9 µM vs. 0.58 µM for untreated<br />

controls). This could be reversed with chamomile pre-treatment. However, this observation<br />

was not statistically significant.<br />

The importance of a chamomile treatment concentration-dependence is further<br />

underscored by the fact that TNBS-inflamed segments also exhibit increased sensitivity to<br />

chamomile inhibition of ACh-induced contractions (IC50 106 µg/mL vs. 175 µg/mL for<br />

untreated controls), particularly in the higher concentration range.<br />

052<br />

Four novel antifungal compounds from the Baltic Sea cyanobacterium<br />

Anabaena sp.<br />

Bui, T. H. 1; Wray, V. 2; Nimtz, M. 2; Fossen, T. 3; Schröder, G. 4; Wende, K. 1; Mundt, S. 1<br />

1Institute of Pharmacy, Department of Pharmaceutical Biology, <strong>Ernst</strong>-<strong>Moritz</strong>-<strong>Arndt</strong>-University,<br />

Friedrich-Ludwig-Jahnstr. 17, D-17487 <strong>Greifswald</strong><br />

2Helmholtz Centre for Infection Research, Inhoffenstraße 7, D-38214 Braunschweig, Germany<br />

3Department of Chemistry and Centre for Pharmacy, University of Bergen, N-5007 Bergen, Norway<br />

R


4 Friedrich Loeffler Institute of Medical Microbiology, University Medicine <strong>Greifswald</strong>, Martin-Luther-<br />

Str. 6, D-17475 <strong>Greifswald</strong><br />

18 Vietnamese and 4 German cyanobacterial strains were screened with the agar diffusion<br />

assay for their antifungal activity to human pathogenous fungi. It was found that Anabaena<br />

sp. strain Bio 33 collected from The Baltic Sea (Rügen Island, Germany) exhibited an<br />

excellent and specific antifungal activity with inhibition zones from 21 to 32 mm against<br />

Candida albicans, Candida krusei, Candida maltosa, Aspergillus fumigatus, Microsporum<br />

gypseum, Trichophyton rubrum and Mucor sp. No cytotoxic activity against the human<br />

bladder carcinoma cell line 5637 (IC50 > 500 μg/mL) and no antibacterial activity against<br />

Bacillus subtilis, Staphylococcus aureus, Escherichia coli and Pseudomonas aeruginosa<br />

were observed. Bioassay-guided fractionation of the 50% methanolic water extract by silica<br />

gel column chromatography followed by repeated reversed phase HPLC (column<br />

LiChrospher RP-18, gradient of MeOH in H2O with 0.05% TFA) resulted in four white<br />

amorphous solids. The structures were elucidated by 1D ( 1H) and 2D spectra (COSY,<br />

TOCSY, NOESY, HSQC, HSQC edited TOCSY and gHMBC), HR-ESI-MS and confirmed<br />

by amino acid analysis and sugar analysis. Spectroscopic data analysis afforded an<br />

unambiguous sequence of R.CHO(S1).CHOH.CONH-Thr(1)-Thr(2)-Thr(3)-HOTyr(4)-<br />

DeThr(5)-Gln(6)-Gly(7)-ThrNMe(8)(S2)-GlnCOOH(9) in which S1 is arabinose-(3-1)galacturonic<br />

acid, S2 is a mannose and R is a aliphatic residue. The four compounds<br />

distinguish from their cyclic/linear core and the presence of chlorine in the aliphatic part.<br />

Culture optimization to enhance the yield of these antifungal peptides is currently<br />

developed.<br />

053<br />

Synthesis of inhibitors of glutathione peroxidases for reversal of cancer drug<br />

resistance<br />

Wilde F1; Lemmerhirt H1; Emmrich T1; Schulz R1; Bednarski P J1, Link A1 1 <strong>Ernst</strong>-<strong>Moritz</strong>-<strong>Arndt</strong> University of <strong>Greifswald</strong>, Friedrich-Ludwig-Jahn-Str. 17, 17489 <strong>Greifswald</strong>,<br />

Germany<br />

Glutathione peroxidases (GPx) contribute substantially to cell protection by reducing<br />

reactive oxygen species (ROS) such as H2O2 and organic hydroperoxides and hence<br />

regulating the redox status of cells to prevent cell death [1]. While H2O2 plays a vital role in<br />

the induction of apoptosis [2], over-expression of GPx has been observed in cancer cells<br />

that have acquired resistance to anticancer agents.<br />

Following a combinatorial chemistry approach, a small library of test candidates was built<br />

starting from a lead compound identified by a molecular modeling project [3].<br />

deletion<br />

of<br />

substituent<br />

replacement<br />

of<br />

basic<br />

heterocycle<br />

To determine the activity of the potential inhibitors, a recently established microtiter method<br />

based on the inhibition of bovine erythrocyte GPx was applied, which allowed for a rapid<br />

screening of compounds.<br />

Here we present the general synthesis of the lead compound and the library members.<br />

Characterization of this class of compounds is hampered by the formation of isomers<br />

leading to complex NMR spectra. A rational for this finding is discussed.<br />

References:<br />

1. Takebe, G. et al.: J. Biol. Chem. 2002, 277(43): 41254-8.<br />

2. Lopez-Lazaro, M.: Cancer Lett. 2007, 252(1): 1-8.<br />

3. Schulz, R.: Diploma thesis, 2008.<br />

adjustment<br />

of<br />

spacer length<br />

modification<br />

of<br />

substitution<br />

pattern<br />

variation<br />

of<br />

aromatic system<br />

054<br />

Structural Insights into Retinal Guanylylcyclase / GCAP-2 Interaction<br />

Determined by Cross-Linking and Mass Spectrometry<br />

Jens Pettelkau1; Thomas Schröder2; Christian Ihling1; Björn E. S. Olausson2; Knut Kölbel1; Christian Lange2; Andrea Sinz1 1 Department of Pharmaceutical Chemistry and Bioanalytics, Institute of Pharmacy, Martin-Luther<br />

University Halle-Wittenberg, Wolfgang-Langenbeck-Strasse 4, 06120 Halle-Saale, Germany<br />

2 Department of Technical Biochemistry, Institute of Biochemistry and Biotechnology, Martin-Luther<br />

University Halle-Wittenberg, Kurt-Mothes-Strasse 3, 06120 Halle-Saale, Germany<br />

Two isoforms of retinal guanylylcylase (ROS-GC 1 and 2), localized in photoreceptor cells,<br />

are responsible for the synthesis of cGMP, the second messenger in phototransduction [1].<br />

ROS-GC 1 and 2 are regulated in a Ca2+-dependent manner from the intracellular site by<br />

guanylylcyclase-activating proteins (GCAPs) [2]. GCAPs are hydrophobic N-terminally<br />

myristoylated proteins containing four EF-hand motifs, which are responsible for Ca2+ sensitivity [3]. Genetic disorders in GCAP genes or parts of the regulatory regions in ROS-<br />

GC genes lead to different retinopathies [4]. Therefore, a detailed knowledge of the<br />

complex formation between GCAPs and ROS-GC is crucial for the development of novel<br />

drugs.<br />

Interactions between a ROS-GC 1 peptide (amino acids 965-981 from the catalytic domain<br />

[5]) and GCAP-2 were investigated applying two complementary types of cross-linking<br />

chemistry. For mapping larger distances, the homobifunctional amine-reactive cross-linker<br />

bis(sulfosuccinimidyl)glutatrate (BS2G) was used. To determine spatially close interactions,<br />

the unnatural photoreactive amino acid photo-leucine that is activated by UV-A radiation,<br />

was incorporated into ROS-GC peptides. After the cross-linking reaction the cross-linked<br />

GCAP-2/GC peptide complexes were enzymatically digested and analysed by highresolution<br />

mass spectrometry.<br />

In the presence of Ca2+, several intramolecular cross-links were identified within N-<br />

terminally myristoylated GCAP-2, all of which were in agreement with the published NMRstructure<br />

(pdb 1JBA) of non-myristoylated GCAP-2. The C-terminal part of GCAP-2, which<br />

is not resolved in the NMR-structure, seems to be highly flexible as indicated by the<br />

numerous intramolecular cross-links within GCAP-2. Analysis of cross-linking samples<br />

without Ca 2+ yielded a limited number of intramolecular cross-linking products of GCAP-2<br />

itself. Several cross-links between the N-terminus of GC peptide and different lysines of<br />

GCAP-2 were identified both in the presence and absence of Ca 2+. After photo-crosslinking,<br />

a limited number of cross-links were obtained pointing to a structurally well-defined<br />

interaction between GCAP-2 and the GC peptide in the presence of Ca 2+. In the absence of<br />

Ca 2+ the structure of GCAP-2 appeared to be much more flexible as indicated by a large<br />

number of diverse photo cross-linking products between GCAP-2 and GC peptide. Based<br />

on the distance contraints imposed by the cross-links it was possible to create a model of<br />

the complex between myristoylated GCAP-2 and the GC peptide that is formed in the<br />

presence of Ca 2+ (Figure 1).<br />

Figure 1: Structural model of the GCAP-2/GC peptide complex. The backbone of the most<br />

representative GC peptide is colored red along with GCAP-2, which is shown as a light blue<br />

cartoon. Cross-linking sites in GCAP-2 obtained from photoaffinity labeling, which were<br />

used for modeling, are shown as light green spheres. Cross-linking sites obtained with<br />

BS2G, which were used for modeling, are shown as light orange spheres. The reactive<br />

sites in the GC peptide are shown as dark green (photo-Leu at positions 5 and 13) and<br />

brown (Tyr-1) spheres.<br />

References:<br />

1. Yang R.B. et al.: Proceedings of the National Academy of Science of the United States of America 1995,<br />

92(2): 602-606.<br />

2. Sharma R.K.: Mol.Cell. Biochem. 2010, 334(1-2): 3–36.<br />

3. Olshevskaya E.V. et al.: J. Biol. Chem. 1997, 272(22): 14327-14333.<br />

4. Sato M. et al.: Graefes Arch. Clin. Exp. Ophthalm. 2005, 243(3): 235-242.<br />

5. Duda T. et al.: Biochemistry 2005, 44(19): 7336-7345.<br />

055<br />

Triterpenes from the fungus Piptoporus betulinus<br />

Alresly, Z. 1; Lindequist, U. 1; Lalk, M. 1; Porzel, A. 2; Arnold, N. 2 ; Wessjohann, L. A. 2<br />

1Institute of Pharmacy, <strong>Ernst</strong>-<strong>Moritz</strong>-<strong>Arndt</strong>-University, Friedrich-Ludwig-Jahn-Straße 17, D-17489<br />

<strong>Greifswald</strong>, Germany<br />

2Leibniz – Institute of Plant Biochemistry, Department of Bioorganic Chemistry (NWC), Weinberg 3,<br />

06120 Halle (Saale , Germany<br />

The mushroom Piptoporus betulinus (Bull. : Fr.) P. Karst. (Polyporaceae), birch polypore,<br />

growth as trunk parasit and saprophyt on Betula pendula Roth. and B. pubescens Ehrh.<br />

(Betulaceae). Fruit bodies of this fungus are used in the European ethnomedicine for the<br />

treatment of cancer and stomach diseases. The mushroom is also known as fungus of the<br />

“iceman” from the Copper Age found in 1991, who carried P. betulinus fruiting bodies<br />

attached to his clothing on his journey in the Alps [1].<br />

The phytochemical knowledge about this mushroom and the justification of its<br />

ethnomedicinal use are limited. Aim of our studies was the phytochemical examination of<br />

extracts prepared from the fruiting bodies of collected mushrooms and the investigation of<br />

isolated compounds for antimicrobial activity.<br />

Fractionation of the ethyl acetate extract led to the isolation of a new bioactive triterpene<br />

with a lanostane basic structure. By the help of spectroscopic methods it could be identified<br />

as 3β-acetoxy-16 hydroxy-24- oxo-5a- lanosta- 8(9) diene -30- oic acid (Fig. 1). In addition,<br />

seven known triterpenes, polyporenic acid C, betulinic acid, betulin, ergosterol peroxide,<br />

9,11dehydroxyergosterol peroxide, 24-methylene-3-oxo-lanosta-8-en-21-oic acid, and<br />

fomefficinic acid, could be isolated and identified by comparison with data from the<br />

literature. All isolated compounds were tested for antimicrobial activity against some Grampositive<br />

and Gram-negative bacteria as well as against some microscopic fungi. The new<br />

triterpene, polyporenic acid C, and fomefficinic acid showed antimicrobial activity against<br />

Staphylococcus aureus.<br />

106 Poster<br />

CH 3<br />

CH3 OH<br />

CH3 CH3 CH3 O O<br />

H3C CH3 Fig. 1: 3β-acetoxy-16α hydroxyl-24-oxo-5α-lanosta-8(9) diene-30-oic acid<br />

References:<br />

1. Lindequist, U.; Niedermeyer, T.H.J.; Jülich, W.D.: eCAM 2005, 2(3): 285–299.<br />

056<br />

A1-barrigenol and rare 17,22-seco oleanolic acid glycosides from the leaves of<br />

Pittosporum angustifolium.<br />

Bäcker, C. 1; Jenett-Siems, K. 2; Siems, K. 3; Wurster, M. 1; Bodtke, A. 4; Chamseddin, C. 4;<br />

Crüsemann, M. 1a; Lindequist, U. 1<br />

1 Department of Pharmaceutical Biology, Institute of Pharmacy, <strong>Ernst</strong> <strong>Moritz</strong> <strong>Arndt</strong> University<br />

<strong>Greifswald</strong>, Friedrich-Ludwig-Jahn-Str. 17, 17489 <strong>Greifswald</strong>, Germany<br />

2 Department of Pharmaceutical Biology, Institute of Pharmacy, Free University of Berlin, Königin-<br />

O<br />

OH<br />

O<br />

CH 3


Luise-Str. 2+4, 14195 Berlin, Germany<br />

3 AnalytiCon Discovery GmbH, Hermannswerder Haus 17, 14473 Potsdam, Germany<br />

4 Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, <strong>Ernst</strong> <strong>Moritz</strong> <strong>Arndt</strong><br />

University <strong>Greifswald</strong>, Friedrich-Ludwig-Jahn-Str. 17, 17489 <strong>Greifswald</strong>, Germany<br />

1a Present address: Kekulé Institute of Organic Chemistry and Biochemistry, Rheinische Friedrich<br />

Wilhelms University Bonn, Gerhard-Domagk-Str. 1, 53121 Bonn, Germany<br />

Pittosporum angustifolium LODD. (Pittosporaceae), a small tree known colloquially as<br />

"gumby gumby", occurs in inland areas of most states of Australia. Especially the leaves<br />

are used by Aboriginals for various medical applications [1]. Recently beneficial effects of<br />

“gumby gumby” as a supportive agent for cancer treatment have been observed [2].<br />

Phylogenetic studies described P. angustifolium as a distinct species, which previously was<br />

considered as a variety of P. phillyreoides (syn. P. phillyraeoides; var. microcarpa) [1].<br />

Phytochemical investigation of the leaves collected in Queensland, Australia, resulted in the<br />

isolation of triterpene saponins known as eryngioside I [3] and new compounds named<br />

pittangretosides A-I. Their structures were elucidated and characterized by NMR,<br />

HRESIMS, hydrolysis and CD measurement. Eight saponins were identified as<br />

A1-barrigenol derivatives whereas two compounds exhibit an unusual 17,22-seco backbone<br />

of oleanolic acid.<br />

Acknowledgments: We sincerely thank Dr. Rudolf Kunze, Berlin, Germany, for the stimulation of this work<br />

and his continuous support as well as Dr. Cornelia Krasser, Yepoon, Australia and Mr. Klaus von<br />

Gliszczynski for the collection and provision of the plant material. We also thank all of the colleagues at the<br />

Institute who were involved in supporting this work.<br />

References:<br />

[1] Cayzer, L.W., Crisp, M.D., Telford, R.H., Aust. Syst. Bot. 2000, 13: 845-902.<br />

[2] Patent, 2009. WO 2009/037225 A2; DE 10 2007 044 094 A1.<br />

[3] Zhang, Z. et al., Phytochemistry 2008, 69: 2070-2080.<br />

057<br />

Homology Modeling of the Kinase Domain of DLK<br />

Hasenpusch, D. 1; Oetjen, E. 2; Lemcke, T. 1<br />

1 Institut für Pharmazie, <strong>Universität</strong> Hamburg, Bundesstr. 45, 201146 Hamburg, Germany<br />

2 Institut für Klinische Pharmakologie und Toxikologie, <strong>Universität</strong>sklinikum Eppendorf, Martinistr.<br />

52, 20246 Hamburg, Germany<br />

An interesting subgroup of protein kinases are the mixed-lineage kinases (MLKs). They<br />

belong to the tyrosine kinase-like kinases (TKL) and show phylogenetic similarity to both<br />

typical serine/threonine kinases and tyrosine kinases. Functional they are a member of the<br />

serine/threonine kinases.<br />

Two interesting members of this group are the DLK (dual leucine zipper kinase) and LZK<br />

(leucine zipper kinase), which form homodimers mediated via hydrophobic interactions of<br />

their leucine zippers. Acting as a MAP3K DLK was shown to result in the activation of the<br />

downstream kinases p38 and JNK (jun-N-terminal kinase). In the insulin producing β-cells<br />

within the pancreatic islets DLK was shown to mediate apoptosis induced by<br />

immunosuppressants and by the cytokine TNFα and to inhibit insulin gen transcription [1–<br />

4]. Given the importance of maintaining β-cell function and mass to prevent the<br />

pathogenesis of diabetes mellitus, the inhibition of DLK represents an interesting and novel<br />

target for developing potential drugs for the therapy of this chronic disease.<br />

So far no crystal structure data of the DLK is available. As a starting point for a structure<br />

based project to identify potential selective DLK inhibitors, we constructed threedimensional<br />

homology models of the kinase domain of DLK. Suitable templates, which<br />

represented different conformations of the kinases, namely the open and closed<br />

conformation of the activation loop, were found in the phylogenetic neighborhood of DLK.<br />

The actual model building was accomplished with MODELLER [5] using standard<br />

procedures and subsequent loop refinement by the loop model routine. Models were<br />

afterwards evaluated using a workflow that combines individual scorings of different<br />

evaluation algorithms (e.g. PROCHECK, ProSA, ERRAT, MODELLER) in a rank-by-rank<br />

ConsensusScoring [6]. Top ranking models were submitted to molecular dynamic<br />

simulations of at least 2 ns, to test for general stability and to allow assuming their final<br />

three-dimensional shape. These models will be used in a future virtual screening<br />

experiment for the search of DLK inhibitors.<br />

References:<br />

[1] Plaumann, S. et al., Mol. Pharmacol. 2008, 73, 652–659.<br />

[2] Klimpel, C., Boerchers, S., Oetjen, E., Arch Pharmacol. 2009, 379, 37-37.<br />

[3] Oehmen, M.-J. et al., Arch. Pharmacol. 2010, 381, 36-36.<br />

[4] Phu do, T. et al. Cell Signal. 2011, 23, 344-353.<br />

[5] Sali, A., Blundel, T. L., J. Mol. Biol. 1993, 234, 779–815.<br />

[6] Kruggel, S., Lemcke, T., Arch. Pharm. (Weinheim) 2009, 342, 327–332.<br />

058<br />

ChemicalToolBoX in action!<br />

Grüning, B. A. 1; Lucas, X. 1; Chbeib, M. 1, Patel, H. 1, Günther, S. 1<br />

1 Institute of Pharmaceutical Sciences, Hermann-Herder-Strasse 9, 79104, Germany<br />

ChemicalToolBox is a set of tools integrated into a workflow-management system to enable<br />

researchers easy-to-use, reproducible, and transparent access to cheminformatic libraries<br />

and drug discovery tools. It includes standard applications for similarity and substructure<br />

searches, clustering of compounds, prediction of properties and descriptors, filtering, and<br />

many other tools that range from drug-likeliness classification to fragmentation and<br />

fragment-merging.<br />

ChemicalToolBox is based on open-source software, web-accessible, freely available, and<br />

easily expandable. It can be downloaded and easily deployed locally or on a cluster.<br />

Here we present some use-cases for the ChemicalToolBox that we have developed in our<br />

lab to demonstrate its capabilities.<br />

As a first application we have created ChemicalBoX, a comprehensive library of about 40<br />

million unique small compounds collected from freely-accessible repositories. The<br />

compounds included in ChemicalBoX have been filtered by physico-chemical properties<br />

and clustered by similarity to allow fast navigation throughout millions of compounds<br />

belonging to many different chemical classes.<br />

In a similar project, called PurchasableBoX, a comprehensive library consisting of several<br />

millions of purchasable small compounds was assembled. Both libraries are unique and, to<br />

the best of our knowledge, are the largest freely-available libraries.<br />

With the fragmentation tool included in ChemicalToolBox, we can build specialized<br />

fragment libraries using databases of compounds as input. Those collections provide the<br />

foundation for calculating focused Mass Spectrometry libraries and for enabling fast and<br />

efficient screening of large amounts of MS data. Furthermore, these fragments can be the<br />

input of a workflow we have created for fragment-based drug design: it merges the<br />

fragments according to predefined synthetic route rules to form new promising compounds<br />

that enlarge the known chemical space.<br />

The use cases presented here provide an insight into the capabilities of ChemicalToolBoX.<br />

By combinations of the various tools many more powerful applications can be designed.<br />

059<br />

The influence of saponins on the cell membrane-cholesterol<br />

Böttger, S. 1, Melzig, M. F. 1<br />

1 Institute of Pharmacy – Pharmaceutical Biology, Freie <strong>Universität</strong> Berlin, Königin-Luise-Str. 2+4,<br />

14195 Berlin, GERMANY<br />

As secondary plant compounds with amphiphilic properties saponins possess surface<br />

activity. They can interact with physical surfaces such as those of aqueous solutions and<br />

lower the surface tension [1]. They also interfere with biologic surfaces such as cell<br />

membranes. The latter case may result in haemolysis and cytotoxicity as a consequence of<br />

perturbation of the membrane integrity at higher saponin concentrations.<br />

The mechanism behind this saponin characteristic is not yet completely understood, but<br />

there are different indications that the membrane-cholesterol is decisively involved in this<br />

process. Saponins have been described to bind/to complex cholesterol [1, 2, 3]. As<br />

cholesterol is known to carry out a cell membrane stabilizing function [4], a loss of the<br />

cholesterol itself or the loss of the ability to carry out this function will lead to cell membrane<br />

instabilities and/or membranolysis.<br />

We developed a cell culture model using ECV-304 urinary bladder carcinoma cells<br />

involving 3H-marked cholesterol and investigated the influence of different types of<br />

saponins on the cell membrane-cholesterol.<br />

References:<br />

1. Böttger, S., Hofmann, K., Melzig, M. F.: Bioorg. Med. Chem. <strong>2012</strong>, 20(9): 2822-2828.<br />

2. Glauert, A. M., Dingle, J. T., Lucy, J. A.: Nature 1962, 196(4858): 953-955.<br />

3. Schlösser, E.: Can. J. Physiol. Pharmacol. 1969, 47(5), 487-490.<br />

4. Meyer, T. et al.: Biospektrum, <strong>2012</strong>, 18(2), 142-145.<br />

060<br />

Synthesis and testing of novel 2,4-thiazolidinedione derivatives as inhibitors of<br />

the enoyl-ACP-reductase InhA of Mycobacterium tuberculosis<br />

Vogel, S. 1; Wagner, S., Waltenberger, C., Sotriffer, C.A., Schirmeister, T. 2<br />

1 Institute of Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, 97074<br />

Würzburg , Germany<br />

2 Institute of Pharmacy and Biochemistry, University of Mainz, Staudinger Weg 5, 55128 Mainz,<br />

Germany<br />

InhA is part of the fatty acid synthase complex type II (FAS II) and catalyzes the reduction<br />

of enoyl-acyl-[acyl-carrier-protein] derivatives of the chain length up to 56 carbons using the<br />

essential cofactor NADH.[1] When inhibited, unsaturated fatty acids accumulate, and the<br />

formation of the cell wall is disturbed.<br />

Computational methods have been applied in a step-wise fashion in order to search for new<br />

InhA inhibitors. Using ligand- and target-based approaches three pharmacophore models<br />

were defined. For screening purposes the ‘drug-like’ subset of the ZINC database and the<br />

vendor database distributed by CCG were used. Hits of the pharmacophore screening were<br />

subdued to hierarchical filtering and visually inspected. The most suitable candidates were<br />

selected for docking procedures with the programs MOE Dock and Autodock 3.0. For<br />

validation purposes, known active compounds were included in the docking simulations.<br />

The most promising hits were chosen for further studies and experimental testing of their<br />

inhibitory activity.<br />

For activity testing two enzyme assays based on the decrease of the absorption or<br />

fluorescence intensity upon reduction of the cofactor NADH were established and validated<br />

using triclosan as positive control. The enzyme InhA was overexpressed in E. coli and<br />

purified.[2] The substrate 2-trans-octenoyl-coenzyme A was synthesized via the mixed<br />

anhydride method.[3]<br />

Based on the results of the screening of a large library of compounds and subsequent<br />

docking experiments the most promising hits with 2,4-thiazolidinedione structure were<br />

chosen for further optimization.<br />

References:<br />

1. X.Hong et al.: Biomacromolecules 2004, 5: 1052-1065.<br />

2. Parikh et al.: Biochemistry 1999, 38: 13623-13634.<br />

3. Goldman/Vagelos: J.Biol.Chem. 1961, 10: 2620-2623.<br />

061<br />

3-Hydroxypyrroles or Pyrrolin-3-ones as novel p38 MAP kinase inhibitors<br />

Freitag, A. 1; Laufer, S. A. 1<br />

1 Institute of Pharmacy, Department of Pharmaceutical and Medicinal Chemistry, Eberhard-Karls-<br />

<strong>Universität</strong>, Auf der Morgenstelle 8, 72076 Tübingen, Germany<br />

p38 MAP kinase controls a plethora of cellular responses to extracellular stimuli such as<br />

cellular stress. The kinase is part of a signalling pathway resulting in the biosynthesis of<br />

pro-inflammatory cytokines like IL-1β and TNF-α. Since their levels are elevated in<br />

chronical inflammatory diseases inhibition of p38 is a promising therapeutic target.<br />

The small molecule inhibitor SB-203580 is one of the first known selective inhibitors of p38.<br />

Poster 107


Due to X-ray crystal strucure analysis, the interactions of the pyridinylimidazole to the<br />

binding site of the kinase are established [1].<br />

Figure 1: left side: binding mode of SB-203580; right side: postulated binding mode of<br />

pyrrolin-3-ones<br />

Being iron-ligands, imidazoles lead to CYP-450 inhibition and probably hepatotoxicity. Our<br />

strategy is to replace the core-heterocycle in order to reduce hepatic toxicity and to improve<br />

the interaction with Lys53. These considerations led us to pyrrolin-3-ones.<br />

Since established synthetic approaches to build pyrroles don’t suit this substitution pattern<br />

a new route has been developed based on reference [2].<br />

One major challenge is tautomerism of the target compound. Depending on substituents<br />

and solvent 3-hydroxypyrrole and pyrrolin-3-one exist at equilibrium.<br />

In position 2 unsubstituted pyrrolinones are extremely tending to polymerize, but the 3hydroxypyrrole<br />

with an ethyl carboxylate substituent in position 2 could successfully be<br />

crystallized and tested in a p38α MAP kinase assay (IC50 0,139 µM).<br />

Acknowledgments: Katharina Bauer and Marcia Goettert for biological testing, Verena Schattel for molecular<br />

modelling.<br />

References:<br />

1. Gallagher, T. F. et al.: Bioorg. & Med. Chem. 1997, 5(1): 49–64.<br />

2. Bauer, H.: Justus Liebigs Annalen der Chemie 1970, 736:1-15.<br />

062<br />

On the Importance of Water Molecules on Ligand Binding in the ATPase of<br />

GyrB<br />

Münsterberg, M.; Lemcke, T.<br />

Institut für Pharmazie, <strong>Universität</strong> Hamburg, Bundesstr. 45, 20146 Hamburg, Germany<br />

Gyrase is a well-known ATP-dependent topoisomerase II, essential for prokaryotic cells. It<br />

is the only known topoisomerase, which catalyses the negative supercoiling of DNA and<br />

consists of two subunits GyrA and GyrB. While GyrA mainly processes the strand passage,<br />

the ATP-hydrolysis is done by the N-terminus of GyrB [1]. Because of its significance in<br />

prokaryotes, gyrase has become a widely used target in antibiotic therapy. Nevertheless,<br />

only fluoroquinolones, inhibitors of the DNA-GyrA-complex, are available for therapy.<br />

Although there are known inhibitors of the ATPase in GyrB, e. g. the aminocoumarines like<br />

novobiocin, there are no drugs on the market which use this target [2]. Meanwhile, the<br />

resistance of bacteria against all major antibiotics forces the development of new antibiotics<br />

with superior effects [3].<br />

Several x-ray crystal structures of GyrB complexes from the protein data bank [4], with<br />

chemically different ligands, contain conserved water molecules. It is well known that water<br />

molecules play an important role for the stability of protein-ligand complexes, because the<br />

presence of water in the ligand binding site can increase or weaken the binding affinity of a<br />

ligand [5,6]. The release of buried water molecules from hydrophobic binding site<br />

environments very often has a beneficial effect on the free energy of binding of an inhibitor.<br />

For a structure based virtual screening it is necessary to know, which water molecule is<br />

important for the complex stability and ligand binding, and which water molecule might be<br />

replaced during docking experiments.<br />

Through the investigation of the GyrB ATPase protein-ligand complexes with different<br />

methods and tools (MD simulations, SuperStar [7] and Rank and HINT [8]) we have<br />

assessed the water molecules in the ATP-binding site. Even though there are huge<br />

conformational differences within the structures, particular water molecules are necessary<br />

for ligand binding in all complexes and thus for successful docking. Other, also conserved,<br />

water molecules in hydrophobic environment might be replaced by a suitable ligand to<br />

perform a successful docking.<br />

Our results will be the foundation for the setup of a virtual high throughput screening in a<br />

structure based design campaign to identify new and selective GyrB ATPase inhibitors.<br />

References:<br />

1. Champoux J. J.: Annu. Rev. Biochem. 2001, 70(1): 369–413.<br />

2. Collin F., Karkare S., Maxwell A.: Appl. Microbiol. Biotechnol. 2011, 92(3): 479–497.<br />

3. Boucher H. W. et al.: Clin. Infect. Dis. 2009, 48(1): 1–12.<br />

4. Berman H. M. et al.: Nucleic Acids Res. 2000, 28(1): 235–242.<br />

5. Lam P. Y. et al.: Science 1994, 263(5145): 380–384.<br />

6. Mikol V., Papageorgiou C., Borer X.: J. Med. Chem. 1995, 38(17): 3361–3367.<br />

7. Verdonk M. L., Cole J. C., Taylor R.: J. Mol. Biol. 1999, 289(4): 1093–1108.<br />

8. Amadasi A. et al.: J. Med. Chem. 2008, 51(4): 1063–1067.<br />

063<br />

Natural surfactant of polyglycerol fatty acid esters type as stabilizers for lipid<br />

nanoparticles<br />

Keck CM1 2; Kovacevic A3; Müller RH1 1Institute of Pharmacy, Department of Pharmaceutics, Biopharmaceutics & NutriCosmetics, Freie<br />

<strong>Universität</strong> Berlin, Kelchstrasse 31, 12169 Berlin, Germany<br />

2Applied Pharmacy Division, University of Applied Sciences Kaiserslautern, Campus Pirmasens,<br />

Carl-Schurz-Strasse 10-16, 66953 Pirmasens, Germany<br />

3Faculty of Pharmacy, Department of Pharmaceutical Technology and Cosmetology, University of<br />

Belgrade, Vojvode Stepe 450, 11000 Belgrade, Serbia<br />

Polyglycerol fatty acid esters (PGE) are non-ionic surfactants with various applications in<br />

pharmaceutical and cosmetic industries. They have been used for many years as food<br />

additives and therefore designed as „orally safe” surfactants. The aim of the study<br />

presented here was to evaluate the performance of PGE as potential stabilizer for SLN and<br />

NLC. PGE may be formed by a polymerization of glycerol followed by an esterification with<br />

fatty acids. The obtained mixture might vary in polymerization degree, kind and position of<br />

esterified fatty acids. The surfactants investigated were differed in the kind and position of<br />

esterified fatty acid (Fig. 1).<br />

SLN composed of 10% Cutina ® CP as solid lipid and NLC containing 6% Cutina ® CP and<br />

4% Miglyol ® 812 as a liquid lipid were produced by hot high pressure homogenization [1].<br />

All samples were stabilized with 1% PGE. Characterization of the particles was performed<br />

by photon correlation spectroscopy (PCS) and laser diffraction (LD). To investigate the<br />

surface properties of the SLN and NLC zeta potential (ZP) was measured in distilled water<br />

adjusted to conductivity of 50 μScm −1. Crystallinity of the SLN and NLC and respective solid<br />

lipid i.e. solid lipid/liquid lipid blend used as a particle matrix was investigated by differential<br />

scanning calorimetry (DSC).<br />

The obtained results indicated that PGE showed a good efficiency in the stabilization of the<br />

SLN and NLC. All samples have revealed size (z-ave) below 200 nm, with narrow size<br />

distribution (polydispersity index < 0.15). The respective z-ave were in good agreement<br />

with LD diameters indicating narrow and unimodal size distribution. PGE based SLN and<br />

NLC possessed the same particle composition, but different crystallinity indices. Particles<br />

stabilized with PI and PDI possessed lower crystallinity than PS based samples. The<br />

differences in DSC parameters are obviously created by the different types of the<br />

surfactants i.e. different interaction with the particle matrix and different surface tension<br />

generated near to the surface of the particles. The differences in the internal structure of<br />

the particles were also reflected in ZP values. With higher melting enthalpy and higher<br />

crystallinity index higher ZP values were obtained.<br />

Conclusions: PGE are suitable stabilizers for SLN and NLC. However, the tendency of the<br />

lipid (used as a particle matrix) to create more crystalline matrix of the lipid nanoparticles<br />

was favored by polyglycerol-6 ester type of the surfactant (PD) whereas the influence of the<br />

polyglycerol-6 isoesters (PI, PDI) was not significant.<br />

A B<br />

C<br />

Chemical structures of the PGE-surfactants used: A- polyglycerol 6-distearate (PD), B-<br />

polyglycerol 6-diisostearate (PDI) and C- polyglycerol 6-monoisostearate (PI)<br />

References:<br />

1. Müller RH, Lucks JS. Arzneistoffträger aus festen Lipidteilchen - Feste Lipid Nanosphären (SLN). 1991,<br />

German patent application P 41 5 62.6.<br />

064<br />

Lectine Mimetics Based on Multivalent Boronic Acids<br />

Claes D1; Maison W1 1 Pharmaceutical and Medicinal Chemistry, Bundesstraße 45, 20146 Hamburg, Germany<br />

Carbohydrates are playing an essential role in biological systems. They are used for energy<br />

storage, as structural substances and for cellular recognition [1]. The so called glycocalix<br />

covers cells and its molecular structure is dependent on the cell type, its level of<br />

differentiation and the activity of carbohydrate modifying enzymes. The recognition of these<br />

differences is a key component of pathogen detection by the native immune system and<br />

makes carbohydrates promising targets for pharmaceuticals [2].<br />

Natural carbohydrate binders are lectins, the first line of defense in our immune system [3].<br />

Lectins show weak affinities to monosaccharides and are therefore binding carbohydrates<br />

multivalently to strengthen these interactions. The geometry, composition and density of<br />

binding epitopes has a profound influence on these binding processes [4]. Notably, many<br />

lectins recognize their carbohydrate ligands in C3-symmetric complexes.<br />

108 Poster<br />

HO<br />

B<br />

O<br />

peptide<br />

O<br />

NH<br />

B O<br />

HO<br />

scaffold<br />

HN O<br />

peptide<br />

O<br />

N<br />

H<br />

boronolectin s<br />

peptide<br />

O<br />

B OH<br />

We propose to mimic lectins with trivalent assemblies of boronic acids. For this purpose we<br />

have synthesized suitable trivalent scaffolds and benzboroxoles, which are known to bind<br />

to non reducing sugars with significant affinities [5].


References:<br />

1. Roseman S. J. Biol. Chem. 2001, 276(45): 41527–42542.<br />

2. Dube D. H., Bertozzi C. R. Nat. Rev. Drug. Dis. 2005, 4(6): 477–488.<br />

3. Lis H., Sharon N. Chem. Rev. 1998, 98(2): 637–674.<br />

4. Kiessling L. L., Gestwicki J. E., Strong L. E. Curr. Opin. Chem. Biol. 2000, 4(6): 696–703.<br />

5. Berube M., Dowlut M., Hall D. G. J. Org. Chem. 2008, 73(17): 6471–6479.<br />

065<br />

Heterologous expression of (putative) polyketide synthases and nonribosomal<br />

peptide synthetases in Bacillus subtilis<br />

Kumpfmüller, J. 1; Kabisch, J. 1; Wenzel, S. 2; Müller, R. 2; Pfeifer, B. 3; Nagel, S. 1; Hiddemann,<br />

L. 1; Lalk, M. 1 Schweder, T. 1<br />

1Institut für Pharmazie, <strong>Universität</strong> <strong>Greifswald</strong>, F.-L.-Jahnstr.17, 17489 <strong>Greifswald</strong>, Germany<br />

2Department of Chemical and Biological Engineering, Science and Technology Center, Tufts<br />

University, 4 Colby Street, Medford, MA 02155, USA<br />

3Institut für Pharmazeutische Biotechnologie, <strong>Universität</strong> des Saarlandes, <strong>Universität</strong>scampus,<br />

Gebäude C2 3, 66123 Saarbrücken, Germany<br />

Polyketides and non-ribosomal peptides constitute an important class of complex natural<br />

products with wide-ranging therapeutic value. They are produced by multifunctional megaenzymes,<br />

the polyketide synthases (PKSs) and the non-ribosomal peptide synthetase<br />

(NRPS), respectively. Looking for new drugs, especially from unculturable microorganisms,<br />

as many marine bacteria are, heterologous expression of putative PKS and NRPS clusters<br />

becomes more and more important. Hence, there is a need for new hosts that can serve as<br />

alternatives to existing expression systems.<br />

In this study, we investigated Bacillus subtilis’s ability to express heterologous<br />

PKSs/NRPSs. First three putative PKSs/NRPSs (RB386, RB6500 and RB11975) from the<br />

marine planctomycete Rhodopirellula baltica were successfully integrated into the<br />

chromosome of B. subtilis. Although all of the recombinant strains showed positive mRNA<br />

results, overexpression of the heterologous protein could be detected and verified via MS<br />

only for RB11975. HPLC-MS-analyses of cell and medium extracts are being used to<br />

identify specifically produced metabolites.<br />

Beside this, we chose the PKS deoxyerythronolide B synthase (DEBS) as a model gene<br />

cluster since it was already successfully expressed by the heterologous hosts Escherichia<br />

coli and Streptomyces coelicolor. The product, 6-deoxyerythronolide B (6dEB), is the<br />

polyketide core of the antibiotic erythromycin. Due to the complexity of the 30kb large<br />

DEBS-cluster it was necessary to develop a new method for its stepwise chromosomal<br />

integration.<br />

To optimize our expression host for the production of polyketides and non-ribosomal<br />

peptides we performed a knockout of two large clusters: the NRPS-operon srfA (26kb,<br />

coding for surfactin synthetase) and the PKS-cluster pksX (78kb, coding for bacillaene<br />

synthase). Hence, we hope to provide more substrates for the heterologous PKS/NRPS<br />

and to reduce the background for an enhanced detection of produced metabolites.<br />

Metabolite analyses of the mutant strains are being used to elucidate the effect of these<br />

mutants on the physiology of B. subtilis.<br />

066<br />

Fed Stomach Model – a novel test device for biorelevant simulation of<br />

intragastric drug dissolution after food intake<br />

Koziolek, M. 1; Görke, K. 1; Garbacz, G. 1, Weitschies, W. 1<br />

1 <strong>Ernst</strong>-<strong>Moritz</strong>-<strong>Arndt</strong>-<strong>Universität</strong> <strong>Greifswald</strong>, Institut für Pharmazie, Biopharmazie &<br />

Pharmazeutische Technologie C_DAT Center of Drug Absorption and Transport, Felix-Hausdorff-<br />

Straße 3, D-17487 <strong>Greifswald</strong><br />

Food ingestion is known to alter physicochemical as well as mechanical conditions in the<br />

upper gastrointestinal (GI) tract. Consequently, drug release from solid oral dosage forms<br />

can be affected, which may have an impact on drug plasma levels by changing one or more<br />

pharmacokinetic parameters. To predict food effects on drug release in the human<br />

stomach, we designed an in vitro dissolution device for biorelevant simulation of fed gastric<br />

conditions, the Fed Stomach Model (FSM). This study aimed at the examination of the<br />

technical functionality of the FSM and the evaluation of the individual effects of the factors<br />

applied.<br />

The construction of the FSM is based on USP paddle apparatus. It is equipped with six<br />

specially designed dissolution cells. Media circulation between dissolution cells and the<br />

vessels of USP paddle apparatus is provoked by a peristaltic pump. By the use of stepping<br />

motor and valve system, both controlled via custom-made computer software, the FSM<br />

enables the simultaneous simulation of predefined movement profiles, flow and pressures.<br />

These parameters are exerted by a central axis driven by the stepping motor and<br />

connected to a pressure regulation module. In the present study, the drug release from<br />

diclofenac extended release (ER) tablets in USP phosphate buffer pH 6.8 was investigated<br />

to examine the individual relevance of the factors applied. In this study, the FSM was<br />

operated in closed-loop mode and the amount of drug dissolved was measured on-line by<br />

fiber optics.<br />

It could be demonstrated with the aid of magnetically labeled tablets that the FSM was able<br />

to simulate intragastric transport velocities equivalent to in vivo data generated by our<br />

group [1]. Furthermore, a valve system connected to a pressure regulation module allowed<br />

the application of biorelevant pressures by air inflation or deflation of a balloon. In<br />

preliminary dissolution experiments, we demonstrated that drug release from diclofenac ER<br />

tablets depended on dosage form movement as well as on pressure. It is thus feasible to<br />

apply test algorithms specific for different gastric regions after food intake.<br />

The Fed Stomach Model can be a valuable tool for biorelevant dissolution testing due to its<br />

potential for precise and reproducible simulation of mechanical parameters present under<br />

fed gastric conditions. Media flow including secretion and emptying, movement patterns<br />

and pressures were integrated into test algorithms representing specific conditions in<br />

fundus and antrum. Thus, the intragastric location of solid dosage forms can be considered<br />

in the FSM. This enables a more realistic in vitro investigation of intragastric drug release.<br />

Further experiments are needed to provide detailed information on mixing behavior and<br />

hydrodynamics. In addition, the dynamics of gastric emptying will be considered in certain<br />

test programs.<br />

Acknowledgments: Federal Ministry of Education and Research (GASTROMAX, FKZ 13 N11368-370)<br />

References:<br />

1. Weitschies, W. et al.: J. Contr. Rel. 2005 108(2-3): 375-385.<br />

067<br />

Molecular Dynamics Simulations and MM/GBSA-Calculations of Docking<br />

Poses to predict the native Binding Mode of thieno[2,3-b]pyridines in PfGSK-3<br />

Poll, B. 1; Kruggel, S. 1; Kunick, C. 2; Brandt, W. 2; Meijer, L. 3, Lemcke, T. 1<br />

1 <strong>Universität</strong> Hamburg, Institut für Pharmazie, Bundesstraße 45, 20146 Hamburg, Germany<br />

2 Technische <strong>Universität</strong> Braunschweig, Institut für Medizinische und Pharmazeutische Chemie,<br />

Beethovenstraße 55, 38106 Braunschweig, Germany<br />

3 Protein Phosphorylation & Human Disease Group, CNRS, Station Biologique, Place Georges<br />

Teissier, B.P. 74, 29682 Roscoff, France<br />

In addition to HIV and Tuberculosis, Malaria is one of the most prevalent infectious<br />

diseases in developing countries, causing over 200 mio. infections every year [1]. Malaria<br />

tropica, the most serious form of malaria, is caused by Plasmodium falciparum. There are<br />

potent malaria therapeutics on the market, however rapid emergence of resistance and<br />

high costs necessitate the development of new drugs [2].<br />

In cooperation with the University of Braunschweig and the CNRS in Roscoff (France) a<br />

thieno[2,3-b]pyridine derivative has been identified as a potential lead structure to inhibit the<br />

plasmodial Glycogen Synthase Kinase 3 (PfGSK-3) [3,4].<br />

One of the crucial aspects of lead structure optimization is the identification of the correct<br />

binding geometry of putative inhibitors. Homology models of PfGSK-3 have been<br />

developed [5] and were used for docking- and 3D-QSAR-studies [6] as well as guide for<br />

structural variations [4]. Different binding modes have been identified and investigated [6].<br />

We used molecular dynamics simulations and MM/GBSA-calculations of the respective<br />

protein-ligand-complexes considering pre-optimized calculation parameters [7] to further<br />

evaluate the binding poses of a series of thieno[2,3-b]pyridine derivatives 1.<br />

Poster 109<br />

R1<br />

1<br />

The GBSA energies were correlated to the biologically measured IC50 values of the<br />

individual compounds for complexes with the proposed binding mode and give further<br />

evidence of the native binding mode [6]. The results of these investigations will be<br />

presented.<br />

References:<br />

1. WHO, World Malaria Report 2010.<br />

2. Noedl, H. et al.: N. Engl. J. Med. 2008, 359: 2619-2620.<br />

3. Droucheau, E. et al.: Biochim. Biophys. Acta. 2004, 1697: 181-196.<br />

4. Brandt, W.: Dissertation, Braunschweig 2009.<br />

5. Kruggel, S., Lemcke, T.: Arch. Pharm. Chem. Life. Sci. 2009, 342: 327-332.<br />

6. Kruggel, S.: Dissertation, Hamburg 2011.<br />

7. Poll, B. et al.: MM-PBSA and MM-GBSA Calculation for Estimating the Free Energy of Binding of<br />

Glycogen Synthase Kinase-3 Inhibitors 2011. Joint Meeting of the German and Austrian pharmaceutical<br />

societies. Innsbruck. Austria.<br />

068<br />

Pharmacometrics – methodologies, establishment and opportunities of a<br />

promising new discipline in Germany<br />

Minichmayr I1 2; Kloft C1 1 Dept. of Clinical Pharmacy and Biochemistry, Institute of Pharmacy, Freie Universitaet Berlin,<br />

Kelchstr. 31, 12169 Berlin, Germany<br />

2 and Graduate Research Training program PharMetrX, Germany<br />

Background: The emerging science of pharmacometrics represents a multidisciplinary<br />

research field bridging clinical pharmacy, pharmacology, medicine, statistics, engineering<br />

and computational methods [mod. 1]. During the past years, pharmacometrics has been<br />

gaining pivotal importance in drug development, regulatory approval, study design and<br />

therapeutic decision making [2]. The present work presents an overview over different<br />

concepts and methodologies within pharmacometrics, outlines possible applications and<br />

gives examples of past pharmacometric accomplishments. Additionally, the current<br />

establishment of pharmacometrics and educational resources in Germany are presented.<br />

Methods: Seminal papers on pharmacometrics were reviewed. Prevalence of<br />

pharmacometrics research at German Institutes of Pharmacy was assessed by systematic<br />

literature search (PubMed, university homepages).<br />

Results: Pharmacometrics applies computational models of biology, anatomy, physiology,<br />

pharmacology, disease and systems biology to describe and quantify interactions between<br />

drugs and patients [mod. 1]. Ultimately, the qualitative, quantitative and temporal<br />

relationships between drug exposure (pharmacokinetics, PK), drug effects<br />

(pharmacodynamics, PD) and favourable or unfavourable responses in connection with<br />

disease and recovery progression are explored and predicted on a subcellular, cellular,<br />

tissue, organ and organism level.<br />

Various approaches exist to characterise drug-patient interactions, ranging from classical<br />

empirical to more mechanistic pharmacometric models. The most prominent concepts –<br />

often separately used in research – are:<br />

1) Classical drug data analysis comprises single individual analysis and population basedmethodologies,<br />

which allow for e.g. the assessment of both inter- and intra-individual and<br />

unexplained variability. Within the latter, the standard two stage approach requires data rich<br />

situations, whereas non-linear mixed effect (NLME) modelling is also apt for modelling<br />

sparse data, typically obtained in patient care, due to simultaneous analysis of several<br />

individuals.<br />

2) Physiologically-based pharmacokinetic/pharmacodynamic (PBPK/PD) models do not<br />

start with study data but anatomical and physicochemical drug data, enabling a more<br />

mechanistic simulation of drug profiles in individual tissues and blood. By means of lumping<br />

techniques, a direct link to classical compartment models can be established [3].<br />

3) Systems biology models consider even more detailed knowledge of underlying complex<br />

biologic processes, e.g. cell signal transduction pathways and their subsequent gene<br />

regulatory network.<br />

Despite the widespread potential of pharmacometrics, educational resources (e.g. scientific<br />

societies, textbooks, journal articles) in this field are scarce. At German Institutes of<br />

R2


Pharmacy, pharmacometric modelling skills and competences are basically gained by<br />

postgraduate training within the scope of a PhD (Universities of Berlin, Bonn, Duesseldorf,<br />

Muenster, planned: Saarbruecken) or a structured graduate research training program<br />

(PharMetrx - Pharmacometrics and Computational Disease Modelling at the Freie<br />

Universitaet Berlin and the University of Potsdam, jointly with five industry partners).<br />

Conclusion: Ideally, all three concepts of pharmacometrics should be linked and<br />

combinedly integrated in future research. Given the tremendous progress and value of the<br />

various applications of pharmacometrics in drug development (“model-based drug<br />

development”), regulatory and therapeutic decisions (“model-based patient care”), the<br />

demand for pharmacometricians is substantially growing. To meet the need in Germany,<br />

several universities already provide postgraduate training. Nonetheless, further expansion<br />

of pharmacometric expertise, particularly in undergraduate pharmacy curricula, is desirable.<br />

References:<br />

1. Barrett JS. et al.: J. Clin. Pharmacol. 2008, 48(5): 632-49.<br />

2. Gobburu JV.: J. Clin. Pharmacol. 2010, 50(9 Suppl): 151-157.<br />

3. Pilari S., Huisinga, W.: J. Pharmacokinet. Pharmacodyn. 2010, 37(4): 365-405.<br />

069<br />

Comparison of the ATP-Binding Sites of IR and IGF-1R Kinase Crystal<br />

Structures and their corresponding TrK Homology Model Counterparts<br />

Löhr, J. H. 1; Lemcke, T. 1<br />

1 <strong>Universität</strong> Hamburg, Institut für Pharmazie, Bundesstraße 45, 20146 Hamburg, Germany<br />

Receptor tyrosine kinases (RTKs) have been pursued as promising targets in cancer<br />

therapy. Transforming extracellular signals into cellular responses, they take part in the<br />

regulation of cell growth and survival. Deregulation of certain RTKs has been linked to<br />

various human tumor conditions and already various small molecule inhibitors have been<br />

approved for therapeutic use.[1]<br />

The insulin-like growth-factor receptor 1 (IGF-1R) and the neurotrophic tyrosine kinase<br />

receptor (TrK) are examples of cancer associated RTKs, where inhibition may prove to be<br />

an effective therapeutic intervention.[2, 3]<br />

With plenty of crystal structures available, structure based design of small molecule<br />

inhibitors of the ATP-Binding site of the IGF-1R kinase is a feasible strategy. However, offtarget<br />

activity on the highly homologous insulin receptor kinase (IR) poses a challenge. In<br />

absence of published three-dimensional models of the TrK, construction and evaluation of<br />

homology models has to precede any structure-based efforts.<br />

Previously, homology models of both active and inactive states of the TrK-1 and TrK-2<br />

kinase domains were built with suitable IR and IGF-1R X-ray structures serving as<br />

templates. Using molecular interaction fields (MIFs) and the GRID/GOLPE approach, the<br />

MIFs were investigated for statistical differences among the different crystal structures of<br />

IR/IGF-1R and the corresponding TrK-1/2 homology models.[4-6] Based on these findings,<br />

geometric and volumetric measurements were evaluated in order to pinpoint structural<br />

differences that emerge from the set of over 30 crystal structures and 10 homology models.<br />

A recently published partly solved crystal structure showing the TrK-3 kinase domain for the<br />

very first time is also taken into account both to assess the homology models and to<br />

highlight structural differences.[7]<br />

References:<br />

1. Arora, A., Scholar, E.M.: J. Pharmacol. Exp. Ther. 2005, 315(3): 971–979.<br />

2. Li, R., Pourpak, A., Morris, S.W.: J. Med. Chem. 2009, 52(16): 4981–5004.<br />

3. Nakagawara, A.: Cancer Lett. 2001, 169(2): 107–114.<br />

4. Goodford, P.J.: J. Med. Chem. 1985, 28(7): 849–857.<br />

5. Kastenholz, M.A. et al.: J. Med. Chem. 2000, 43(16): 3033–3044.<br />

6. Baroni, M. et al.: Quant. Struct.-Act. Relat. 1993, 12(1): 9–20.<br />

7. Albaugh, P. et al.: ACS Med. Chem. Lett. <strong>2012</strong>, 3(2): 140–145.<br />

070<br />

Two-dimensional capillary gel electrophoresis (2-D-CGE) as powerful<br />

instrument for quantitation and characterisation of biomolecules<br />

Hahne, T.; Cianciulli, C.; Wätzig, H.<br />

Institute of Medicinal and Pharmaceutical Chemistry, Technische <strong>Universität</strong> Braunschweig,<br />

Beethovenstr. 55, 38106 Braunschweig, Germany<br />

Capillary gel electrophoresis (CGE) is a highly selective separation technique, used more<br />

and more in pharmaceutical industry for analysis of biomolecules. Thus, CGE is replacing<br />

conventional gel electrophoresis stepwise, because of some advantages such as ease of<br />

handling and time-saving. Even if CGE is a powerful and selective tool for bioanalysis the<br />

selectivity was increased by adding a second dimension. According to our approach the<br />

molecules are separated due to their isoelectric point in the first dimension and then due to<br />

their molecular weights in the following second dimension, as known from classical CGE. In<br />

this study a protein mixture consisting of bovine serum albumin (BSA), ovalbumin, ßlactoglobulin,<br />

myoglobin and a monoclonal antibody was used to investigate the<br />

performance of this innovative separation technique. Reproducibilities, resolutions, signalto-noise<br />

ratios and limits of quantitation with different concentrations of the biomolecules<br />

were investigated.<br />

071<br />

A comparison: Quantification of therapeutically used organic nitrates via LC-<br />

UV and LC-ESI-MS in biological samples after solvent and solid-phase<br />

extraction respectively<br />

Puerstinger, J. 1; Seeling, A. 1<br />

1 Institute of Pharmaceutical/Medical Chemistry, Friedrich-Schiller University, Philosophenweg 12,<br />

07747 Jena, Germany<br />

Pentaerithrityl tetranitrate (PETN), isosorbide dinitrate (ISDN) and glyceryl trinitrate (GTN)<br />

are organonitrate coronary vasodilators and, consequently, used in the treatment of<br />

coronary insufficiency and angina pectoris. Sustained therapeutical effects of PETN and<br />

ISDN are useful in the prophylaxis of attacks of angina pectoris but lead to a slower onset<br />

in comparison to GTN which plays an important role in the management of cardiac<br />

infarction. Furthermore, recent investigation points to a remarkable benefit of organic<br />

nitrates in the therapeutic fields of diabetes mellitus [1] and pulmonary hypertension [2].<br />

Bioactivation via several enzymatic reduction pathways [3] results in less nitrated polyols<br />

which contribute to the vasodilator potency [4].<br />

In order to quantify the therapeutically used organic nitrates and to monitor their<br />

denitrification processes, we developed miscellaneous chromatographic separation<br />

methods to determine PETN, ISDN and GTN as well as their less nitrated metabolites in<br />

biological liquids such as human plasma and whole blood samples using solvent or solidphase<br />

extraction for the segregation of the analytes and, subsequently, UV or ESI-MS for<br />

their detection.<br />

Acknowledgments: Measurements on LC-ESI-MS instruments could be realized in the research group of<br />

Prof Christian G. Huber, Department of Chemistry and Bioanalytics, University of Salzburg, Austria.<br />

References:<br />

1. Wenzel P. et al.: Arterioscler. Thromb. Vasv. Biol. 2007, 27(8): 1729-1735.<br />

2. Daiber A. et al.: Mol. Pharmacol. 2004, 66: 1372-1382.<br />

3. Munzel T., Daiber A., Mulsch A.: Circ. Res. 2005, 97(7): 618-628.<br />

4. Wenzel P. et al.: Br. J. Pharmacol. 2007, 150(4): 526-533.<br />

072<br />

Pulmonary deposition of three market dry powder inhalers using an idealized<br />

pediatric and adult mouth-throat model<br />

Below, A. 1; Bickmann, D. 2; Breitkreutz, J. 1<br />

1 Institute of Pharmaceutics and Biopharmaceutics, Heinrich-Heine-University, 40225 Düsseldorf,<br />

Germany<br />

2 Boehringer Ingelheim Pharma GmbH & Co. KG, 55216 Ingelheim am Rhein, Germany<br />

The efficiency of aerosol delivery to the patient´s lung depends on three main parameters:<br />

inhalation device, formulation and patient. Marketed inhalation products are in most cases<br />

developed according to the needs of adult patients and less for children. Changes in upper<br />

airway geometry and breathing pattern due to maturation from childhood to adolescent are<br />

usually not considered. Because of respiratory diseases like childhood asthma and cystic<br />

fibrosis even young children have to use inhaled medicines. The efficiency of inhaled<br />

products is especially in preschool children unknown. The complex interaction between<br />

inhaled product and patient is the scope of this study. In a first step the influence of the<br />

upper airway geometry in dependence of age on the performance of three market products<br />

[1-2] was investigated. Afterwards, also the impact of different flow rates was considered.<br />

For the in vitro assessment of particle deposition via next generation impactor (NGI), three<br />

commercially available multidose dry powder inhalers (Salbu Novolizer ® [1], SalbuHexal ®<br />

Easyhaler ® [2], Asmanex ® Twisthaler ®) were connected to two different inlets: idealized<br />

pediatric and adult upper airway model. The pediatric model represents 4-5-year-old<br />

children. In dependence of the device different steady flow rates through the model were<br />

achieved. The inhalation volume was adjusted to 4L (adult model) and 1L (adult and<br />

pediatric model). Deposited mass in the model (extrathoracic deposition) and at the model<br />

outlet (pulmonary deposition) was collected, determined by HPLC and related to label<br />

claim.<br />

An influence of inhalation volume on pulmonary deposition (PD) was observed for the<br />

Novolizer (15% vs. 22% at 75 L/min (4 kPa)). Easyhaler and Twisthaler PD were not<br />

influenced by inhalation volume accordingly. The comparison of adult and pediatric models<br />

has shown higher pulmonary doses using the adult mouth-throat model for all three<br />

products, e.g. Twisthaler 21% vs. 27% (46 L/min (4 kPa)). Different flow rates have an<br />

influence on product performance especially on Twisthaler performance. Using the pediatric<br />

mouth-throat model PD increased from 15% (30 L/min (2 kPa) to 21% (46 L/min (4 kPa)).<br />

Novolizer and Easyhaler performance is very robust; PD is almost the same for all<br />

investigated flow rates.<br />

Physical upper airway models as impactor inlets simulate the human upper airways more<br />

correctly than the USP/Ph.Eur. inlet. Using the adult mouth-throat model higher pulmonary<br />

doses were observed than for the pediatric model. Because there is still a lack of in vivo<br />

data of preschool children using dry powder inhalers in vitro in vivo correlation is not<br />

feasible. If in vivo deposition data will be available in future, the developed model may be<br />

validated. Based on different product performance we believe that for pediatric product<br />

developments child-appropriate models should be used.<br />

References:<br />

1. Below, A. et al.: 18th ISAM conference 2011<br />

2. Below, A.: 8th PBP World Meeting <strong>2012</strong>.<br />

073<br />

Biosynthesis of structurally diverse paracylophanes by Vietnamese<br />

cyanobacteria<br />

Preisitsch, M. 1, Neidhardt, I. 1, Pham, T. L. H. 1, Bodtke, A. 2, Niedermeyer, T. H. J. 3, Mundt,<br />

S. 1<br />

1 Institute of Pharmacy, Department of Pharmaceutical Biology, <strong>Ernst</strong> <strong>Moritz</strong> <strong>Arndt</strong> University<br />

<strong>Greifswald</strong>, Friedrich-Ludwig-Jahn-Str. 17, 17489 <strong>Greifswald</strong>, Germany<br />

2 Institute of Pharmacy, Department of Pharmaceutical/Medicinal Chemistry, <strong>Ernst</strong> <strong>Moritz</strong> <strong>Arndt</strong><br />

University <strong>Greifswald</strong>, Friedrich-Ludwig-Jahn-Str. 17, 17489 <strong>Greifswald</strong>, Germany<br />

3 Cyano Biotech GmbH, Magnusstraße 11, 12489 Berlin, Germany<br />

Cyanobacteria have been identified as a promising source of new, highly diverse, bioactive<br />

natural products in the research of novel pharmaceutical leads. Especially cyclic peptides<br />

and lipopeptides, alkaloids, lactones, amides, esters, derivatives of fatty and amino acids,<br />

biosynthesized by marine cyanobacteria, represent the largest part of recently isolated<br />

compounds with mainly antibiotic, antifungal, cytotoxic or enzyme-inhibiting activities [1]. In<br />

contrast to these metabolites, naturally occurring paracyclophanes have been isolated less<br />

frequently from cyanobacteria [2].<br />

Bioassay-guided separation of the methanol extract from the Vietnamese freshwater<br />

cyanobacterium Nostoc sp. CAVN 10 resulted in five [7.7]paracyclophanes,<br />

carbamidocyclophanes A-E, including carbamido moieties and a varying substitution<br />

pattern of the chlorinated butyl side chains. Carbamidocyclophanes A-C exhibited cytotoxic<br />

activity against MCF-7 (breast cancer cell line) and Fl cells (human amniotic epithelial cell<br />

line) and moderate antibacterial activity against Staphylococcus aureus (ATCC 6538)[3].<br />

Further screenings for carbamidocyclophanes in various cyanobacteria, in particular in<br />

Nostoc strains, revealed a second carbamidocyclophane producing strain, Nostoc sp.<br />

CAVN 2. HPLC-DAD/IT-TOF-HRMS analysis of separated CAVN 2 cyclophanes resulted in<br />

an identification of already published [2-4] and also previously unknown cyclophanes.<br />

Feeding experiments with Nostoc sp. CAVN 2, using different halogen salts, yielded in the<br />

first-time detection of brominated carbamidocyclophanes.<br />

An optimized, highly rapid one-step extraction and separation procedure for the detection of<br />

110 Poster


carbamidocyclophanes as well as closely related structural analogues and first insights into<br />

the phylogenetic relationship of axenic carbamidocyclophane biosynthesizing<br />

cyanobacteria are presented.<br />

Acknowledgments: The authors thank Dr. M. Meixner for 16S rDNA amplification and sequencing of<br />

investigated strains as well as Mr. S. Heiden for the assistance in the construction of the phylogenetic tree.<br />

References:<br />

1. Singh, R. K. et al.: J. Antibiot. 2011, 64(6):401-412.<br />

2. Chlipala, G. E. et al.: J. Nat. Prod. 2010, 73(9): 1529-1537.<br />

3. Bui, H. T. N. et al.: J. Nat. Prod. 2007, 70(4): 499-503.<br />

4. Kang, H.-S. et al.: Phytochem. <strong>2012</strong>, 79(0): 109-115.<br />

074<br />

Cannabinoids stimulate mesenchymal stem cell migration via a mitogenactivated<br />

protein kinase pathway<br />

Schmuhl, E. 1; Ramer, R. 1; Peters, K. 2; Hinz, B. 1<br />

1 Institute of Toxicology and Pharmacology, University of Rostock, Schillingallee 70, D-18057<br />

Rostock, Germany<br />

2 Department of Cell Biology, University of Rostock, Schillingallee 69, D-18057 Rostock, Germany<br />

Mesenchymal stem cells (MSC) are known to be involved in various regenerative<br />

processes such as cardiac, ocular, skin and bone tissue healing. However, little is known<br />

about the pharmacotherapeutical options aiming at tissue healing steps such as the<br />

mobilization and homing of MSC. Here, we show that cannabidiol (CBD), a nonpsychoactive<br />

cannabinoid, stimulates the migration of human adipose-derived MSC in both<br />

Boyden chamber and in vitro scratch wound assays. In Boyden chambers CBD (0.01 – 3<br />

µM) was shown to promote cell migration in a time- and concentration dependent manner.<br />

This promigratory action was inhibited by AM-630 (CB2 receptor antagonist) and by O-1602<br />

(G protein-coupled receptor [GPR] 55 agonist). Moreover, CBD activated the mitogenactivated<br />

protein kinase (MAPK) pathway as evidenced by increased phosphorylation of<br />

extracellular signal-regulated kinase (ERK) 1/2. Blockade of ERK activation by PD98059<br />

prevented CBD-stimulated MSC migration, whereas inhibition of p38 MAPK by SB203580<br />

was inactive in this respect. Furthermore, AM-630 and O-1602 were found to attenuate<br />

CBD-induced ERK activation. An ERK-dependent promigratory action was likewise<br />

demonstrated for the phytocannabinoid Δ9-tetrahydrocannabinol, the GPR55 antagonist O-<br />

1918 and the selective CB2 receptor agonist JWH-133. We conclude that cannabinoids<br />

promote MSC migration via receptor-dependent ERK activation, possibly contributing to<br />

tissue healing.<br />

075<br />

Accuracy temperature control as a meaningful tool for microwave assisted<br />

method development and optimization<br />

Härter A<br />

Anton Paar GmbH, Helmuth-Hirth-Str. 6, 73760 Ostfildern<br />

During the last decade microwave-assisted synthesis has become a powerful tool for<br />

pharmaceutical method development and optimization. Controlled microwave heating under<br />

sealed vessel conditions has - shown to dramatically reduce reaction times, increase<br />

product yields, and enhance product purities by reducing unwanted side reactions<br />

compared to conventional synthetic methods. Modern microwave reactors control and<br />

regulate important parameters such as temperature, pressure and power. Due to this<br />

plurality of control parameters, the essential of accurate temperature for reaction<br />

optimization is often unacknowledged.<br />

We will introduce the basics of optimal temperature measurement, including tips for heating<br />

- microwave transparent solvents, effects due to external cooling, the importance of reliable<br />

stirring and the influence of reactor stability. The concept of accurate temperature control is<br />

outlined, which enables performing optimized protocols in any applicable scale in any<br />

instrument of our modular equipment family without reoptimization. This increases the<br />

efficiency of microwave-mediated synthesis even further, speeding up the entire process of<br />

academic and industrial research.<br />

076<br />

Using sedimentation velocity centrifugation to study the aggregation process<br />

of proteins<br />

Wolff, M. 1 2; Nagel-Steger L. 1 2; Willbold, D. 1 2<br />

1 Heinrich-Heine-<strong>Universität</strong>, Institut für Physikalische Biologie, 40225 Düsseldorf, Germany<br />

2 Forschungszentrum Jülich, ICS-6, 52425 Jülich, Germany<br />

The pathogenic aggregation of proteins plays an important role in the development of a<br />

number of neurodegenerative diseases. For example in Alzheimer’s or Parkinson’s Disease<br />

the self-assembly of misfolded cellular proteins causes the formation of different oligomeric<br />

structures which finally leads to amyloid fibril formation. In order to identify different<br />

assemblies in the aggregation process we employ the method of sedimentation velocity<br />

(SV) analysis. Using SV analysis gives us the opportunity to study protein size and shape<br />

distributions in solution under defined conditions. The advantages of SV analysis are that 1)<br />

no interaction with liquid or solid phases influences the measurement as known for other<br />

separation techniques like e.g. size exclusion chromatography, and 2) that due to the<br />

fractionation power of the method large aggregates do not impair the results as in light<br />

scattering methods.<br />

One of the most important targets in Alzheimer’s therapy is the 39 to 43 amino acids long<br />

proteolytic fragment of the amyloid precursor protein, called amyloid β peptide for its<br />

property to form amyloid fibrils. Beginning with the characterization of the hydrodynamic<br />

properties of the monomeric Aβ peptide preparations, we will analyze early steps in<br />

aggregate formation in order to identify small Aβ assemblies of defined size, which are<br />

significantly populated in freshly prepared Aβ solutions. With this approach we want to gain<br />

insight into mechanistic details of the aggregation process. Moreover this approach is<br />

aimed at the identification of novel drug targets for Alzheimer’s disease therapy.<br />

077<br />

New Furanoid and seco-Labdanoid Diterpenes isolated from Leonurus<br />

cardiaca L.<br />

Brückner, A. 1; Hennig, L. 2; Rauwald, H. J. 1<br />

1University of Leipzig, Department of Pharmaceutical Biology, Johannisallee 21-23, D-04103<br />

Leipzig, Germany<br />

2University of Leipzig, Department of Chemistry, Institute for Organic Chemistry, Johannisallee 39,<br />

D-04103 Leipzig, Germany<br />

Continuing our search for possible cardioactive single constituents of primary and refined<br />

antiarrhythmic extracts of Leonurus cardiaca L. [1] we report herewith the isolation of three<br />

new diterpenes besides known substances, like leosibiricin, sitosterol and stigmasterol [2].<br />

The structures of isoleosibirin, leopersin F and 7-epi-leopersin F were determined by 1d/2d<br />

1H/ 13C NMR spectroscopical experiments. These labdane type diterpenes have been found<br />

only once in other plants of the genus leonurus, namely in the related Lamiaceae Leonurus<br />

sibiricus and Leonurus persicus [3, 4]. Thus these compounds may prove useful as<br />

analytical marker substances as well as possible pharmacologically active compounds, as<br />

for example shown for the vasorelaxant activity of similar diterpenes isolated of Marrubium<br />

vulgare [5].<br />

References:<br />

[1] Ritter, M et al.: Planta Med. 2010, 76: 572-582.<br />

[2] Knöss, W.: Plant Cell Reports 1995, 14: 790-793.<br />

[3] Tasdemir, D.; Wright, A.D.; Sticher, O.: J. Nat. Prod. 1996, 59: 131-134.<br />

[4] Savona, G.; Piozzi, F.; Rodriguez, B.: Phytochemistry 1982, 21 (11): 2699-2701.<br />

[5] El Bardai, S.; Morel, N.; Wibo, M.: Planta Med. 2003, 69: 75-77.<br />

078<br />

Characterization of the pharmacokinetic properties of visnagin and Ammi<br />

visnaga water extract following oral administration in rats<br />

Haug, K. G. 1; Weber, B. 1; Hochhaus, G. 1; Butterweck, V. 2<br />

1 Department of Pharmaceutics, College of Pharmacy, University of Florida, 1600 SW Archer<br />

Road, Gainesville, FL,32610, USA<br />

2 Institute for Pharma Technology, School of Life Sciences, University of Applied Sciences<br />

Northwestern Switzerland, Gruendenstrasse 40, 4132 Muttenz, Switzerland<br />

The furanochromone visnagin is one of the main compounds of Ammi visnaga L. (syn.<br />

Khella, Apiaceae) with potential effects on kidney stone prevention. After determination of<br />

the pharmacokinetic (PK) properties of visnagin after intravenous bolus administration in<br />

rats [1], the aim of the present study was the evaluation of the PK properties of visnagin<br />

and an aqueous Ammi visnaga extract (AVE) after oral administration in rats. In two<br />

separate experiments three doses of visnagin (2.5, 5, and 10 mg/kg) solubilized in 25%<br />

Captisol® and three doses of AVE (standardized on visnagin and containing equivalent<br />

amounts of visnagin) were administered by oral gavage to male Sprague-Dawley rats,<br />

respectively. Plasma samples were extracted and subsequently analyzed using a validated<br />

LC-MS/MS method [1]. Plasma concentration-time profiles (Figure 1) were explored by<br />

non-compartmental analysis. Visnagin plasma exposure (median AUClast and AUCinf) was<br />

significantly increased for all three doses (up to 10-fold for the low dose) when administered<br />

as extract compared to the pure agent (Figure 2). For both the AVE and the pure<br />

compound, AUClast and AUCinf increased disproportionately with an increase in dose (Figure<br />

1 & 2). Visnagin resided significantly longer in the body when given in form of AVE with up<br />

to three times longer median MRTlast and MRTinf for the low dose. Cmax values after AVE<br />

administration were elevated and occurred at later time points in comparison to equivalent<br />

doses of pure visnagin. The terminal half-life increased with dose for both AVE and pure<br />

visnagin, reaching a maximum value of 1.94 h for the 10 mg/kg pure compound group.<br />

In conclusion, exposure of visnagin is enhanced after extract administration and could<br />

result in a superior efficacy of AVE compared to an equivalent dose of visnagin.<br />

Figure 1: Visnagin plasma concentration (geometric mean � SEM) vs. time profiles after<br />

administration of the pure compound (a) and AVE (b), sorted by dosing group (Low: 2.5<br />

mg/kg, Medium: 5 mg/kg, High: 10 mg/kg).<br />

Figure 2: AUCinf (a) and AUClast (b) (median + SD) of visnagin after administration of pure<br />

compound vs. AVE, sorted by dosing group (Low: 2.5 mg/kg, Medium: 5 mg/kg, High: 10<br />

mg/kg), numbers represent Bonferroni adjusted p-values, AUCinf values that are based<br />

upon extrapolated AUC > 50% were excluded.<br />

Reference:<br />

Haug, K.G. et al.: Eur J Pharm Sci <strong>2012</strong>, 45 (1-2): 79-89<br />

079<br />

Synthesis and biological studies on novel water-soluble gold(I)alkynyl<br />

complexes as antitumor agents and thioredoxin reductase inhibitors.<br />

Andermark, V. 1; Meyer, A. 1; Ott, I. 1<br />

1 Institute of Medicinal and Pharmaceutical Chemistry, Technische <strong>Universität</strong> Braunschweig,<br />

Beethovenstraße 55, 38106 Braunschweig, Germany<br />

Poster 111


Besides the use of gold(I) complexes, i.e. auranofin, as disease modifying antirheumatic<br />

drugs (DMARDs)they are also known for their potential as antitumor drugs and highly<br />

efficient inhibition of thioredoxin reductase (TrxR)[1], an enzymewhich is overexpressed in<br />

tumor cells.TrxR contains a selenocystein in the active site, making it an excellent target for<br />

soft lewis acids like gold(I)-complexes[2]. Inhibition of TrxR leads to increased reactive<br />

oxygen species (ROS) levels, cell cycle arrest and can induce apoptosis [3].<br />

This work bases on recent results, which show that phosphinegold(I)alkynyl complexes can<br />

inhibit TrxR in the nanomolar concentration range and have anti-angiogenic properties in<br />

zebrafish embryos[4].To improve the water-solubility of the phosphinegold(I)alkynyl<br />

complexes the bulky, hydrophobic triphenylphosphine-ligand of the most active complexes<br />

will be replaced by more polar phosphine ligands, while the most promising alkynyl ligands<br />

will be kept.<br />

The tri-(2-furyl)phosphinegold(I)chloride (3)complex is prepared by the reaction of<br />

equimolar amounts of dimethylsulfidegold(I)chloride (1) and tri-(2-furyl)phosphine (2).<br />

Afterwards 3 is reacted with an equimolar amount ofan alkyne (4) in basic conditions,<br />

yielding aphosphinegold(I)alkynyl(5) complex (scheme 1). Synthesized compounds are<br />

confirmed by 1H, 13C, 31P NMRand their purity is evaluated by elemental analysis.<br />

The cytotoxicity of the synthesised complexes, as well asthe metal free alkynes and<br />

phosphinesis evaluated using HT-29 colon carcinoma cells and MCF-7 breast cancer cells<br />

in the colorimetric crystal violet assay. Additionallycurrent results onTrxR inhibition,<br />

glutathione reductase inhibition and cellular uptake into tumor-cells will be presented.<br />

Scheme 1.Synthesis of the compounds 3 and 5 as an example for the general synthetic<br />

route.<br />

Acknowledgments: Financial support bythe Deutsche Forschungsgemeinschaft (DFG, project FOR630)is<br />

gratefully acknowledged.<br />

References:<br />

1. Ott, I.: Coord. Chem Rev.2009, 253: 1670-1681.<br />

2. Gromer, S. et al.: J. Biol. Chem. 1998,273(32): 20096-20101<br />

3. Urig, S., Becker, K.: Semin.Cancer Biol. 2006,16: 452-465<br />

4. Meyer A. et al.:Angew. Chemie<strong>2012</strong>accepted<br />

080<br />

Influence of opioid receptor activation and inhibition on synaptic transmission<br />

under hypoxia<br />

Bloßfeld, M. 1; Merkenschlager, A. 2; Nieber, K. 1<br />

1 University Leipzig, Institute of Pharmacy, Pharmacology for Natural Sciences, Talstraße 33,<br />

04103 Leipzig, Germany<br />

2 <strong>Universität</strong>sklinikum Leipzig, Klinik und Poliklinik für Kinder und Jugendliche, Liebigstraße 20a,<br />

04103 Leipzig<br />

Activation of delta opioid receptors by [D-Ala2, D-Leu5] enkephalin (DADLE) has been<br />

reported to have neuroprotective effects on rat embryonic cortical neurons and PC12 cells<br />

under hypoxia. A neurotoxic impact has also been shown for micromolar concentrations<br />

[1,2]. There are limited information about effects of naloxon, a mu receptor antagonist,<br />

during hypoxia. The aim of the present study was to examine the influence of DADLE and<br />

naloxone on the amplitude of electrically evoked postsynaptic potentials (PSPs) under<br />

normoxic (95% O2, 5% CO2) and hypoxic (1% O2, 5% CO2, 94% N2) conditions. The<br />

experiments were done using intracellular recordings on cortical pyramidal cells in brain<br />

slices of adult rats. DADLE (1 µM) decreased the amplitude of PSPs. Naloxone (100 µM)<br />

inhibited synaptic transmission in neurons from 10 week old rats whereas in neurons of<br />

brain slices of rats older than 10 weeks no influence could be found. Naloxone (100 µM)<br />

inhibited completely the effect of DADLE (1µM). Both components had no influence on<br />

input resistance and membrane potential. Exposure of the slices to hypoxia (5 min) resulted<br />

in a decreased amplitude of PSPs. DADLE (1 µM), superfused 5 min before hypoxia, did<br />

not alter the hypoxia-decreased PSPs. Contrary, naloxone (100 µM) antagonised the PSP<br />

amplitude reduction. This effect was age-independent. We conclude that during hypoxia<br />

naloxon (100µM) but not DADLE (1µM) normalizes synaptic transmission as an indicativ of<br />

a neuroprotective effect.<br />

References:<br />

1. Su, D.S. et al.: Neurosci. Lett. 2007, 423, 113-117<br />

2. Sun K., Su D.S. and Wang X.: Brain Res. 2009, 1292, 100-106<br />

081<br />

Glutathione transferase isoenzymes in corneal tissues, human cornea<br />

construct and corneal cell lines<br />

Kölln C1; Reichl S1 1 Institut für Pharmazeutische Technologie, Technische <strong>Universität</strong> Carolo-Wilhelmina zu<br />

Braunschweig, Mendelssohnstr. 1, 38106 Braunschweig, Germany<br />

The human cornea represents the main barrier for active drug substances from ophthalmic<br />

formulations. Therefore, excised cornea and corneal cell culture models are used for in vitro<br />

drug absorption studies. However, little is known about drug metabolism during<br />

transcorneal passage. The purpose of this study was to determine expression and activity<br />

of glutathione transferases (GST) in human corneal cell lines and human cornea construct<br />

in comparison to excised animal corneas. Isoenzymes GSTO1 and GSTP1 are supposed<br />

to be considered in more detail.<br />

Presence of GST isoenzymes in corneal cell lines, cornea construct and excised corneal<br />

tissues was determined by western blot and immunohistochemical staining using specific<br />

antibodies against isoenzymes GSTO1 and GSTP1. In this study corneal cell lines HCE-T<br />

(epithelial), HCK-Ca (stromal) and HENC (endothelial) were investigated as well as tissue<br />

engineered human cornea construct [1, 2]. Porcine and rabbit corneas were studied in<br />

comparison. The activity was measured by spectrophotometrically methods. Total GST<br />

activity was profiled using 1-Chloro-2,4-dinitrobenzene (CDNB) [3], specific GSTO1 activity<br />

by using S-(4-Nitrophenacyl)-glutathione [4] and specific GSTP1 activity was investigated<br />

using ethacrynic acid [5].<br />

The presence of glutathione transferases in human cornea construct and all corneal cell<br />

lines was verified on protein level by western blot and immunohistochemical staining,<br />

respectively. In case of corneal cell lines, activity of investigated isoenzymes seems to be<br />

comparable in HCE-T and HCK-Ca, whereas total GST activity was found to be highest in<br />

HCE-T. In comparison, HENC showed lowest activity in all studies. Excised rabbit and<br />

porcine corneas showed higher specific GSTP1 and GSTO1 activity as well as total GST<br />

activity compared to human cornea construct.<br />

Known to be present in human cornea, glutathione transferase activity is detectable in all<br />

investigated human corneal cell lines, as well as in human cornea construct and excised<br />

animal corneas.<br />

Acknowledgments: We are grateful to the German Federal Institute for Risk Assessment (BfR), which<br />

funded this work under grant no. 3-1328-30652054369.<br />

References:<br />

1. Hahne M, Reichl S: Int J Pharm. 2011, 416(1): 268-279.<br />

2. Hahne M et al.: J Pharm Sci. <strong>2012</strong>, accepted<br />

3. Habig WH, Pabst MJ, Jakoby WB: J Biol Chem. 1974, 249(22): 7130-7139.<br />

4. Board PG et al.: Anal Biochem. 2008, 374(1): 25-30.<br />

5. Habig WH, Jakoby WB: Methods Enzymol. 1981, 77: 398-405.<br />

082<br />

Novel inhibitors of the arginine methyltransferase PRMT6<br />

Wagner, T. 1; Jung, M. 1<br />

1 Institute of Pharmaceutical Sciences, Albert-Ludwigs-<strong>Universität</strong> Freiburg, Albertstr. 25, 79110<br />

Freiburg, Germany<br />

Background<br />

Arginine protein methyltransferases (PRMTs) represent a SAM dependent class of protein<br />

modifying enzymes that are able to mono- and dimethylate the guanidino group of specific<br />

arginine residues within certain protein substrates e.g. histone proteins, p53, FOXO<br />

transcription factor and other protein modifying enzymes where PRMTs usually methylate<br />

glycine-alanine-arginine patches (GAR motifs). The human family of PRMTs consists of<br />

eleven members (PRMT1-11) which are devided into two classes. Type I<br />

methyltransferases are able to methylate arginine residues in an asymmetrical manner and<br />

type II methyltransferases catalyse the formation of symmetrically substituted arginine<br />

residues. PRMTs 1–4, 6 and 8 are type I enzymes and PRMTs 5, 7 and 9 are PRMTs of<br />

type II. [1] PRMTs are involved in the regulation of various physiological processes like<br />

apoptosis, cell differentiation, metabolism, DNA recombination and HIV tat transactivation.<br />

Thus, PRMT inhibitors are interesting potential compounds for drug discovery. [2]<br />

Inhibitor search is highly dependent on high throughput screening (HTS) which relies on<br />

suitable assay systems. Here we used a new homogeneous bead based in vitro assay<br />

system for screening new PRMT6 inhibitors. Therefore we surveyed some known PRMT1<br />

inhibitors that have been identified in our group previously.<br />

Material and Methods<br />

A method for the expression and purification of PRMT1 was set up in order to be able to<br />

screen larger amounts of compounds as potential inhibitors of this enzyme. Compounds<br />

have been screened and PRMT1 inhibitors have been identified by the use of an ELISA<br />

based, heterogeneous assay system (Delfia) in our group previously. In order to test the<br />

selectivity of the PRMT1 inhibitors towards other arginine methyltransferases we used an<br />

alphaLISA based method for exploring the enzyme PRMT6. Therefore a biotinylated<br />

peptide fragment of the histone protein H3 (amino acids 1-21) is used as substrate for the<br />

enzyme PRMT6. This peptide is able to bind to a streptavidine coated donor bead which<br />

contains phthalocyanine as a photosensitizer which converts ambient oxygen to singlet<br />

oxygen upon excitation with light at a wavelength of 680 nm. In the case of the peptide<br />

being methylated by PRMT6 at the arginine residue at position 2 of the peptide after<br />

addition of the cofactor S-adenosylmethionine (SAM) an antibody directed against this<br />

modification and coated to acceptor beads is able to bind to the modified substrate but not<br />

to the unmodified state. Thereby donor and acceptor beads are brought into close proximity<br />

so that the singlet oxygen emitted by the donorbeads is able to convert a thioxene<br />

derivative on the acceptor beads. The addition product degrades with chemoluminsecence<br />

which finally leads to the excitation of the lanthanide europium which is also present in the<br />

acceptor beads. The latter can be detected by time resolved fluorescence readout in an<br />

EnVision plate reader (commercial detection system by PerkinElmer) at a wave length of<br />

615nm.<br />

Results<br />

We were able to profile PRMT inhibitors against PRMT6 by using a new alphaLISA based<br />

assay system. Most compounds inhibit both PRMT1 and PRMT6 in low micromolar<br />

concentrations. But we could further show selectivity for PRMT1 towards PRMT6 for some<br />

compounds. We also identified one compound that seems to inhibit PRMT6 selectively<br />

compared to PRMT1. The presented assay protocols are also suitable for HTS of larger<br />

compound libraries.<br />

References:<br />

1. Bissinger, E.-M. et al.: Med. Chem. Commun. 2010, 1(2): 105–172.<br />

2. Bissinger, E.-M. et al.: Bioorg. Med. Chem. 2011, 19(12): 3717–3731.<br />

083<br />

The estrogen receptor as a target for anabolic effects of phytoecdysteroids in<br />

mammals<br />

Haupt, O1); Tchoukouegno Ngueu, S1); Diel, P1); Parr, MK1 2)<br />

1 Center for Preventive Doping Research, German Sport University, Cologne, Germany<br />

2 Institute of Pharmacy, Freie <strong>Universität</strong> Berlin, Germany<br />

Phytoecdysteroids, such as ecdysterone (chemical structure in figure 1), are structurally<br />

similar or identical to insect-moulting hormones and have been detected in various plant<br />

species. In mammals ecdysteroids are reported to produce a range of effects including the<br />

stimulation of protein synthesis and physical performance. Therefore, ecdysterone is<br />

promoted as active component in several over-the-counter supplements and may be<br />

misused by athletes to increase muscle mass.<br />

The anabolic-androgenic potency of ecdysterone was studied in-vivo. In a Hershberger<br />

assay analogue design male Wistar rats were treated for 21 days either with 5 mg/kg<br />

ecdysterone s.c. or placebo. After necropsy, the weights of the prostate, seminal vesicle<br />

and levator ani muscle were measured. The muscle fiber size of the soleus muscle and the<br />

gastrocnemius muscle was determined. The anabolic effect of ecdysterone resulted in an<br />

112 Poster


increase of the fiber size in both muscles. An androgenic effect could not be observed.<br />

In order to elucidate the molecular mechanism involved in the anabolic activity of<br />

ecdysterone, cell culture experiments were conducted using the mouse skeletal muscle cell<br />

line C2C12. Differentiation of C2C12 cells towards myotubes was induced. The resulting<br />

myotubes were incubated in the presence of ecdysterone, dihydrotestosterone (DHT) and<br />

antiandrogenic or antiestrogenic substances, such as flutamide (an antiandrogen) and ZK<br />

191703 (an antiestrogenic substance). Additionally co-incubation of with ecdysterone and<br />

the antagonists was performed. Cells were fixed and myotube diameters were determined<br />

by glutaraldehyde-induced autofluorescence and morphometry.<br />

The treatment of the cells with ecdysterone resulted in an increased myotube-diameter.<br />

This stimulation could be antagonized by an antiestrogen, which suggests an estrogen<br />

receptor mediated mechanism. Flutamide, an anti-androgen, could not antagonize this<br />

effect, which supports our hypothesis that the anabolic effect of ecdysterone is mediated by<br />

an estrogen receptor mediated pathway.<br />

In summary these observations are relevant for the development of new strategies for the<br />

treatment of skeletal muscle injuries such as sarcopenia and cachectic diseases but also to<br />

provide scientific data that allow for the classification of ecdysterone in preparations.<br />

Currently in Germany a categorization either as novel food or as pharmaceutical is<br />

discussed.<br />

Figure 1: Chemical structure of ecdysterone<br />

084<br />

Regulation of 5-lipoxygenase gene expression by AF4 and MLL as<br />

transcriptional regulators<br />

Ahmad, K. 1; Marschalek, R. 2; Steinhilber, D. 1<br />

1 Institute of Pharmaceutical Chemistry, Max-von-Laue-Str. 9, D-60438 Frankfurt am Main,<br />

Germany<br />

2 Institute of Pharmaceutical Biology, Max-von-Laue-Str. 9, D-60438 Frankfurt am Main, Germany<br />

Besides its function in inflammation, recent findings support a role of 5-lipoxygenase (5-LO)<br />

as critical regulator of leukemia, creating a further link between inflammation and cancer<br />

[1]. The mechanism of this interaction still remains unclear.<br />

5-LO which is encoded by the ALOX5 gene is an enzyme that catalyses the first two steps<br />

in the biosynthesis of the pro-inflammatory leukotrienes derived from arachidonic acid [2]. It<br />

is known that ALOX5 gene expression is controlled by 1,25-dihydroxyvitamin D3 (calcitriol)<br />

and transforming growth factor β (TGFβ) [3]. The resulting upregulation of the 5-LO gene<br />

expression is mediated by the binding of TGFβ effector proteins SMADs and the vitamin D<br />

receptor (VDR) to response elements within the 5-LO gene and is due to transcript<br />

elongation [4,5].<br />

Concerning the transcript elongation it is described that the AF4 (ALL-1 fused gene on<br />

chromosome 4) protein is involved in the regulatory process of the transcript elongation and<br />

exhibits chromatin remodelling activities [6]. Furthermore the chromosomal translocation of<br />

the AF4 gene and the MLL (mixed lineage leukemia) gene leads to the production of der4<br />

(AF4-MLL) and der11 (MLL-AF4) fusion proteins. These fusion proteins are also associated<br />

with the pathomechanism of leukemia [7].<br />

To elucidate the involvement of AF4, MLL and their fusion proteins on the 5-LO gene<br />

expression, reporter gene assay experiments were performed. Thus different 5-LO reporter<br />

gene constructs were cotransfected with AF4, MLL, der4 or der11.<br />

The results revealed that both AF4 and MLL are involved in the control of 5-LO gene<br />

expression as transcriptional regulators. Furthermore the presented data suggest that der4<br />

and der11 are able to deregulate the 5-LO gene expression either in the step of transcript<br />

initiation or at the step of transcript elongation. Thus, MLL and its fusion proteins provide a<br />

potential link between 5-LO and leukemia development.<br />

Acknowledgments: Hans Kröner - Graduiertenkolleg<br />

References:<br />

1. Chen, Y. et al.: Nat Genet. 2009, 41(7): 783-92.<br />

2. Rådmark, O. et al.: Trends Biochem Sci. 2007, 32(7): 332-41<br />

3. Brungs, M. et al.: Proc. Natl. Acad. Sci. USA 1995, 92(1): 107-11<br />

4. Seuter, S., Sorg, BL., Steinhilber, D.: Biochem Biophys Res Commun. 2006, 348(4): 1403-10<br />

5. Stoffers, KL. et al.: J Mol Biol. 2010, 395(4): 884-96<br />

6. Bitoun, E., Oliver PL., Davies, KE.: Hum Mol Genet. 2007, 16(1): 92-106<br />

7. Gaussmann, A. et al.: Oncogene. 2007, 26(23): 3352-63<br />

085<br />

In vitro study of colonic microbial metabolization and absorption kinetics of<br />

olsalazine in the TIM-2 model simulating the conditions in the proximal large<br />

intestine<br />

Glöckl G1;Venema K2;Wilson CG3; Weitschies W1 1Institute of Pharmacy, <strong>Ernst</strong> <strong>Moritz</strong> <strong>Arndt</strong> University, <strong>Greifswald</strong>, Germany<br />

2TNO Healthy Living, Zeist, The Netherlands<br />

3Institute of Pharmacy and Biomedical Sciences, Strathclyde University, Glasgow, UK<br />

It is well-known that the colonic epithelium expresses a lot of transporters, either for uptake<br />

or efflux, as well as metabolizing enzymes. The epithelial metabolism is superimposed by<br />

the metabolic action of the microbiota colonizing the human colon. The complex interplay<br />

between these processes is the origin of the limited understanding of the colonic absorption<br />

process. The latter is of special importance when administering standard oral extended<br />

release formulations.The microbial metabolic activity is difficult to investigate in vivo<br />

(location) as well as in vitro (anaerobic environment). Therefore, TNO’s in vitro model of the<br />

large intestine (TIM-2) wasdesignedto mimic the conditions in the proximal part ofthe<br />

human colon [1].<br />

Some prodrugs, e.g. for the medication of chronic inflammatory bowel diseases, are<br />

specifically cleaved in the colon by means of microbial reducing enzymes. The aim of the<br />

presented study is to investigate in vitrohow the azo-prodrugolsalazineis cleaved by<br />

faecalmicrobiota and further absorbed using the TIM-2 model.<br />

The computer controlled TIM-2 system consists of glass tubes and flexible walls allowing<br />

the content to be mixed in a peristaltic movement. The device is continuously flushed with<br />

nitrogen generating an anaerobic environment and kept at body temperature. The pH is<br />

adjusted to 5.8 by adding NaOH. The system is further equipped with a set of hollow fiber<br />

membranes allowing the fermentation products (predominately short chain fatty acids) to be<br />

eliminated from the system. Therefore, a dialysis liquid containing electrolytes and vitamins<br />

in a phosphate buffer pH 5.8 is pumped through the system. The volume of the luminal<br />

content is kept at 120 mL using an integrated level sensor.For a continuous measurement<br />

of the luminal redox potential an electrode (InLab ® Redox, Mettler) was integrated in a<br />

specially designed glass compartment.<br />

The system is inoculated with standard pooled faecalmicrobiota obtained from healthy<br />

volunteers. For feeding, a standard ileal efflux medium mainly consisting of peptides and<br />

polysaccharides is continuously delivered to the system. Each day 25 mL of luminal<br />

contentare withdrawn and replaced by dialysis liquid to simulate passage of stool.<br />

Olsalazine was utilized as model drug to evaluate the metabolic function of the microbiota.<br />

The prodrugolsalazine is cleaved by microbial azoreductases to form two molecules of the<br />

active mesalazine. Both compounds can easily be quantified with UV/Vis spectrometry in<br />

the lumen as well as in the dialysate.<br />

After addition of 500 mg of olsalazineto the TIM-2 system via the sampling port we could<br />

measure the elimination of olsalazine and the appearance and subsequent elimination of its<br />

metabolite mesalazine. The elimination rate was inversely proportional to the molecular<br />

weight and thus faster for mesalazine.<br />

The integration of a redox electrode into the system allowed the continuous measurement<br />

of the intraluminal redox potential. After an initial drop of the potential in the growth phase<br />

of the microbiota(adaptation period of about 16 hours) a strongly reducing environment was<br />

generated (stable ecosystem maintained for at least 48 hours). During this period, the<br />

system is suitable to simulate the metabolization and absorption of the model drug<br />

olsalazine and probably of other actives. In future studies we will further check if these data<br />

can be correlated with in vivo data.<br />

Acknowledgments: G.G. thanks W. Borst, A. Maathuis, J. Lelieve d and M. Minekus (TNO) for their<br />

assistance and GalenusPrivatstiftung for a personal grant. The project was funded by the German federal<br />

ministry of education and research (InnoProfile, FKZ: 03IP612).<br />

References:<br />

1. Minekus, M. et al.:Appl. Microbiol. Biotechnol.1999, 53(1): 108-114.<br />

086<br />

Development of long-term stable hydrogel matrices for a vessel-simulating<br />

flow-through cell<br />

Semmling, B. 1; Seidlitz, A. 1; Nagel, S. 1; Schütt, R. 1; Brand, T. 1; Grabow, N. 2; Sternberg, K. 2;<br />

Weitschies, W. 1<br />

1 Institute of Pharmacy, EMA University of <strong>Greifswald</strong>, Felix-Hausdorff-Straße 3, 17487 <strong>Greifswald</strong>,<br />

Germany<br />

2 Institute for Biomedical Engineering, University of Rostock, Friedrich-Barnewitz-Straße 4, 18119<br />

Rostock, Germany<br />

Natural polymeric gels are often unstable and very sensitive to erosion. Therefore synthetic<br />

polymeric materials, gelling agents such as polyvinylalcohol (PVA) or polyacrylamide (PAA)<br />

are interesting due to various properties: stability, elasticity, diffusibility, hydrophilicity, nontoxicity.<br />

Modifications of the composition and the cell entrapment in the hydrogel are<br />

feasible. Drug dissolution as well distribution processes of coated drugs of drug-eluting<br />

stents (DES) were determined in the vessel-simulating flow-through cell (vessel-simulating<br />

FTC) [1]. The current study focuses on the approach of developing long-term stable<br />

hydrogel matrices for the vessel-simulating FTC by replacing the conventional polymer<br />

matrix (2 % (w / w) sodium alginate matrix, AM) by 15 % (w / w) PVA-hydrogel matrices<br />

(PVA-M), 10 % (w / w) PAA matrices (PAA-M) and 2 % (w / w) agarose hydrogel matrices<br />

(Ag-M). The distribution coefficient (DC) of the fluorescent model drug substance<br />

triamterene between the hydrogel and aqueous media, long-term stability of the novel<br />

hydrogel compartments and drug distribution between the three compartments: hydrogel,<br />

dissolution media and stent were investigated.<br />

The distribution coefficient of the model substance between the investigated long-term<br />

stable hydrogel matrices and dissolution media indicate differences in dependency on the<br />

respective hydrogel matrix (data Ag-M and PAA-M not shown): Compared to the reference<br />

matrix (DC of AM after 72 h was found to be about 1.4) the drug delivered into the hydrogel<br />

compartment accumulated up to 2.3 (after 72 h, PVA-M). Test results of the examination of<br />

the long-term stability of novel hydrogel matrices show no signs of gel erosion within 4<br />

weeks perfusion (data not shown) indicating mechanical long-term stability of the novel<br />

hydrogels under typical test conditions.<br />

Release profiles from triamterene coated DES (Eudragit ® RL / RS 30 / 70, dissolution<br />

media PBS 7.4, dissolution temperature 37 °C ± 0.5 °C, flow rate 35 mL / min [2]) obtained<br />

with the vessel-simulating FTC using the novel matrices (PAA-M, PVA-M and Ag-M)<br />

indicate a fast increase in the amount of dissolved drug in the dissolution media (Ag-M:<br />

82 % after 12 h) with a simultaneous fast decrease in residual drug amount in the DES<br />

coatings (Ag-M: 14 % after 12 h). After 12 hours 4 % of the total drug amount was detected<br />

in the agarose matrices. In comparison to release profile of fluorescent model drug using<br />

Ag-M, a slightly different dissolution profile of drug release using the reference hydrogel<br />

matrix (AM) could be observed. After 12 hours about 77 % of the total drug amount eluted<br />

from DES into dissolution media was detected. The residual drug amount in stent coating<br />

increases continuously and was found to be about 18 % after 12 hours. At the same<br />

sampling point the total drug amount in the sodium alginate hydrogel compartment<br />

accumulated up to 5 %. The difference in detected drug amount in the compartments<br />

dissolution media, hydrogel (AM, Ag-M, PAA-M and PVA-M) and DES may be attributed to<br />

different distribution coefficient of triamterene between the hydrogel compartments and<br />

PBS 7.4.<br />

In conclusion, the methods of hydrogel matrix preparation (PAA-M, PVA-M and Ag-M) were<br />

established successfully. The stability studies confirmed sufficient mechanical robustness<br />

of the matrices under long-term test conditions. Additionally, the developed matrices for the<br />

vessel-simulating FTC enable the determination of delivered drug not only in dissolution<br />

media representing the blood, but also in hydrogel matrices simulating the vessel wall as<br />

target organ structure. Consequently, the use of the gel compartment allows the adaption of<br />

the in vitro test closer to the physiological situation and therefore enables the estimation of<br />

the drug distribution of DES under more realistic test conditions.<br />

Poster 113


Acknowledgments: Financial support by the Federal Ministry of Education and Research (BMBF) within<br />

REMEDIS “Höhere Lebensqualität durch neuartige Mikroimplantate” (FKZ: 03IS2081) is gratefully<br />

acknowledged.<br />

References:<br />

1. Neubert, A. et al.: J. Control. Release 2008, 130(1): 2-8.<br />

2. Di Mario, C et al.: Am. J. Cardiol. 1993, 71(14): 54D-61D.<br />

087<br />

New titanium(IV) complexes as potential anticancer drugs<br />

Schur, J. 1; Manna, C.M. 2; Tshuva, E.Y. 2; Deally, A. 3; Tacke, M. 3; Köster, R. 4, Ott, I. 1<br />

1 Institute of Medicinal and Pharmaceutical Chemistry, TU Braunschweig, Beethovenstr. 55, 38106<br />

Braunschweig, Germany<br />

2 The Hebrew University of Jerusalem, Department of Inorganic Chemistry, 91904 Jerusalem,<br />

Israel<br />

3 University College Dublin, UCD School of Chemistry and Chemical Biology, Belfield, Dublin 4,<br />

Ireland<br />

4 Zoological Institute, Department of Cell Physiology, TU Braunschweig, Spielmannstr. 8, 38106<br />

Braunschweig, Germany<br />

Since the serendipitous discovery of the antitumor agent cisplatin in the 1960s, research on<br />

metal-based compounds has made remarkable progress. Nowadays, platinum-based<br />

metallodrugs (e.g. oxaliplatin) play a major role in medical anticancer treatment.<br />

Nevertheless, the activity is limited to a small spectrum of cancers because of increasing<br />

resistance phenomena of these drugs and several side effects caused by high toxicity.<br />

Therefore, in the last two decades, research has been extended to non-platinum<br />

metallodrugs.<br />

An innovative compound in the 1990s was the titanium-based metallodrug titanocene<br />

dichloride that reached clinical trials but failed because of decreased hydrolytic stability and<br />

formulation problems [1]. Based on these promising works and with the aim of improving<br />

cytotoxicity and hydrolytic stability better stabilized titanium-containing compounds e.g.<br />

titanocene Y and a new class of non-Cp diamino bis(phenolato) “salan” Ti(IV) complexes<br />

were recently developed and seem to hold a high potential for anticancer therapy [2,3].<br />

Figure 2: Metallodrugs: (a) Oxaliplatin, (b) Titanocene dichloride, (c) Titanocene Y, (d)<br />

Salan-Ti(IV) complex<br />

In our study the mentioned lead compounds were investigated biologically in comparison to<br />

titanocene dichloride. Very interesting properties for the novel class of salan-Ti(IV)<br />

complexes (see figure 1) and titanocene Y (as a substituted titanocene) could be obtained<br />

and may be a step forward in target identification of titanium-based anticancer drugs.<br />

Figure 3: Zebrafish embryos (94 hpf) exposed to 100 µM Salan-Ti(IV)<br />

The lead compound of new diamino salan-Ti(IV) complexes and titanocene Y have an<br />

improved cytotoxicity in the low micromolar range and a higher accumulation in cancer cells<br />

compared to titanocene dichloride. A relationship between the antiproliferative activity, the<br />

cellular uptake and the protein binding ability of the tested Ti-containing compounds was<br />

noted. In addition, the toxicity of the new titanium-based complexes in zebrafish embryos [4]<br />

was studied and no toxic effects could be observed up to a concentration of 100 µM (see<br />

figure 2). Further investigations were performed on the DNA-binding efficacy, the nuclear<br />

drug content and the drugs accumulation in mitochondria. Our results indicate that novel<br />

titanium(IV) complexes possibly have overcome the disadvantages of the well known<br />

titanocene dichloride, which underlines their high potential as anticancer drugs.<br />

References:<br />

1. Melendez, E., Crit. Rev. Oncol. Hematol. 2002, 42: 309–315.<br />

2. Sweeney, N.J. et al., J. Organomet. Chem. 2005, 690: 4537–4544.<br />

3. Peri, D. et al., Chem. Eur. J. 2009, 15: 2403–2415.<br />

4. Parng, C. et al., ASSAY Drug Dev. Technol. 2002, 1: 41–48.<br />

088<br />

Anti-infectives with novel mode of action: Interruption of P. aeruginosa cell-tocell<br />

communication by PqsD inhibitors<br />

Maurer, C. K. 1; Storz, M. P. 1; de Jong, J. C. 1; Weidel, E. 1; Zimmer, C. 1; Steinbach, A. 1;<br />

Hartmann, R. W. 1 2<br />

1 Helmholtz-Institute for Pharmaceutical Research Saarland, Campus C2.3, 66123 Saarbrücken,<br />

Germany<br />

2 Department of Pharmaceutical and Medicinal Chemistry, Saarland University, Campus C2.3,<br />

66123 Saarbrücken, Germany<br />

The opportunistic pathogen P. aeruginosa controls the coordinated production of virulence<br />

factors as well as biofilm formation via a cell density dependent cell-to-cell communication<br />

system known as quorum sensing (QS) [1]. The pqs QS system employs the signal<br />

molecules PQS and its precursor HHQ to activate PqsR [2], a transcriptional regulator that<br />

drives the expression of many genes involved in pathogenicity.<br />

PqsD is a key enzyme in the biosynthesis of HHQ and PQS [3]. Since reduction of HHQ<br />

production is accompanied by reduced mortality in infected mice [4], PqsD proves to be an<br />

interesting target for drug discovery.<br />

Two different drug design strategies were applied: (A) a ligand based approach including<br />

analogues of the natural substrate anthraniloyl-CoA (ACoA) and mimics of the<br />

corresponding transition state as well as (B) the development of compounds derived from<br />

known inhibitors of FabH, an enzyme, which is structurally and functionally related to PqsD<br />

[5,6]. Thereby, inhibitors with IC50 values in the nM to low µM range have been identified.<br />

SPR experiments revealed, that, while (2-nitrophenyl)methanol compounds derived from<br />

approach (A) resulted in time-dependent irreversible PqsD inhibition with binding within the<br />

anthraniloyl binding site, 2-benzamidobenzoic acid derivatives from (B) are reversible<br />

inhibitors that do not occupy the anthraniloyl binding site.<br />

The effect of representative compounds from both classes on the extracellular levels of the<br />

signalling molecules HHQ and PQS in P. aeruginosa PA14 was investigated. While<br />

2-benzamidobenzoic acids had no significant effect, (2-nitrophenyl)methanol derivatives<br />

strongly reduced extracellular HHQ and PQS levels without affecting cell viability.<br />

References:<br />

1. Dubern, J. F.; Diggle S. P.: Mol. BioSyst. 2008, 4: 882.<br />

2. Cao, H. et al.: Proc Natl Acad Sci USA 2001, 98(25): 14613-8.<br />

3. Pistorius, D. et al.: ChemBioChem. 2011, 12(6): 850-3.<br />

4. Lesic, B. et al.: PLoS Pathogens 2007, 3: 1229.<br />

5. Bera, A. K. et al.: Biochemistry 2009, 48: 8644-8655.<br />

6. Nie, Z. et al.: J. Med. Chem. 2005, 48: 1596-1609.<br />

089<br />

A Vascular function of Cyclin dependent kinase 5 (Cdk5)<br />

Liebl J. 1, Moser M. 2, Hager B. 1, Zhang S. 1, Fürst R. 1, Bibb J. A. 3, Adams R. H. 4, Vollmar A.<br />

M. 1, Zahler S. 1<br />

1Center of Drug Research, Ludwig-Maximilians University, Munich, Germany<br />

2Department of Molecular Medicine, Max Planck Institute of Biochemistry, Martinsried, Germany<br />

3Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, Texas<br />

4Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, Münster,<br />

Germany<br />

Cyclin-dependent kinase 5 (Cdk5) is essential for CNS development and function. Whereas<br />

its role in neurons has been studied in detail, there still exists only limited knowledge about<br />

its functions in other systems. During recent years, more and more reports indicated<br />

functions of Cdk5 in non-neuronal tissues and we could show that Cdk5 regulates<br />

endothelial cell migration in vitro.<br />

This work elucidates an important function of Cdk5 in the vascular system in vivo. We<br />

specifically disrupted the Cdk5 gene in the mouse endothelium using the Cre-loxP system.<br />

Floxed Cdk5 mice were intercrossed with mice expressing Crerecombinase under control<br />

of the Tie2 promoter (Tie2Cre) or a tamoxifen-inducible VE-Cadherin promoter<br />

(Cdh5(PAC)-CreERT2), respectively. Breedings of Cdk5fl/wtTie2Cre males and Cdk5fl/fl<br />

females resulted in offspring in which only 15% pups harbored the Cdk5fl/flTie2Cre<br />

genotype instead of expected 25% and most of the endothelial-specific Cdk5 knockout<br />

mice did not survive past postnatal day 28. Analysis of the genotypes of embryos at<br />

different stages indicated that Cdk5fl/flTie2Cre embryos frequently died during late<br />

embryogenesis between E16.5 and E18.5. Endothelial-specific Cdk5 knockout leads to a<br />

non-separation phenotype between blood and lymphatic system including edema<br />

formation, blood-filled and dilated lymphatic vessels, defective lymphatic valve formation,<br />

and impaired draining function of the lymphatic system. Concerning blood vessels, we<br />

found a hypervascularization of endothelial-specific Cdk5 knockout mice. This was<br />

indicated by an increased number of branching points in the yolk sacs and skin of E16.5<br />

Cdk5fl/flTie2Cre embryos and increased branching and sprouting in the developing retina<br />

of d6 pups with Cdk5fl/flCdh5(PAC)-CreERT2 genotype after tamoxifen treatment as well<br />

as after pharmacological inhibition of Cdk5 with the small molecule roscovitine. Most<br />

importantly, we found a reduction of tumor growth of subcutaneous implanted wildtype<br />

B16F1 melanoma xenografts in endothelial-specific Cdk5 knockdown mice. Histological<br />

analysis of tumors showed an immature vascularization of tumors grown in<br />

Cdk5fl/flCdh5(PAC)-CreERT2 mice.<br />

To sum up, our results reveal an important vascular function of Cdk5 in angiogenesis and<br />

lymphangiogenesis, suggesting Cdk5 as druggable target for anti-angiogenic therapy.<br />

090<br />

A novel assay for the analysis of inhibitors of ergosterol biosynthesis<br />

Staudacher V; Müller C; Bracher F<br />

Ludwig-Maximilians-<strong>Universität</strong> München, Department für Pharmazie - Zentrum für<br />

Pharmaforschung, Butenandtstr. 5-13, 81377 München, Germany<br />

Systemic fungal infections are of increasing importance in intensive care, due to the rising<br />

portion of immunocompromised patients (cancer, AIDS, and organ transplantation patients)<br />

in clinical routine. These infections are characterized by a very high mortality, and very<br />

potent and fast-acting antifungal agents are needed for life-saving therapy. Moreover,<br />

resistance to established antifungal drugs (e.g. fluconazole) gains in clinical significance.<br />

Ergosterol is the central sterol in fungal metabolism, and inhibition of ergosterol<br />

biosynthesis represents a classical, but still highly effective strategy in antifungal therapy.<br />

Over the decades, inhibitors of several enzymes in ergosterol biosynthesis have been<br />

marketed, the most prominent ones being the azole-type inhibitors of the enzyme C14demethylase.<br />

The morpholine antifungal amorolfine inhibits two enzymes in ergosterol<br />

biosynthesis, but is not suitable for systemic application, and the squalene epoxidase<br />

inhibitors (naftifine, terbinafine) mainly target dermatophytes.<br />

Notably, the post-squalene part of ergosterol biosynthesis contains a significant number of<br />

additional enzymes which occur only in fungi, but not in mammals, and therefore are<br />

potential targets for new, selective antifungal drugs. In our ongoing research on the<br />

development of new antifungals [1] we focused on new inhibitors of enzymes in ergosterol<br />

biosynthesis. In order to perform effective optimization of first lead structures, there was a<br />

need to work out a screening system which enables us to (a) identify the target enzyme in<br />

ergosterol biosynthesis unambiguously, and (b) gain quantitative data on the effect of the<br />

inhibitors on overall ergosterol biosynthesis.<br />

For this purpose we developed a convenient whole-cell screening assay, which in a first<br />

step enables us to identify substances, which interfere with enzymes of the post-squalene<br />

part of ergosterol biosynthesis, and in a second step to determine IC50 values for inhibition<br />

114 Poster


of ergosterol biosynthesis.<br />

In this assay three different yeasts, Candida glabrata, Saccharomyces cerevisiae and<br />

Yarrowia lipolytica, are treated with test substances. After alkaline cell lysis the sterols are<br />

extracted with an organic solvent, and the crude extract is purified by dispersive solid phase<br />

extraction (dSPE). After silylation the sterols are analyzed by GC-MS.<br />

The obtained sterol patterns provide an indication of the inhibited enzyme(s), since<br />

inhibition of defined enzymes leads to the accumulation of specific sterols. By this way we<br />

can identify inhibitors of any enzyme in the post-squalene part of ergosterol biosynthesis in<br />

one single cellular assay, with no need for having the single purified enzymes in hands.<br />

Furthermore, we extended this qualitative screening method to a quantitative determination<br />

of the effect on total ergosterol biosynthesis. In analogy to a screening system worked out<br />

by us for inhibitors of cholesterol biosynthesis recently [2], incubation with the inhibitors in<br />

the presence of 13C-acetate, followed by GC-MS analysis of the labeled ergosterol isotopes<br />

allows for the convenient determination of IC50 values of the inhibitors.<br />

The system was calibrated with several known antifungals targeting ergosterol<br />

biosynthesis, and meanwhile applied to the identification of the target enzymes of a number<br />

of new inhibitors of ergosterol biosynthesis.<br />

References:<br />

1. Renard D, Perruchon J, Giera M, Müller J, Bracher F: Bioorg. Med. Chem. 2009, 17(23) 8123-8137.<br />

2. Giera M, Plössl F, Bracher F: Steroids 2007, 72(8): 633-642.<br />

091<br />

Cysteine-rich LIM-only protein 4 (CRP4) opposes angiotensin II induced<br />

pathological heart hypertrophy and fibrosis<br />

Straubinger, J.; Majer, M.; Ruth, P.; Lukowski, R.<br />

Institute of Pharmacy, Department of Pharmacology, Toxicology and Clinical Pharmacy, University<br />

of Tübingen, Germany<br />

Background: Cardiac hypertrophy is an adaptive response of the heart to many cardiovascular<br />

disorders including hypertension, infarction and defects of the valves. Although<br />

physiological heart hypertrophy, which refers to an adaption of the heart muscle to healthy<br />

exercise or pregnancy, and pathological hypertrophy share common features, only the<br />

pathological remodelling of the heart leads to fibrosis, increased diastolic stiffness, fatal<br />

failure and death. Clinical studies and functional analysis of genetically modified mice have<br />

established that natriuretic peptides (NP) like ANP and BNP and their cardiac receptors<br />

diminish development of pressure and volume induced heart hypertrophy and fibrosis via<br />

the second messenger cyclic guanosine-3´,5´-monophosphate (cGMP). However, the<br />

signaling events downstream of cGMP and its major effector cGMP kinase I (cGKI) are less<br />

clear. Here, we studied the role of the cysteine-rich LIM-only protein (CRP4) for the cardiac<br />

response to an increase in afterload and healthy exercise training. CRP4 is directly targeted<br />

by cGKI in the cardio-vascular system and a highly related homologue of the muscle LIM<br />

protein CRP3/MLP. CRP/MLP knockout mice develop a dilated cardiomyopathy and<br />

defects in CRP3/MLP are the cause for dilated and hypertrophic cardiomyopathies in<br />

human.<br />

Methods: Gene-targeted CRP4 knockout (KO) mice and their heterozygous (HET) and wild<br />

type (WT) littermates were subjected to a chronic pressure-dose of angiotensin II (AngII).<br />

Cardiac hypertrophy, as defined by the heart-to-body weight ratios (HW/BW), was<br />

determined in AngII treated mice and compared to littermate animals that received saline or<br />

were trained by a duration-controlled swimming exercise protocol to induce a physiological<br />

adaption of the heart muscle. Changes in hypertrophy marker genes, putative effects of<br />

AngII on components of the NP/cGMP pathway and the expression pattern of other<br />

members of the CRP protein family were analyzed in total mRNA and protein isolated from<br />

the ventricles. These experiments were corroborated by the localization of CRP4 in the<br />

myocardium and Sirius red stainings as a quantitative measure of fibrosis.<br />

Results: Under basal conditions and upon AngII infusion CRP4 mRNA and protein were<br />

detectable in the myocardium from WT mice, whereas CRP4 was absent from KO hearts<br />

and significantly reduced in HET mice. HW/BW ratios of all three genotypes were not<br />

different at baseline, but increased in HET and KO animals in response to infusion of AngII.<br />

Sirius red staining and quantitative RT-PCR experiments revealed an increase in interstitial<br />

fibrosis and a diminished production of anti-fibrotic factors such as BNP in HET and KO<br />

hearts. In contrast, neither the increase in cardiac mass nor the extent of fibrosis were<br />

altered upon physiological growth of the heart muscle between the different genotypes.<br />

Conclusion: Hearts of CRP4 KO mice are more susceptible to AngII-induced remodelling,<br />

whereas the lack of CRP4 does not alter physiological cardiac hypertrophy. Therefore, we<br />

conclude that CRP4 is a beneficial mediator in NP/cGMP/cGMP kinase I signaling to<br />

oppose pathological heart growth and fibrosis induced by cardiac Gαq-pathways.<br />

092<br />

Comparison of binding requirements in a series of benzimidazol-2-yl-aminosubstituted<br />

(L)-amino acids by simple QSAR models for the NMDAR glycineB<br />

site and the transporter LAT1<br />

Krauß, A. 1; Rotmann, A. 2 3; Martiné, U. 2; Closs, E. I. 2; Dannhardt, G. 1<br />

1 Institute of Pharmacy and Biochemistry, Johannes Gutenberg-University, Staudingerweg 5,<br />

55128 Mainz, Germany<br />

2 Department of Pharmacology, Johannes Gutenberg-University, Obere Zahlbacher Straße 67,<br />

55101 Mainz, Germany<br />

3 Department of Parasitology, Ruprecht Karls-University, Im Neuenheimer Feld 324, 69120<br />

Heidelberg, Germany<br />

Central bioavailability is a major concern in the development of new ligands targeting the<br />

glycine binding site of the NMDA receptor, as potential applications comprise inter alia<br />

epilepsy, chronic pain, dementia (Alzheimer’s disease, Parkinson’s disease) or<br />

schizophrenia [1]. Uptake by amino acid transporters [2], especially large neutral amino<br />

acid transporters (system L), is therefore considered as a versatile strategy to cross the<br />

blood-brain barrier without disruption. The concept was proven for the low affinity glycineB<br />

site antagonist 4-chlorokynurenine as reviewed in [3], and recently applied to analogues of<br />

pregabalin [4] and analogues of melphalan [5].<br />

This work is based on a congeneric series of benzimidazol-2-yl-amino-substituted (L)amino<br />

acids obtained in a seven-step linear synthesis and assayed for NMDAR glycineB<br />

site binding (displacement of [ 3H]-MDL-105,519 from pig cortical membranes) as well as<br />

system L activity (competition with [ 3H]-L-Phe uptake in C6 rat glioblastoma cells). Two<br />

corresponding QSAR models have been derived, utilizing GraphPad Prism 3.00, GraphPad<br />

Instat 3.06 and NLREG 6.4. for stepwise multiple (non)linear regression. The NMDAR-<br />

QSAR suggests a parabolic relationship with STERIMOL parameter L and, likewise<br />

negative but linear, correlations with Swain and Lupton R and the hydrogen bond indicator<br />

variable HD. The LAT-QSAR discloses an increasing lipophilicity (ClogP) and a decreasing<br />

pka value (of the guanidino group, mainly experimental) as favourable for affinity. Taken<br />

together, within a restricted applicability domain, parallel steric resp. lipophilic demands<br />

dominate a certain opposite trend in the electronic effects. Semi-empirical quantum<br />

mechanical calculations with Spartan 04 (Wavefunction, Inc.) at the HF 3-21G(*) level [6]<br />

may explain these electronic prerequisites partly by =N---HN intramolecular hydrogen<br />

bonding, this proving advantageous for the NMDA receptor, but disadvantageous for LAT.<br />

However, the pronounced conformational flexibility of the scaffold allows for various binding<br />

modes, each with the free amino acid moiety as the most important and common motif.<br />

Moreover, compared to the rather specific and sterically demanding interactions at the<br />

glycineB site of the NMDAR [7], the system L-type transporters are known for their broad<br />

substrate profile, ranging from large neutral (LAT1) to large and small neutral (LAT2) amino<br />

acids [8]. A joint further development of compounds considering both targets is therefore<br />

regarded as possible.<br />

Acknowledgements: The VCCLAB (Virtual Computational Chemistry Laboratory, http://www.vcclab.org) is<br />

appreciated as a fast and user-friendly access to calculated log P values, enabling a comparison of different<br />

methods [9].<br />

References:<br />

1. Bauman, A. et al.: J. Label. Compd. Radiopharm. 2011, 54(10): 645–656.<br />

2. Closs, E. I. et al.: J. Nutr. 2004, 134(10): 2752S–2759S.<br />

3. Kohl, B. K.; Dannhardt, G.: Curr. Med. Chem. 2001, 8(11): 1275–1289.<br />

4. Belliotti, T. et al.: J. Med. Chem. 2005, 48(7): 2294–2307.<br />

5. Matharu, J. et al.: Bioorg. Med. Chem. Lett. 2010, 20(12): 3688–3691.<br />

6. Krauß, A.; Dannhardt, G.: Joint Meeting of the Czech, German and Hungarian Pharmaceutical Societies,<br />

2006, Marburg, Germany Abstract C045, p.115.<br />

7. Jansen, M.; Dannhardt, G.: Eur. J. Med. Chem. 2003, 38(10): 855–865.<br />

8. Verrey, F. et al.: Pflügers Arch. - Eur. J. Physiol. 2004, 447(5): 532–542.<br />

9. Tetko, I. et al.: J. Comput. Aid. Mol. Des. 2005, 19(6): 453–463.<br />

093<br />

A comparative study of in vitro methods analyzing endothelial barrier function:<br />

conventional macromolecular permeability measurements versus<br />

sophisticated impedance sensing<br />

Hornburger, M. C. 1; Mayer, B. A. 1; Vollmar, A. M. 1; Wegener, J. 2; Fürst, R. 1<br />

1 Department of Pharmacy, Center for Drug Research, Pharmaceutical Biology, University of<br />

Munich, Butenandtstr. 5-13, 81377 Munich, Germany<br />

2Institute for Analytical Chemistry, Chemo- & Biosensors, University of Regensburg, <strong>Universität</strong>sstr.<br />

31, 93053 Regensburg, Germany<br />

Endothelial barrier dysfunction, i.e. the opening of interendothelial junctions, and the<br />

consequent formation of edema are hallmarks of many severe diseases, such as sepsis,<br />

acute lung injury, or atherosclerosis. Drugs that specifically target endothelial barrierregulating<br />

systems are lacking and there is a strong demand to identify new<br />

pharmacological targets. For the validation of these targets, the barrier function of cultured<br />

endothelial cells is analyzed by two different approaches: 1) the classic measurement of<br />

macromolecular permeability and 2) the modern real-time cell impedance-sensing methods,<br />

which employ alternating currents to analyze changes of cellular morphology. Impedance is<br />

a complex physical quantity consisting of an imaginary capacitive (cell membrane<br />

properties) and a real resistive (intercellular junction stability) contribution.<br />

We aimed at deciphering the most appropriate method for the characterization of<br />

endothelial barrier-modulating agents. Therefore, we compared macromolecular<br />

permeability measurements (two-compartment Transwell ® system, membrane pore size 0.4<br />

μm, tracer: FITC-dextran 40 kDa) with three different commercially available impedance<br />

sensing devices: (i) ECIS ® (Applied Biophysics) and (ii) xCELLigence (Roche), with the<br />

cells directly grown on gold electrodes, and (iii) cellZscope ® (nanoAnalytics), where cells<br />

are placed on a permeable membrane (pore size 0.4 μm) between two electrodes.<br />

First, we tested for the correlation between macromolecular permeability and impedance<br />

measurements. Therefore, we employed the barrier-disruptive mediators TRAP (thrombin<br />

receptor-activating peptide), histamine, and TNF-α, as well as the barrier-stabilizing<br />

compound forskolin (adenylyl cyclase activator) and the Rho kinase (ROCK) inhibitor Y-<br />

27632. The impact of TRAP and forskolin on endothelial barrier function nicely correlated in<br />

all tested systems: forskolin elevated impedance values and reduced dextran flux, whereas<br />

TRAP evoked a fast and sustained decrease of impedance and a significant increase in<br />

macromolecular permeability. Interestingly, the histamine-induced barrier breakdown could<br />

not be detected by the Transwell ® assay, whereas a rapid and transient decrease of<br />

endothelial impedance occurred. The barrier-protecting ROCK inhibitor evoked a massive<br />

and long-lasting decline of impedance, whereas dextran flux was again not affected.<br />

Second, we compared the three impedance systems in order to disclose individual<br />

advantages or limitations. A key feature of the ECIS ® system is the opportunity to break<br />

down impedance in its resistive and capacitive contributions and, moreover, to discriminate<br />

betweenchanges of cell-cell and/or cell-matrix junctions. In contrast, xCELLigence only<br />

provides information on the overall impedance magnitude, internally referenced and<br />

normalized ascell index. Independent of the applied barrier-modulating stimulus, both<br />

systems returned essentially similar results with respect to the time course of the cell<br />

layers’ integral ionic permeability. The filter insert-using cellZscope ® device, however,<br />

reached its limit of sensitivity due to the low overall resistance of the endothelial cells under<br />

study (transendothelial electrical resistance< 5 Ω cm²).<br />

In conclusion, impedance sensing is a very sensitive, label-free and easy-to-use method to<br />

monitor changes in cellular morphology in real-time. Alterations in endothelial permeability<br />

are associated with these morphological changes, which are reflected by different ion fluxes<br />

in impedance sensing and by macromolecular fluxes in the Transwell ® assay. For the<br />

evaluation of new drug targets in endothelial barrier regulation, we therefore recommend to<br />

be aware of these differences and to combine impedance sensing with measurements of<br />

macromolecular permeability, since each method alone cannot fully determine all<br />

consequences of barrier-modulating agents.<br />

094<br />

A novel mouse model for studying the in vivo functions of the sodiumactivated<br />

potassium channel Slack (Slo2.2)<br />

Bausch A1; Hausmann M1; Baranski J1, Meyerdierks N1, Ruth P1 §, Lukowski R1 §,<br />

Poster 115


1 Institute of Pharmacy, Department of Pharmacology, Toxicology and Clinical Pharmacy,<br />

University of Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, Germany<br />

§ These authors contributed equally to this work.<br />

Background: Sodium-activated potassium channels of the Slack (Slo2.2) subtype are<br />

abundantly present in many neurons [1]. On a cellular level Slack channels might be<br />

involved in action potential repolarisation, setting the resting membrane potential in some<br />

neurons, slow after hyperpolarisation, burst firing and adaptation of firing rate during high<br />

frequency firing thereby shaping neuronal activity and excitability [1]. Although the<br />

molecular properties of Slack channels have been investigated in detail [2] their exact in<br />

vivo-functions remain elusive. Evidence suggests that Slack may play an important role for<br />

physiological processes such as spatial hearing [3], olfaction [4] and nociception [5]. In<br />

addition to physiological functions Slack channels have been indicated to exert control over<br />

pathophysiological processes as cytoprotection following ischemic injury [2,6].<br />

Methods: To investigate the function of the sodium-activated potassium channel Slack<br />

(Slo2.2) in vivo we recently generated knockout mice (KO) with a targeted deletion of the<br />

pore forming exon. Additionally, using the Cre/loxP recombination system, we have<br />

established a mouse line that carries a “floxed” version of the pore forming exon and<br />

therefore enables a conditional (spatial and temporal) inactivation of the Slack gene.<br />

Reverse-Transcription-PCR (RT-PCR) and real-time-PCR were used to analyse the<br />

expression pattern and levels of Slack transcripts and transcripts of highly related<br />

potassium channels in Slack KO and littermate WT animals. Slack protein expression<br />

pattern was assessed by immunohistochemistry (IHC) and Western Blot using our novel<br />

anti-Slack-antibodies. Initial tests of the hypothesis that the Slack KO mice show a motor<br />

phenotype were performed by analysis of footprint patterns and the ability of Slack KO mice<br />

and their WT littermates to maintain balance in the “beam walk test”. Olfactory functions<br />

were investigated using food-finding (“buried pellet test”), innate preference and avoidance<br />

tests.<br />

Results: Slack KO mice are viable, fertile and do not show gross phenoptypical<br />

abnormalities under physiological conditions except for a delayed body-weight gain during<br />

the first 6 to 8 weeks after birth. Using RT-PCR we could confirm the absence of the pore<br />

region in Slack transcripts of Slack KO mice in all brain regions tested (e.g. cerebellum,<br />

neocortex and olfactory bulb). These results were confirmed by IHC analysis, which further<br />

demonstrated the absence of the Slack protein in 12-µm serial cross-sections of Slack KO<br />

brains. Although a high Slack-mRNA-level and Slack immunoreactivity were detected in<br />

brain regions that are involved in motor coordination Slack KO mice did not show an<br />

impaired motor function. All measures in the footprint pattern tests and performance results<br />

of the beam walk test were not different between Slack KOs and WT littermates.<br />

Furthermore, strong Slack-immunoreactivity was detected in the olfactory bulb with the<br />

highest protein density in the mitral cell layer and the outer plexiform layer containing the<br />

dendrites of the mitral cells. Together with the decrease in body weight of young Slack KO<br />

mice these findings raised the possibility that an olfactory disorder caused impaired<br />

suckling of the newborns. Indeed, the retrieval time for Slack KO mice to recover the buried<br />

pellet in the food-finding test was significantly prolonged compared to age- and littermatched<br />

WT mice.<br />

Conclusion: Our preliminary results indicate that Slack deficiency does not cause any<br />

gross abnormalities under physiological conditions. However, Slack ablation in sensory<br />

olfactory neurons and/or second-order neurons (e.g. periglomerular cells, mitral cells) of the<br />

olfactory bulb may be associated with mild olfactory dysfunctions. Together, we provide a<br />

powerful novel tool to study the functions and molecular details of Slack channels in a preclinical<br />

relevant model in vivo. Our ongoing and future investigations of these mice will help<br />

us to address the question whether Slack channels potentially constitute novel drug targets<br />

for stroke, epilepsy, ataxia or olfactory disorders.<br />

References:<br />

1. Bhattacharjee, A. and L.K. Kaczmarek: Trends Neurosci. 2005, 28(8): 422-8.<br />

2. Yuan, A. et al.: Neuron 2003, 37(5): 765-73.<br />

3. Yang, B., R. Desai, and L.K. Kaczmarek, J Neurosci. 2007, 27(10): 2617-27.<br />

4. Lu, S. et al.: J Neurophysiol. 2010, 103: 3311–3319.<br />

5. Tamsett, T.J., K.E. Picchione, and A. Bhattacharjee, J Neurosci, 2009, 29(16): p. 5127-34.<br />

6. Wojtovich, A.P. et al.: PLoS One 2011. 6(12): e28287.<br />

095<br />

Expression analysis of drug transporter proteins in RPMI 2650 cell line and<br />

excised human nasal mucosa<br />

Dolberg, A. M. 1; Reichl, S. 1<br />

1 Institut für Pharmazeutische Technologie, Technische <strong>Universität</strong> Carolo-Wilhelmina zu<br />

Braunschweig, Mendelssohnstraße 1, 38106 Braunschweig, Germany<br />

The nasal mucosa is an interesting site for application of drugs with systemic action, since it<br />

has a number of advantages as a delivery route, including easy accessibility, extensive<br />

vascular supply and avoidance of gastrointestinal degradation as well as first pass<br />

metabolism. Therefore more and more applications for nasal systemic drug delivery have<br />

been developed during the last decades, and their number is still increasing [1].<br />

In the development of drugs for nasal application, identification and characterization of the<br />

different uptake and efflux systems are necessary. P-glycoprotein (P-gp) and multidrug<br />

resistance-associated proteins (MRP) are classified as ATP binding cassette (ABC)<br />

transporters based on their sequences, organisation of the ATP-binding domains and efflux<br />

function. ABC proteins represent a large family of integral membrane transporters that<br />

utilize the energy of ATP hydrolysis to carry specific substrates across membranes [2]. The<br />

solute carrier gene (SLC) superfamily encodes another large family of membrane-bound<br />

transporters, located in almost every cellular and organelle membranes. Proteins of the<br />

SLC family include passive transporters, ion-coupled transporters and exchangers [3].<br />

A comparison was performed between excised human nasal mucosa from turbinectomy<br />

surgeries and our in vitro model based on immortalized human nasal epithelial cells (RPMI<br />

2650) concerning the mRNA expression of different efflux and uptake transporter proteins.<br />

First investigations to prove functionality of different ABC transporters in RPMI 2650 cells<br />

was carried out using permeation studies and uptake assays.<br />

The mRNA expression of eight ABC transporters as well as 13 SLC transporters of excised<br />

human tissue and RPMI 2650 epithelial model were investigated and compared. The<br />

results showed promising similarity in the possible presence or absence of these efflux and<br />

influx transporters in human tissue and the in vitro model. A similarity between RPMI 2650<br />

models and human nasal mucosa has already been shown for barrier properties to<br />

evaluate passive drug permeation [4].<br />

To verify the functionality of the first transporter protein, P-gp, bidirectional transport studies<br />

using rhodamine 123 with RPMI 2650 cells grown on polyethylene terephthalate filter were<br />

performed. Indirect immunofluorescence staining for P-gp was also carried out. The<br />

findings confirmed the assumption that P-gp is not functionally expressed in these epithelial<br />

cells.<br />

An uptake assay using 5(6)-carboxy-2′,7′-dichlorofluorescein as a substrate for MRP1 and<br />

MRP5 was carried out. The result indicated functional expression of these efflux<br />

transporters in RPMI 2650 cells. Identification and localization of the responsible<br />

transporter proteins have to be proved by immunohistochemistry.<br />

Further investigations will be focused on western blot, immunohistochemical analysis and<br />

permeation as well as uptake studies using specific substrates for active transport to verify<br />

the protein expression and functionality of further drug transporters in human nasal mucosa<br />

and in vitro model based on RPMI 2650 cell line.<br />

Acknowledgments: We are grateful to the German Federal Institute for Risk Assessment (BfR), which<br />

funded this work under grant no. 3-1329-469. Furthermore, the authors thank Prof. Dr. Schroeder, Dr.<br />

Schmidt, Städtisches Klinikum Braunschweig and Dr. Reintjes, HNO-Praxis Schlosscarree Braunschweig for<br />

the supply of human nasal mucosa specimens.<br />

References:<br />

1. Dimova, S. et al.: Toxicology In Vitro 2005, 19: 107–122.<br />

2. Jones, P.M., George, A.M.: Cell. Mol. Life Sci. 2004, 61: 682–699.<br />

3. He, L., Vasiliou, K., Nebert, D.W.: Human Genomics 2009, 3: 195–206.<br />

4. Wengst, A., Reichl, S.: Eur. J. Pharm. Biopharm. 2010, 74: 290–297.<br />

096<br />

A new heterogeneous assay for measuring NAD + -dependent histone<br />

deacetylase (sirtuin) activity<br />

Schiedel, M. 1; Jung, M. 1 2<br />

1 Institute of Pharmaceutical Science, Albert-Ludwigs-<strong>Universität</strong>, Albertstr. 25, 79104 Freiburg,<br />

2 Freiburg Institute of Advanced Studies (FRIAS)<br />

Background<br />

Sirtuins represent a specific NAD +-dependent class of histone deacetylases (HDACs). By<br />

using NAD + as a cofactor, these enzymes cleave off the acetyl groups from the epsilonamino<br />

group of lysines in histones and other proteins, e.g. p53, FOXO proteins, p300 or<br />

HIV tat. The human family of sirtuins consists of seven members, which are distributed to<br />

different cell compartments and involved in the regulation of various physiological<br />

processes like apoptosis, cell differentiation, metabolism, DNA recombination and HIV tat<br />

transactivation. Thus, sirtuin inhibitors are interesting potential drugs for drug discovery [1].<br />

Inhibitor search is highly dependent on high throughput screening (HTS) which relies on<br />

suitable assay systems. We introduced a new heterogeneous assay based on fluorescence<br />

detection of Ac-p53-TAMRA, a known nonhistone substrate for sirtuins [2].<br />

Material and Methods<br />

Ac-p53-TAMRA is a fluorescence-labelled and biotinylated fragment of the<br />

tumorsuppressor p53. Because the immobilized Ac-p53-TAMRA, which is bound on<br />

streptavidin microplates, is not able to serve as Sirt-substrate, we set up a preincubation in<br />

1 mL Epi-tubes. After three hours at 37 °C the reaction is stopped and trypsin is added,<br />

which hydrolyzes only the deacetylated substrate. In the next step the test samples are<br />

dispensed on a streptavidin-coated microplate in order to immobilize the biotinylated<br />

substrate and to wash away the unbound components, including the fluorescence-tagged<br />

cleavage product of tryptic digestion. Finally the amount of remaining uncleaved but still<br />

biotinylated and TAMRA-labelled substrate is determined by fluorescence readout.<br />

Results<br />

We were able to establish a new nonisotopic, heterogeneous assay for NAD +-dependent<br />

histone deacetylases, which enables in-vitro-testing of library components with intrinsic<br />

fluorescence or quenching properties, that are prone to false-negative or –positive results in<br />

homogeneous fluorescence based assay systems. The presented detection protocol is<br />

suitable for HTS.<br />

References:<br />

1. Trapp, J., Jung, M.: Curr Drug Targets 2006, 7(11): 1553-1560.<br />

2. Milne, J.C. et al.: Nature 2007, 450(7170): 712-716.<br />

097<br />

Influence of different solvents on the applicability of a small intestinal<br />

microdialysis system in vivo<br />

Schönherr, D. 1; Hanke, U. 1; Siegmund, W. 2; Becker, D. 3; Weitschies, W. 1<br />

1University of <strong>Greifswald</strong>, Center of Drug Absorption and Transport, Institute of Pharmacy, Felix-<br />

Hausdorff-Straße 3, 17487 <strong>Greifswald</strong>, Germany<br />

2University of <strong>Greifswald</strong>, Center of Drug Absorption and Transport, Institute of Pharmacology,<br />

Felix-Hausdorff-Straße 3, 17487 <strong>Greifswald</strong>, Germany<br />

3Novartis Pharma AG, Technical Research & Development, Novartis Campus, WSJ-177.4.14,<br />

4056 Basel, Switzerland<br />

For better understanding the mechanisms of drug absorption after oral drug administration<br />

a new small intestinal microdialysis system was developed. To characterize the influence of<br />

different solvents on the applicability for in vivo studiesseveral experiments were<br />

performedin vitro.<br />

The small intestinal microdialysis system is made up of a minimodule which is built up of 30<br />

hollow fiber capillaries which consist of a polysulfone membrane (Fresenius Polysulfone ®)<br />

and a tubing system with an inflow, an outflow, and a tube for separating samples via air<br />

bubbles. The applicability of the system was tested with paracetamol (acetaminophen) as<br />

model substance using phosphate buffer USP pH 6.8, hydrochloric acid pH 1.2 and a<br />

biorelevant buffer pH 6.8 as solvent.For all experiments phosphate buffer USP pH 6.8<br />

served as perfusion medium as well. Two pumps were used to transport the samples<br />

(300 µL) after an intended equilibration time and to produce air bubbles separating the<br />

samples in the outflow tube. To examine the sensitivity of the system, the minimodule was<br />

inserted into a chamber filled with solvent. After pre-incubation, a solution of the drug<br />

substance was added (resulting theoretical concentration of 0.02 and 2.0 g/L).<br />

Subsequently, solvent was added to dilute the drug solution (resulting theoretical<br />

concentration of 0.01 and 1.0 g/L). Drug concentrations were calculated using UV<br />

spectroscopy and a previously determined minimodule scaling factor.<br />

Drug concentrations as well as concentration changes over time were successfully<br />

monitored for both drug concentrations and all investigated solvents. The functional<br />

applicability of the microdialysis system at both pH values was proven.<br />

The suitability of the new small intestinal microdialysis system to monitor concentration<br />

changes over time in vitro was demonstrated for different solvents. The applicability of the<br />

system in vivo will be examined in the near future.<br />

116 Poster


We thank Fresenius Medical Care GmbH for providing the hollow fiber capillaries. Financial support from<br />

Novartis Pharma AG is gratefully acknowledged.<br />

098<br />

Identification of benzimidazole derivatives as inhibitors of leukotriene<br />

biosynthesis by virtual screening targeting 5-lipoxygenase-activating protein<br />

(FLAP)<br />

Luderer, S. 1, Banoglu, E. 2, Altenhofen, W. 3, Gerstmeier, J. 4, Pergola, C. 4, Werz, O. 4<br />

1 Department of Pharmaceutical Analytics, Pharmaceutical Institute, Eberhard-Karls-University<br />

Tuebingen, Auf der Morgenstelle 8, D-72076 Tuebingen, Germany<br />

2 Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Gazi University, Etiler, 06330<br />

Yenimahalle, Ankara, Turkey<br />

3 Chemical Computing Group, AG, Kaiser-Wilhelm-Ring 11, 50676 Koeln, Germany<br />

4 Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University<br />

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

Leukotrienes (LTs) are lipid mediators formed from arachidonic acid (AA) by the enzyme 5lipoxygenase<br />

(5-LO). 5-LO converts AA from membrane phospholipids to LTA4, aided by<br />

the 5-lipoxygenase-activating protein (FLAP). LTA4 is further metabolized to LTB4 and<br />

cysteinyl-LTs (cys-LTs) by LTA4 hydrolase or LTC4 synthase [1]. LTs are key mediators in<br />

inflammatory, allergic and cardiovascular diseases, but only Zileuton as direct 5-LO<br />

inhibitor (Zyflo CR®) and Cys-LT receptor antagonists (e.g. Montelukast, Singulair ©) are<br />

currently available on the market and used for the treatment of asthma.<br />

One promising target for pharmacological suppression of leukotriene biosynthesis is 5lipoxygenase-activating<br />

protein (FLAP). Here, we applied a virtual screening targeting<br />

FLAP based on a combined ligand- and structure-based pharmacophore model which led<br />

to the identification of 8 compounds (see below) as potential FLAP inhibitors [2].<br />

Compounds 1, 2 and 3 barely inhibited 5-LO product formation with IC50 values > 10 µM<br />

both in a cell-based and a cell-free system. Compounds 4, 5, 6 and 8 reduced 5-LO product<br />

synthesis in intact neutrophils, but also inhibited recombinant 5-LO suggesting a direct<br />

interference with 5-LO. In contrast, the benzimidazole derivative 7 potently suppressed LT<br />

formation in intact neutrophils (IC50=0.31μM) but essentially failed to inhibit 5-LO in cell-free<br />

systems suggesting an interaction with FLAP. Compound 7 was also active in a human<br />

whole blood assay, and acted anti-inflammatory in an in vivo model of acute inflammation,<br />

i.e. the carrageenan-induced pleurisy in rats. For structural optimization, a series of further<br />

benzimidazole-based derivatives were synthesized leading to more potent analogues with<br />

IC50 values between 0.12-0.19 μM. Our results disclose the benzimidazole scaffold bearing<br />

as a new chemotype for further development of anti-leukotriene agents.<br />

References:<br />

1. Radmark, O., et al., 5-Lipoxygenase: regulation of expression and enzyme activity. Trends Biochem Sci,<br />

2007. 32(7): p. 332-41.<br />

2. Banoglu, E., et al., Identification of novel benzimidazole derivatives as nhibitors of leukotriene<br />

biosynthesis by virtual screening targeting 5-lipoxygenase-activating protein (FLAP). Bioorg Med Chem,<br />

<strong>2012</strong>. 20(12): p. 3728-41.<br />

099<br />

The fourth molybdenum enzyme mARC: Expression and N-reductive activity in<br />

different cell lines<br />

Jakobs H. 1; Reichmann D. 2; Bittner F. 2; Mendel R. 2; Clement B. 1;Havemeyer A. 1<br />

1Department of Pharmaceutical / Medicinal Chemistry, Pharmaceutical Institute, Christian-<br />

Albrechts-University of Kiel, Gutenbergstraße 76, 24118 Kiel, Germany<br />

2Department of Plant Biology, Technical University of Braunschweig, Humboldtstraße 1, 38106<br />

Braunschweig, Germany<br />

mARC (“mitochondrial amidoxime reducing component”) is a mammalian molybdenum<br />

enzyme that was discovered in our lab [1]. The enzyme is located in the outer mitochondrial<br />

membrane and together with the electron transport proteins cytochrome b5 and its NADHdependent<br />

reductase it is capable of reducing N-hydroxylated compounds such as the<br />

model substrate benzamidoxime [2,3].This is actually where the enzyme got its name from.<br />

Besides amidoximes, the enzyme system is also able to reduce other N-hydroxylated<br />

structures such as guanidines and sulphonamides and is thus involved in reductive drug<br />

metabolism[2]. For this reason it can be employed for the activation of<br />

N-hydroxylatedprodrugs using e.g. the prodrug principle of “amidoximes instead of<br />

amidines” [4]. Moreover, the mARC containing enzyme system is also able to reduce<br />

N-hydroxylated DNA-bases[2], which could be a mechanism of detoxification.However, the<br />

endogenous function of mARC is still unknown. The human genome codes for two mARC<br />

proteins, referred to as mARC1 and mARC2 [5].<br />

Our studies upon tissue distribution carried out with porcine material revealed highest<br />

expression levels for mARC in liver, thyroid and kidney [6].<br />

In continuation to our studies with primary hepatocytes, porcine hepatic and extrahepatic<br />

tissue fractions and recombinant human enzymes, we tested extrahepatic cell lines both on<br />

mARC1 and mARC2 expression and N-reductive activities. Cell lines included kidney cell<br />

lines HEK293, RC124, HMCL and Colo357, a pancreatic cancer cell line. Furthermore, we<br />

studied 3T3-L1, a fibroblast-like murine cell line that differentiates under appropriate<br />

conditions into adipocyte-like cells.<br />

N-reductive activity and expression of both mARC1 and mARC2 can be found in all studied<br />

cell lines. N-reduction in all human cell lines obey Michaelis-Menten-kinetics, but show<br />

differences in Vmax and Km. Conversion rates for benzamidoxime correlate with the<br />

expression of mARC1 but not with the expression of mARC2.<br />

All studied human cell lines can therefore be used to study both the enzyme system in<br />

general and the N-reduction of N-hydroxylated compounds.<br />

With regard to murine 3T3-L1,N-reductive activity is enhanced during differentiation into an<br />

adipocyte-like phenotype.<br />

Others found that enzymes and transporters that take part in the TCA cycle, fatty acid<br />

oxidation and ATP synthesis as well as mARC2 show changes in expression during<br />

differentiation of 3T3-L1 into an adipocyte-like cell type [7].Together with the fact that<br />

N-reductive activityis increased during differentiation of 3T3 L1 this could be a hint that<br />

mARC is involved in metabolic processes. Further studies on this topic will be necessary.<br />

References:<br />

1. Havemeyer, A. et al.:J. Bio. Chem.2006, 281(46): 34796–34802.<br />

2. Gruenewald, S. et al.: J. Med. Chem.2008, 51(24): 8173–8177.<br />

3. Neve, E. et al.: J. Biol. Chem.2011, 287(9): 6307–6317.<br />

4. Clement B.:Inventor. Methoden zur Behandlung und Prophylaxe der Pneumocystiscariniipneumonie<br />

(PCP) und anderen Erkrankungen sow e Verbindungen und Formulierungen zum Gebrauch bei besagten<br />

Methoden. 1993, German patent P4321444.4, PCT/DE 94/00756 (1994); U.S. patent 5,786,383 (1998 July<br />

28); European patent 0708640 (1998 September 16)<br />

5. Wahl, B.et al.: J. Biol. Chem.2011, 285(48): 37847–37859.<br />

6. Clement, B. et al.: Drug Metab. Rev.2010,42(S1): 1–323.<br />

7. Newton, B. et al.:J. Proteome Res. 2011,10(10): 4692–4702.<br />

100<br />

QSAR studies of the N-reduction of p-substituted benzamidoximes<br />

Treuer, E. 1; Reichmann, D. 2; Bittner, F. 2; Mendel, R. 2; Clement, B. 1;Havemeyer, A. 1<br />

1Pharmaceutical Institute, Christian-Albrechts-University, Gutenbergstraße 76, 24118 Kiel,<br />

Germany<br />

2 Department of Plant Biology, Technical University of Braunschweig, Humboldtstraße 1, 38106<br />

Braunschweig, Germany<br />

mARC (mitochondrial Amidoxime Reducing Component) is a molybdenum containing<br />

protein, which is localized in the outer mitochondrial membrane [1]. The genome of<br />

mammalians codes for two different forms of this protein (mARC 1 and mARC 2). Together<br />

with the electron transport proteins cytochrome b5 and cytochrome b5reductase, it catalyzes<br />

the N-reduction of different compounds. On the one hand, it transforms toxic and mutagenic<br />

N-hydroxylated base analogues [2]. On the other hand, it is involved in the activation of<br />

several prodrugs like N-hydroxypentamidine and guanoxabenz [3].<br />

For the development of new amidoximeprodrugs, we investigate structure-activity<br />

relationships of the N-reduction of p-substituted benzamidoximes. Therefore, several<br />

compounds were incubated with OMV (outer membrane vesicles) which was isolated from<br />

pig liver mitochondria. The resulting amidine was quantified with HPLC analysis and Km<br />

and Vmax were calculated. The influence of the electronic properties of the substituent in<br />

para-position on the N-reductive activity was measured. There are first hints that the<br />

conversion rates for the N-reduction of benzamidoximes with electron donating groups in<br />

para-position are higher than for compounds with electron withdrawing substituents.<br />

However, the differences are low and all para-substituted benzamidoximes were intensively<br />

reduced. Thus the prodrug principle amidoximes (N-hydroxyamidines) instead of amidines<br />

can be applied to allbenzamidine containing drug candidates irrespective of the<br />

substituents at the aromatic ring.<br />

References:<br />

1. Havemeyer, A. et al.: J. Biol. Chem.2006, 281(46): 34796-34802.<br />

2. Havemeyer, A., Lang, J., and Clement, B.: Drug.Metab.Rev. 2001, 43(4): 524-539.<br />

3. Gruenewald, S. et al.: J. Med.Chem. 2008, 51(24): 8173-8177<br />

101<br />

Development of an ELISA assay for screening of p38δ MAPK inhibitors<br />

Goettert M. a ,Bauer S. a , Shaalan N. b , Graeser, R. c , Laufer, S. a<br />

a Department of Pharmaceutcal and Medicinal Chemistry, Institute of Pharmacy, Eberhard Karls<br />

University of Tübingen, Auf der Morgenstelle 8, D-72076 Tübingen, Germany<br />

b Department of Pharmaceutical Biology, Faculty of Pharmacy and Biotechnology, German<br />

University of Cairo, Egypt<br />

c ProQinase GmbH, Breisacher Str. 117, D-79106 Freiburg, Germany<br />

p38 mitogen activated protein kinases (MAPKs) are members of a larger group of serine/<br />

threonine protein kinases.<br />

They contribute in a variety of cellular processes such as gene expression, mitosis,<br />

differentiation, cell survival/apoptosis and biosynthesis/ release of pro- inflammatory<br />

cytokines [1].<br />

In many inflammatory diseases, e.g. rheumatoid arthritis (RA), the role of p38α isoform is<br />

widely investigated.<br />

Activated rheumatoid arthritis synovial fibroblasts (RASFs) are considered as key cells in<br />

the development of RA since they mediate the most relevant pathways of joint destruction<br />

n[2,3].<br />

The p38δ MAPK is activated in RASFs by a cytokine-independent pathway, leading to the<br />

expression of matrix metalloproteinases, e.g. MMP-1 and MMP-3, which contributes to the<br />

destruction of articular cartilage and bone [3,4].<br />

In the RA synovial tissue, all four isoforms- α, β, γ and δ- of the p38 MAPK have been<br />

detected. Particularly, p38δ MAPK occurs predominantly in the site of invasion and bone<br />

destruction. For this, the participation of p38δ MAPK becomes more and more evident<br />

[2,5].<br />

For the purpose of routine screening, we developed a direct 96 Well plate ELISA assay to<br />

identify inhibitors of p38δ MAPK. After incubation with a candidate inhibitor, the activity of<br />

p38δ MAPK is measured by the phosphorylation degree of activation transcription factor 2<br />

(ATF-2). The phosphorylated ATF-2 is directly detected by a monoclonal peroxidase<br />

conjugated antibody. Being a natural substrate of the p38 MAPKs, ATF- 2 phosphorylation<br />

is inversely correlated with the inhibitor potency.<br />

Poster 117


Based on already successfully established ELISA assays for p38α MAPK and JNK3, the<br />

advantages of this assay are its accuracy, easy handling, rapidness and the avoidance of<br />

using radioisotopes.<br />

References:<br />

1. Schindler, J.F., J.B. Monahan, and W.G. Smith, p38 pathway kinases as anti-inflammatory drug targets.<br />

Journal of Dental Research, 2007. 86(9): p. 800-811.<br />

2. Ospelt, C., et al., Toll-like receptors in rheumatoid arthritis joint destruction mediated by two dist nct<br />

pathways. Annals of the Rheumatic Diseases, 2004. 63: p. 90-91.<br />

3. Huber, L.C., et al., Synovial fibroblasts: key players in rheumatoid arthritis. Rheumatology, 2006. 45(6): p.<br />

669-675.<br />

4. Goettert, M., et al., Development of a p38δ mitogen activated protein kinase ELISA assay for the<br />

quantitative determination of inhibitor activity. Journal of Pharmaceutical and Biomedical Analysis, <strong>2012</strong>.<br />

5. Korb, A., et al., Differential tissue expression and activation of p38 MAPK alpha, beta, gamma, and delta<br />

isoforms n rheumatoid arthritis. Arthritis and Rheumatism, 2006. 54(9): p. 2745-2756.<br />

102<br />

Is Dimethylarginine-Dimethylaminohydrolase-2 (DDAH-2) involved in<br />

methylarginine metabolism?<br />

Altmann K. S. 1, Havemeyer A. 1, Beitz E. 1, Clement B. 1<br />

1Pharmaceutical Institute, Department of Pharmaceutical and Medicinal Chemistry, Christian-<br />

Albrechts-University of Kiel, Gutenbergstraße 76, 24118 Kiel, Germany<br />

DDAH isoenzymes, discovered in 1989 by Ogawa et al., catalyze the hydrolytic degradation<br />

of free endogenous methylarginines, Nω-monomethyl-L-arginine (L-NMMA) and<br />

Nω,Nω'-dimethyl-L-arginine (ADMA). [1,2] These methylarginines occur as endogenous nitric<br />

oxide synthase (NOS) inhibitors and reduce the formation of nitric oxide (NO).[3,4] By<br />

modulating the rates of L-NMMA and ADMA DDAH enzymes exhibit a therapeutic target to<br />

modulate the bioavailability of NO. [5,6]<br />

The postulated metabolism is evidenced for DDAH-1 but the involvement of DDAH-2 in the<br />

degradation of ADMA and L-NMMA is still a matter of debate.<br />

Experimental Procedures:<br />

Therefore, we determined the isoform specific DDAH protein expression profile in various<br />

porcine tissues by immunoblotting and correlated it with respective DDAH activity levels.<br />

Tissue DDAH activity was measured by the conversion of ADMA alone and in the presence<br />

of a DDAH inhibitor. Furthermore, metabolism of ADMA, L-NMMA and alternative DDAH<br />

substrates, S-methyl-L-thiocitrulline (SMTC) andNω-amino-L-arginine, was studied in<br />

porcine tissues each containing one DDAH isoform.<br />

Results:<br />

DDAH activity was detected in homogenates of porcine kidney, liver, and brain, i.e. tissues<br />

with high expression levels of DDAH-1, but not in those of spleen or thyroid, where DDAH-2<br />

predominates. By the same token neither methylarginines nor alternative DDAH substrates<br />

were converted in the DDAH-2-rich porcine thyroid cytosolic fractions, whereas all<br />

substrates were metabolized by the DDAH-1-rich kidney cytosol preparation.<br />

In addition, L-citrulline formation in porcine tissues was reduced to 50 % in the presence of<br />

1 mM of the DDAH inhibitor Nω-(2-methoxyethyl)-L-arginine (L-257), evidencing DDAH<br />

activity as a major source for the conversion. [7]<br />

Conclusion:<br />

Together, these data show that DDAH-1 and DDAH-2 have distinct tissue distributions as<br />

well as substrate selectivity indicating different roles in NO metabolism. While DDAH-1<br />

metabolizes the endogenous NOS inhibitors L-NMMA and ADMA, a physiological function<br />

of DDAH-2 has yet to be defined.<br />

References:<br />

1. Ogawa, T., Kimoto, M., and Sasaoka, K.: J. Biol. Chem. 1989, 264(17):10205–10209.<br />

2. Leiper, J. M. et al.: Biochem. J. 1999, 343Pt 1: 209–214.<br />

3. Anthony, S., Leiper, J., and Vallance, P.: Vasc Med. 2005, 10Suppl 1: S3-9.<br />

4. MacAllister, R. J. et al.: Br. J. Pharmacol. 1996, 119(8): 1533–1540.<br />

5. Dayoub, H. et al.: Circulation. 2003, 108(24): 3042–3047.<br />

6. Feng, M. et al.: Int. J. Cardiol. 2010, 144(2): 180–186.<br />

7. Rossiter, S. et al.: J. Med. Chem. 2005, 48(14): 4670–4678.<br />

103<br />

New heterocyclic sigma receptor ligands and their role in the activation of<br />

apoptotic pathways<br />

Korpis K1, Weber F2, Wünsch B2, Bednarski P J1 1 Institute of Pharmacy, University of <strong>Greifswald</strong>, F.-L.-Jahn-Straße 17, 17487 <strong>Greifswald</strong>, Germany<br />

2 Institute of Pharmacy, University of Münster, Hittorfstraße 58-62, 48149 Münster, Germany<br />

Sigma receptors are present in peripheral organs and in the central nervous system. They<br />

are divided into two distinct subtypes: σ1 and σ2. Both subtypes are involved in<br />

neuromodulatory processes and in particular the σ1 receptor is under evaluation for the<br />

treatment of a number of neurological disorders such as depression or schizophrenia. 1 In<br />

addition to the relevance of these receptors in neurological effects both subtypes are<br />

expressed in a high density in different human tumor cell lines. σ1 receptor antagonists and<br />

σ2 agonists have been shown to be involved in programmed cell death (apoptosis),<br />

whereas σ1 receptor agonists and σ2 receptor antagonists have antiapoptotic and<br />

neuroprotective activity. More specifically, Spruce et al. have shown that σ1 antagonists<br />

induce caspase-dependent apoptosis, which is in accord with recent observations that σ1<br />

agonists prevent caspase-3 activation. On the other hand, σ2 agonists have been shown to<br />

activate a caspase independent apoptotic pathway and have been proposed as putative<br />

anti-cancer drugs. 2<br />

The aim of this work is to determine the role of newly synthesized σ receptor ligands in the<br />

activation of apoptotic pathways.<br />

Flow cytometry with Annexin V and propidium iodide was performed to observe cell<br />

membrane alterations due to apoptosis. Experiments with human cancer cells treated with<br />

our compounds showed apoptosis levels increasing from 1 % in the control to 31 % in the<br />

treated cells. Another apoptosis assay was used to measure the expression of members of<br />

the caspase family to confirm apoptosis as the mechanism of cell death. Treatment of the<br />

multiple myeloma cell line RPMI 8226 caused dose-dependent activation of caspase-3,<br />

caspase-8 and caspase-9, indicating the activation of both the intrinsic and the extrinsic<br />

apoptotic pathway.<br />

However, Western blot analysis showed only cleaved products of caspase-8 and not<br />

caspase-9, evidence for activation of only the extrinsic pathway. These data suggest that<br />

our compounds do not suppress the expression of pro-survival Bcl-2 family proteins, such<br />

as Bcl-2.<br />

Further studies will address the crucial role in the extrinsic apoptotic pathway, which is<br />

initiated by activation of death receptors (i.e. TRAIL) and if there are other apoptosisindependent<br />

pathways of programmed cell death, for example autophagy.<br />

References:<br />

1. Abate, C. et al.: J. Med. Chem. 2011, 54(4): 1022-1032.<br />

2. Riganas, S. et al.: Bioorganic & Medicinal Chemistry <strong>2012</strong>, 20(10): 3323-3331.<br />

104<br />

A cell culture based assay for studies on inhibitors of the inducible nitric oxide<br />

synthase (iNOS) and effects of cold atmospheric pressure plasma treatment of<br />

keratinocytes and lymphocytes<br />

Schulz U1, Bundscherer L2, von Woedtke Th3, Masur K2, Morgenstern O1 1 Institut für Pharmazie, <strong>Ernst</strong>-<strong>Moritz</strong>-<strong>Arndt</strong>-<strong>Universität</strong> <strong>Greifswald</strong>, Friedrich-Ludwig-Jahn-Str. 17,<br />

17489 <strong>Greifswald</strong>, Germany<br />

2 Zentrum für Innovationskompetenz plasmatis, AG “Zelluläre Effekte” im INP <strong>Greifswald</strong> e.V.,<br />

Felix-Hausdorff-Str. 2, 17489 <strong>Greifswald</strong>, Germany<br />

3 Campus PlasmaMed im INP <strong>Greifswald</strong> e.V., Felix-Hausdorff-Str. 2, 17489 <strong>Greifswald</strong>, Germany<br />

The inducible nitric oxide synthase (iNOS) is a target of great interest for development of<br />

drugs for COPD and asthma [1], septic shock [2] and autoimmune diseases e.g. diabetes<br />

type 1 [3] and multiple sclerosis [4]. We synthesized some potential iNOS inhibitors with<br />

general structure 1 based on the fact that substances with thiourea or thiosemicarbazide<br />

structure elements showed iNOS-inhibitory properties before [5].<br />

To investigate the effect of our compounds on iNOS, we established a cell culture based<br />

assay using RINm5F cells that express the target under influence of interleukin-1β (IL-1β)<br />

and interferon-γ (IFN-γ) [6]. We optimized the produced amount of nitric oxide (measured<br />

as nitrite) using various combinations of the two cytokines with two cell counts. Additionally,<br />

we varied the incubation time pre and post addition of IL-1β and IFN-γ.<br />

Here we display the collected data that show that IFN-γ is, as expected, unable to induce<br />

iNOS on its own [7]. IL-1β alone increases nitrite concentration higher in short preincubation.<br />

IFN-γ in addition to IL-1β is able in very little concentrations to push the NO<br />

production to a maximum. We found a combination of low IL-1β and IFN-γ concentrations<br />

after a pre-incubation of 24 h and 48 h incubation after cytokine addition to be most<br />

effective.<br />

With these parameters we screened our derived substances for activity and found 7<br />

compounds to inhibit the target significantly higher than the reference aminoguanidine.<br />

Additionally to the screening of the chemical substances, we used the iNOS test model with<br />

modification to study effects of cold atmospheric pressure plasma treatment of<br />

keratinocytes (HaCaT cells) and lymphocytes (Jurkat cells). We searched for increased<br />

nitrite concentrations due to treatment of RINm5F cells with plasma or with supernatant of<br />

plasma-treated HaCaT and Jurkat cells. The aim was to investigate the influence of the<br />

plasma based secretion of the cytokines IL-1β and IFN-γ in order to induce NO production<br />

from the RINm5F cells. Possible inhibitory effects from produced cytokines e.g. TGF-β were<br />

investigated as well [8].<br />

Acknowledgments: Zentrum für Innovationskompetenz plasmatis, AG “Zelluläre Effekte” im INP <strong>Greifswald</strong><br />

e.V., Felix-Hausdorff-Str. 2, 17489 <strong>Greifswald</strong>, Germany<br />

The authors like to gratefully acknowledge the contribution of Liane Kantz to this work.<br />

References:<br />

1. Sugiura H, Ichinose M: Nitric Oxide 2011, 25(2): 138-144.<br />

2. Fortin C F, McDonald P P, Fulop T, Lasur O: Shock 2010, 33(4): 344-352.<br />

3. Cnop M, Welsh N, Jonas J C, Jörns A, Lenzen S, Eizirik D L: Diabetes 2005, 54(suppl 2): S97-S107.<br />

4. Steinert J R, Chernova T, Forsythe I D: Neuroscientist 2010, 16(4): 435-452.<br />

5. Al-Amiery A A, Al-Majedy Y K, Ibrahim H H, Al-Tamimi A A: Org. Med. Chem. Lett. <strong>2012</strong>, 2(1): 4-7.<br />

6. Ankarcrona M, Dypbukt J M, Brune B, Nicotera P: Exp. Cell Res. 1994, 213(1): 172-177.<br />

7. Cetkovic-Cvrlje M, Eizirik D L: Cytokine 1994, 6(4): 399-406.<br />

8. Mabley J G, Cunningham J M, John N, Di Matteo M A, Green I C: J. Endocrinol. 1997, 155(3): 567-575.<br />

105<br />

New Glutathione Peroxidase Inhibitors<br />

Lemmerhirt, H. 1; Schulz, R. 1; Emmrich, T. 1; Wilde, F. 1; Link, A. 1; Bednarski, P. J. 1<br />

1<strong>Ernst</strong>-<strong>Moritz</strong>-<strong>Arndt</strong>-University, Institute of Pharmacy, Friedrich-Ludwig-Jahn-Straße 17, 17489<br />

<strong>Greifswald</strong>, Germany.<br />

Cancer cells have a variety of mechanisms to protect themselves from anti-cancer drugs.<br />

For example, they express drug efflux transporters like P-gp, increase repair of DNA<br />

damage or mutate targeted enzymes and receptors. A relatively new postulated<br />

mechanism of resistance is the over-expression of enzymes that deal with oxidative stress.<br />

It has been postulated that glutathione peroxidases (GPx, EC 1.11.1.9) is a key enzyme in<br />

the development of resistance. [1] The primary function of GPx is to prevent cells from<br />

oxidative stress by degrading peroxides like hydrogen peroxide to water or organic<br />

hydroperoxides to the corresponding alcohols.<br />

Hydrogen peroxide is postulated to be an important messenger of apoptosis. If cancer cells<br />

treated with anti-cancer drugs were to over-express GPx, then the signals for apoptosis<br />

would be weakened, leading to survival of more cancer cells [2].<br />

The aim of our work is to develop specific and reversible inhibitors of GPx to reverse drug<br />

resistance. Already some inhibitors of GPx are known, such as misonidazole and several<br />

118 Poster


thiol compounds but they have disadvantages; misonidazole shows neurotoxicity and thiols<br />

are unspecific and inhibit GPx irreversible. Furthermore thiols are instable with regard to<br />

oxidation.<br />

R. SCHULZ identified several promising lead structures by means of virtual screening with<br />

AutoDock and determined their GPx inhibitory activity by an enzyme assay [3]. One of them<br />

(1) was chosen for structural modification to improve inhibitory activity.<br />

The synthesized compounds have been tested for enzyme inhibitory activity in a microtiter<br />

based assay. The method is based on the oxidation of glutathione by GPx in the presence<br />

of hydroperoxides. The resulting glutathione disulfide (GSSG) is reduced back to GSH by<br />

glutathione reductase (GR) with consumption of NADPH. The rate of decrease of NADPH,<br />

monitored at λ = 340 nm, corresponds to the catalytic rate of GPx.<br />

We have now identified two new compounds that inhibit GPx with lower IC50 values than the<br />

lead compound. The substances will now be tested in vitro cell culture on human cancer<br />

cells resistant to common anti-cancer drugs. For example, ovarian carcinoma cell lines<br />

resistant to cisplatin will be used to assess whether anti-cancer-drug-resistance can be<br />

reversed with the new compounds.<br />

References:<br />

1. Saga, Y., et al.: Oncol Rep 2008, 20 (6): 1299-303.<br />

2. Gouaze, V., et al.: J Biol Chem 2002, 277 (45): 42867-74.<br />

3. Schulz, R.: Diploma Thesis 2008.<br />

106<br />

Stimulation and Inhibition of Angiogenesis by Non-Thermal Atmospheric-<br />

Pressure Plasma<br />

Haertel B. 1; Eiden K. 1; Deuter A. 1; Wende K. 1; von Woedtke T. 2; Lindequist U. 1<br />

1 Institute of Pharmacy, University of <strong>Greifswald</strong>, D-17489 <strong>Greifswald</strong>, Germany<br />

2 Leibniz-Institute for Plasma Science and Technology e.V. (INP), D-17489 <strong>Greifswald</strong>, Germany<br />

The study of angiogenesis has broad clinical implications e.g. in the fields of wound<br />

healing, oncology, hematology or ophthalmology. The formation of new blood vessels is an<br />

essential feature of tissue remodeling. Further, angiogenesis is a prerequisite for growth of<br />

solid tumors and development of metastasis. However, it is also involved in the<br />

pathophysiology of hematologic malignancies. For improving wound healing angiogenesis<br />

should be promoted, whereas in treating tumors angiogenesis should be inhibited. Inhibition<br />

of angiogenesis is also important in the treatment of macular edema or age-related macular<br />

degeneration. Angiogenesis depends on the expression of mediators initiating events<br />

leading to the formation of new microvessels. Non-thermal plasma induces a variety of<br />

effects on living cells in vitro, including production of fibroblast growth factor-2 (FGF-2) or<br />

reactive oxygen species by endothelial cells. Both FGF-2 and ROS belong to mediators of<br />

angiogenesis. However, up to now there are no quantitative data using non-thermal plasma<br />

in more complex angiogenesis models as the hen’s egg test on the chorioallantoic<br />

membrane (HET-CAM) or the aortic ring test (AOR). It is very important to know whether<br />

and how plasma influences angiogenesis in complex models.<br />

This study focused on the effects of plasma generated by the kINPen 09 [1] on<br />

angiogenesis using two different models: HET-CAM and rat aortic ring (AOR) test. In both<br />

models kINPen 09 treated medium (30 to 300s) was applied. ImageJ was used to analyze<br />

vessel area and fractal dimension after treating the CAM from embryonic day 11 to 13.<br />

Aortic rings were prepared from either LEW.1W or WOK.W rats. They were embedded in<br />

matrigel and treated daily for 4 days starting at day 4 after embedding. We developed a<br />

semi quantitative method to quantify outgrow of microvessels from aortic rings.<br />

In both models natural and spontaneous vessel formation was detected. In the HET-CAM<br />

assay vessel area and fractal dimension were significantly enhanced at embryonic day 14<br />

by the 120s-plasma treated medium compared to untreated controls. There was no effect of<br />

plasma (60 or 120s) on vessel growth of aortic rings prepared from LEW.1W rats. The<br />

angiogenic activity of rings from WOK.W rats was significantly (p


to its regulatory C2-like domain (C2ld). Though the crystal structure of 5-LO has been<br />

resolved, many of the distinct interaction sites and the involvement of the C2ld are still<br />

unclear. Therefore the C2ld (amino acids 1-115, fused to maltose binding protein) was<br />

expressed and purified in two steps via amylose affinity chromatography and, after<br />

cleavage, nickel chelating affinity chromatography. Gel filtration studies revealed a high<br />

tendency of the isolated domain to form soluble aggregates. But the single mutation of<br />

tryptophan 75 to alanine led to a dramatic improvement of its biophysical properties leading<br />

to mainly monomeric protein. The proper folding of the tryptophan mutant was successfully<br />

shown by its ability to bind Ca 2+ in a thermal shift assay. Crosslinking of 5-LO or the<br />

isolated C2ld with CLP revealed that the catalytic domain is also involved in the binding of<br />

CLP.<br />

Here we present the purification of a single-mutant that renders the 5-LO C2ld accessible<br />

for interaction studies elucidating the involvement of the regulatory domain to the binding of<br />

small molecules or proteins.<br />

References:<br />

1. Hammarberg T. et al. J Biol Chem. 2000, 275(49): 38787-93.<br />

2. Radmark O. et al. Trends Biochem Sci. 2007, 32(7): 332-41.<br />

3. Rakonjac M. et al. Proc Natl Acad Sci U S A. 2006, 103(35): 13150-5.<br />

4. Dincbas-Renqvist V. et al. Biochim Biophys Acta. 2009, 1789(2): 99-108.<br />

110<br />

Synthesis of annelated and substituted imidazolones and –thiones targeted at<br />

human NAD + -dependent histone deacetylases<br />

Beese K1; El Gaghlab K1; Rumpf T2; Jung M2; Link A1 1Institute of Pharmacy, <strong>Ernst</strong>-<strong>Moritz</strong>-<strong>Arndt</strong>-University <strong>Greifswald</strong>, Friedrich-Ludwig-Jahn-Str. 17,<br />

17489 <strong>Greifswald</strong>, Germany<br />

2Institute of Pharmaceutical Sciences, Albert-Ludwigs-University Freiburg, Albertstr. 25, 79104<br />

Freiburg, Germany<br />

Histones can be modified enzymatically by insertion and elimination of chemical groups<br />

mainly upon basic amino acids. A very significant histone modification is the acetylation<br />

forming lightly packed, active euchromatin that facilitates the accessibility of genes for<br />

transcription factors, whereas deacetylation leads to tightly packed, inactive<br />

heterochromatin, which results in a repression of transcription.<br />

NAD +-dependent histone deacetylases (HDACs), known as sirtuins, regulate epigenetic<br />

gene expression by deacetylation of histones and non-histone proteins. Thus, an increased<br />

enzyme activity is associated with the development of cancer, HIV and neurodegenerative<br />

diseases, and inhibitors of HDACs might represent promising drugs.<br />

In continuation of our efforts to identify modulators of HDACs with improved activity and<br />

selectivity we designed and synthesized analogs of a novel lead structure [1] reported by<br />

our group, recently, depicted below. Compounds such as 1-substituted1,3dihydroimidazo[4,5-c]quinolin-2-ones/thiones,1-substituted5-nitro-1,3-dihydronaphth[1,2d]imidazol-2-ones/thiones,<br />

and 1,5-disubstituted1,3-dihydrobenzimidazol-2-ones/thiones,<br />

that can be regarded as non-hydrolyzable analogs of the lactone splitomicin, were prepared<br />

and tested in a new nonisotopic, heterogeneous assay.<br />

introduction<br />

of<br />

hetero atoms<br />

or<br />

substituents<br />

annelation<br />

and /or<br />

substitution<br />

replacement<br />

by<br />

oxygen<br />

introduction<br />

of<br />

aliphatic or aromatic<br />

sidechains<br />

Acknowledgements: Technical assistance of Dr. A. Bodtke and K. Böheim is gratefully acknowledged.<br />

K.E.G. was recipient of grant DFG Li 765/4-2.<br />

References:<br />

1. Freitag, M. et al.: Bioorg Med Chem 2011, 19(12): 3669–3677.<br />

111<br />

ICAM-1 confers the TIMP-1-dependent anti-invasive action of cannabinoids on<br />

human lung cancer cells<br />

Ramer, R. 1; Bublitz, K. 1; Merkord, J. 2; Haustein, M. 1; Borchert, P. 1; Linnebacher, M. 2; Hinz,<br />

B. 1<br />

1 Institute of Toxicology and Pharmacology, University of Rostock, Schillingallee 70, D-18057<br />

Rostock, Germany<br />

2 Section of Molecular Oncology and Immunotherapy, Department of General Surgery, University<br />

of Rostock, Schillingallee 69, D-18057 Rostock, Germany<br />

Cannabinoids inhibit cancer cell invasion via increasing tissue inhibitor of matrix<br />

metalloproteinases-1 (TIMP-1). This study investigates the role of intercellular adhesion<br />

molecule-1 (ICAM-1) within this action. In the lung cancer cell lines A549, H358, and H460,<br />

cannabidiol (CBD; 0.001-3 μM) elicited concentration-dependent ICAM-1 up-regulation<br />

compared to vehicle via cannabinoid receptors, transient receptor potential vanilloid 1, and<br />

p42/44 mitogen-activated protein kinase. Up-regulation of ICAM-1 mRNA by CBD in A549<br />

was 4-fold at 3 μM, with significant effects already evident at 0.01 μM. ICAM-1 induction<br />

became significant after 2 h, whereas significant TIMP-1 mRNA increases were observed<br />

only after 48 h. Inhibition of ICAM-1 by antibody or siRNA approaches reversed the antiinvasive<br />

and TIMP-1-upregulating action of CBD and the likewise ICAM-1-inducing<br />

cannabinoids Δ 9-tetrahydrocannabinol and r(+)-methanandamide when compared to<br />

isotype or nonsilencing siRNA controls. ICAM-1-dependent anti-invasive cannabinoid<br />

effects were confirmed in primary tumor cells from a lung cancer patient. In athymic nude<br />

mice, CBD elicited a 2.6- and 3.0-fold increase of ICAM-1 and TIMP-1 protein in A549<br />

xenografts, as compared to vehicle-treated animals, and an antimetastatic effect that was<br />

fully reversed by a neutralizing antibody against ICAM-1 [% metastatic lung nodules vs.<br />

isotype control (100%): 47.7% for CBD + isotype antibody and 106.6% for CBD + ICAM-1<br />

antibody]. Overall, our data indicate that cannabinoids induce ICAM-1, thereby conferring<br />

TIMP-1 induction and subsequent decreased cancer cell invasiveness.<br />

112<br />

Can Heparin influence the Cyr61 pathway for affecting cell adhesion<br />

processes?<br />

Schmitz, P. 1; Gerber, U. 1; Schütze, N. 2; Bendas, G. 1<br />

1 Department of Pharmacy, University Bonn, 53121 Bonn, Germany<br />

2 Orthopedic Center for Musculoskeletal Research, University Würzburg, 97074 Würzburg,<br />

Germany<br />

The cysteine-rich protein 61 (Cyr61, CCN1), a member of the CCN-family, is a matricellular<br />

protein expressed and secreted by various cells, which induce e.g. angiogenetic effects and<br />

enhance the tumorigenicity of solid tumors. Besides a range of functions Cyr61 mediates<br />

adhesion and migration of tumor cells by activating different members of the integrin<br />

adhesion receptor family. Integrins are crucial for different steps in course of the<br />

haematogenous metastasis. Heparin possesses antimetastatic effects by blocking<br />

adhesion receptors; selectins and selected integrins, e.g.VLA-4 (very late activation antigen<br />

4) [1,2], but heparin should also be able to bind Cyr61.<br />

However, neither the molecular mechanisms of Cyr61 affecting VLA-4 mediated adhesion<br />

nor interference by heparin in this pathway have been described. This study focused these<br />

aspects.<br />

To confirm direct binding of heparin to Cyr61, SAW biosensor studies were performed<br />

using a series of modified heparins and recombinant CCN1 proteins. To further focus on<br />

the Cyr61/VLA-4 binding pathway and potential impact on that by heparin, the impact of a<br />

Cyr61 downregulation in MV3 melanoma cells by shRNA technology on VLA-4 mediated<br />

cell adhesion was analysed by microscopic detection.<br />

Our data suggest a reduced VLA-4 mediated cell binding of Cyr61 knockdown MV3 clones<br />

in an adhesion assay, while exogenous recombinant Cyr61 restores the binding<br />

capacities.This confirms that VLA-4 is partly controlled by Cyr61 activity and make Cyr61<br />

as attractive target to interfere with metastasis. Concerning heparin interference, kinetic<br />

binding data suggest a direct interaction between heparin and Cyr61 for the first time.<br />

Binding affinities of fractionated heparin in the micromolar range were intensified by Nacetylation<br />

or 2-O-desulfation of heparin. Furthermore, Cyr61 accumulation at the cell<br />

surface glycosaminoglycan Syndecan-4 could be simulated demonstrating high binding<br />

affinities. However, this binding is non-reversible by adding high concentrations of heparin.<br />

Our data provide evidence for a certain impact of Cyr61 with VLA-4 mediated melanoma<br />

cell binding. The hypothesis of heparin interference in this pathway is strongly supported<br />

providing prospects for therapeutic interference in tumorigenicity and metastasis by<br />

blocking Cyr61 activity.<br />

We gratefully thank the “G.Ronzoni“ Institute, Milano for providing us the modified heparins.<br />

References:<br />

1. Schlesinger, M., et al., Thromb Res. 5 (<strong>2012</strong>): 603-10.<br />

2. Bendas, G., Borsig, L.; Int J Cell Biol. <strong>2012</strong>.<br />

113<br />

Cyclooxygenase-2 and peroxisome proliferator activated receptor gamma<br />

confer cannabidiol-induced apoptosis of human lung cancer cells<br />

Heinemann, K. 1; Ramer, R. 1; Merkord, J. 2; Salamon, A. 2; Linnebacher, M. 3; Hinz, B. 1<br />

1 Institute of Toxicology and Pharmacology, University of Rostock, Schillingallee 70, D-18057<br />

Rostock, Germany<br />

2 Department of Cell Biology, University of Rostock, Schillingallee 69, D-18057 Rostock, Germany<br />

3 Section of Molecular Oncology and Immunotherapy, Department of General Surgery, University of<br />

Rostock, Schillingallee 69, D-18057 Rostock, Germany<br />

The antitumorigenic mechanism of cannabidiol (CBD) is still controversial. This study<br />

investigates the role of cyclooxygenase-2 (COX-2) and peroxisome proliferator activated<br />

receptor γ (PPARγ) in CBD´s proapoptotic and tumor-regressive action. In lung cancer cell<br />

lines (A549, H460) and primary cells from a lung cancer patient CBD elicited decreased<br />

viability associated with apoptosis with the strongest toxic impact on primary tumor cells.<br />

Apoptotic cell death by CBD was suppressed by NS-398 (COX-2 inhibitor), GW-9662<br />

(PPARγ antagonist) and siRNA targeting COX-2 and PPARγ. CBD-induced apoptosis was<br />

paralleled by upregulation of COX-2 and PPARγ expression with a maximum induction of<br />

COX-2 mRNA after 8 h and continuous increases of PPARγ mRNA when compared to<br />

vehicle. In response to CBD tumor cell lines exhibited increased levels of COX-2dependent<br />

prostaglandins (PGs) among which PGD2 and 15-deoxy-Δ12 14-PGJ2 caused a<br />

translocation of PPARγ to the nucleus and induced a PPARγ-dependent apoptotic cell<br />

death. Moreover, in A549-xenografted nude mice CBD caused upregulation of COX-2 and<br />

PPARγ in tumor tissue, and tumor regression that was fully reversible by GW-9662.<br />

Together, our data demonstrate a novel proapoptotic mechanism of CBD involving initial<br />

upregulation of COX-2 and PPARγ and a subsequent nuclear translocation of PPARγ by<br />

COX-2-dependent PGs.<br />

114<br />

Synthesis of Zwitterionic Ligands for PET-Imaging in Melanoma<br />

Kriemen, E. 1; Maison, W. 1<br />

1 Pharmaceutical and Medicinal Chemistry, Bundesstraße 45, 20146 Hamburg, Germany<br />

Malignant melanoma, although it is far less prevalent than non-melanoma skin cancers, is<br />

the major cause of death from skin cancer.[1] This is because of the high aggressiveness of<br />

melanoma metastases and their resistance to conventional chemotherapy and external<br />

beam radiation therapy. Therefore an early diagnosis of melanoma is the key issue for<br />

increasing patient survival.[2]<br />

For our approach we focus on PET (positron emission tomography) as an imaging<br />

technique, which relies on decay characteristics of positron emitting (β+-decay)<br />

120 Poster


adionuclides.[3] Modern PET scanners have high sensitivities, but are still limited by<br />

specific binding/uptake to normal tissues and organs which lead to high background.<br />

Recent data showed that “zwitterionic” tracer molecules lower background binding<br />

dramatically by not interacting with serum proteins due to charge shielding.[4]<br />

With the mentioned principle in mind we designed a melanoma targeted radiotracer 1. It<br />

contains a DOTA moiety for proper chelating a radionuclide such as 89Zr and 68Ga.[5] The<br />

classical DOTA molecule is modified by geometrically balanced charge (positive charge<br />

with quaternary ammonium cations and negative charge with sulfonate groups).[4] This<br />

zwitterionic DOTA derivative is conjugated to CycMSH, a cyclic targeting peptide with high<br />

specificity to the melanocortin receptor MC1. The MC1 receptor is overexpressed in more<br />

than 80% of human metastatic melanoma tumor samples and is thus a valuable tumor<br />

marker.[6,7]<br />

We present the synthesis of a zwitterionic DOTA derivative and it’s conjugation to CycMSH<br />

by a solid phase protocol and evaluate the binding properties of this targeted imaging<br />

reagent to MC1.<br />

References:<br />

1. Siegel, R., Naishadham, D., Jemal, A.: CA Cancer J. Clin. <strong>2012</strong>, 62(1): 10–29.<br />

2. Anderson, C. M. et al.: Oncology 1995, 9(1): 1149–1158.<br />

3. Frangioni, J. V.: J. Clin. Oncol. 2008, 26(24): 4012–4021.<br />

4. Choi, H.S. et al.: Angew. Chem. Int. Ed. 2011, 50(28): 6258–6263.<br />

5. Verel, I., Visser, G. W. M., van Dongen, G. A.: J. Nucl. Med. 2005, 46(1): 164–171.<br />

6. Hadley, M. E.: Pigment Cell. Res. 1996, 9(5): 213–234.<br />

7. Guo, H.: J. Nucl. Med. 2011, 52(4): 608–616.<br />

115<br />

Is lysophosphatidylcholine (LysoPC) the key to explain the antimetastatic<br />

effects of empty liposomes?<br />

Ross, T. 1; Schlesinger, M. 1; Raynor, A. 2; Jantscheff, P. 2; Massing U. 2; Bendas, G. 1<br />

1 University of Bonn, Department of Pharmaceutical Chemistry II, An der Immenburg 4, 53121<br />

Bonn, Germany<br />

2 Tumor Biology Center Freiburg, Breisacher Str. 117, 79106 Freiburg, Germany<br />

Liposomes are extensive subject to current anticancer research. However, liposomal<br />

phospholipids (PL) are only regarded as passive carrier constituents. The findings of<br />

Graeser et al. [1] showing that empty liposomes composed of saturated PLs have<br />

antimetastatic effects in different mouse models suggest PL actively interfering in the<br />

metastatic course. Since cellular adhesion receptors are of key importance for the<br />

metastatic route of tumor cells, we proposed affected cell adhesion as the underlying<br />

mechanism. Recently we presumed lysophosphatidylcholine (LysoPC) as key molecule for<br />

this activity. One might assume that, due to the EPR-effect, liposomes accumulate in<br />

neoplastic tissue leading to an increased uptake and metabolisms of saturated PL. Since<br />

phospholipase A2 release is known to be often associated with tumor activity, increased<br />

levels of saturated LysoPC in tumor tissue might result. Jantscheff et al. [2] could show a<br />

reduction of melanoma cell adhesion in vitro as well as an inhibition of metastasis like lung<br />

invasion in vivo of cells pretreated with LysoPC. However, the underlying molecular<br />

mechanisms of LysoPC activity remain to be elucidated. Since any toxic activities of<br />

LysoPC or induction of apoptosis could be excluded, our current research focuses on the<br />

involvement of the plasma membrane affected by LysoPC and consequences for<br />

membrane associated signalling pathways.<br />

Using gas chromatography a rapid change in the lipid composition of the tumor cell<br />

membranes favouring the LysoPC derived fatty acids (FA) was shown under the influence<br />

of different LysoPC derivatives. In order to evaluate the impact of FA saturation on tumor<br />

membrane rigidification and underlying functional consequences, we compared the fully<br />

saturated Lyso-PC C18:0 and the unsaturated C18:1-derivative in MV3 melanoma cells.<br />

Using trimethylammoniumdiphenylhexatriene as a fluorescence anisotropy probe we could<br />

detect a significant rigidification of the cell membrane by C18:0 LysoPC, while C18:1<br />

LysoPC possessed only minor changes. The changes in membrane rigidity correspond with<br />

a diminished receptor-mediated migration capacity of the cells, as shown in microscopic<br />

wound healing assays. Thus, one might assume that LysoPC-induced membrane<br />

rigidification is involved in the reduced receptor mediated cell binding which might be one<br />

reason for the antimetastatic effects of LysoPC.<br />

To further focus on the molecular mechanisms of diminished receptor mediated cell binding<br />

we present preliminary data on the phosphorylation of the proteoglycan syndecan-4, which<br />

is an important element in the formation of focal adhesions complexes. Summarizing, we<br />

propose LysoPC as an active biological molecule affecting the plasma membrane fluidity<br />

with strong consequences for the formation of focal adhesion complexes. These data could<br />

also provide new insights and prospects for liposomal carriers in tumor therapy.<br />

References:<br />

1. Graeser, R. et al.: Pancreas 2009, 38(3): 330-337<br />

2. Jantscheff P. et al.: Mol Cancer Ther 2011, 10(1): 186-197<br />

116<br />

NEFA determination for functional lipidomics: comprehensive UPLC-MS/MS<br />

quantitation, qualification and parameter prediction [1]<br />

Hellmuth C, Weber M, Koletzko B, Peissner W<br />

Division of Metabolic and Nutritional Medicine, Dr. von Hauner Children’s Hospital, Ludwig-<br />

Maximilians-University of Munich, 80337 Munich, Germany<br />

Objectives<br />

Metabolomics is an increasing field in pharmaceutical research and development.<br />

Metabolic profiling depicts abundance changes in endogenous metabolites [2]. E.g. levels<br />

of nonesterified fatty acids (NEFA) are highly associated with obesity, diabetes and<br />

metabolic syndrome. For instance, NEFA are elevated in T2DM adolescents in fasting state<br />

as well as after glucose challenge [3]. Though, straightforward quantitative methods for<br />

determination of NEFA species are still missing.<br />

Methods<br />

Serum samples were prepared by simple protein precipitation, avoiding cumbersome and<br />

solvent-consuming extraction or derivatization procedures. Short run time with high<br />

resolution was achieved by UPLC separation coupled to LC-MS/MS detection.<br />

Results<br />

Accurate quantification of 36 NEFA species in 20 µl of healthy human plasma was<br />

achieved, the highest numbers ever reported for a LC-MS application. The use of qualifier<br />

ions supports unequivocal analyte verification. Sample preparation is fast and nonexpensive.<br />

In combination with automated liquid handling, total assay time per sample is<br />

less than 15 minutes.<br />

Conclusion<br />

The presented method provides a rapid and comprehensive determination of NEFA in<br />

human plasma. This enables application in clinical trials with high sample number analyzed<br />

in short time and with low costs. Usage of 20 µl plasma facilitates application in studies<br />

with children or adolescents, when blood is obtained by finger sticks or heel pricks.<br />

References:<br />

1. Hellmuth C. et al.: Anal Chem. <strong>2012</strong> 84(3):1483-90<br />

2. Xu EY, Schaefer WH, Xu Q: Curr Opin Drug Discov Devel. 2009 12(1):40-52<br />

3. Nadeau KJ et al.: J Clin Endocrinol Metab. 2009 94(10):3687-95<br />

117<br />

The impact of DNA methylation on cellular differentiation of immune cells<br />

Flemming S. 1; Grützkau A. 2, Häupl T. 3; Günther S. 1<br />

1 Pharmaceutical Bioinformatics, Institute of Pharmaceutical Sciences, University of Freiburg<br />

2 German Arthritis Research Center, Berlin<br />

3 Department of Rheumatology and Clinical Immunology, Charité University Hospital, Berlin<br />

Epigenetic changes in DNA methylation are associated with regulation of gene expression<br />

and play an important role in cellular differentiation. Such changes might be of importance<br />

in chronic inflammatory diseases where autoreactive immune cells are thought to<br />

perpetuate inflammation and organ destruction. To explore these variations and its<br />

regulatory mechanism, we have gathered genome-wide DNA methylation data in human<br />

immune cells extracted from peripheral blood, specifically T-helper cells (CD4), Tsuppressor<br />

cells (CD8), macrophages (CD14), granulocytes (CD15), B-lymphocytes<br />

(CD19) and natural killer cells (CD56). Furthermore, we have analyzed methylation patterns<br />

associated to cell differentiation of T- and B-cells and the correlation with gene expression<br />

data.<br />

Cells from four healthy donors were sorted by FACS technology and the measurement was<br />

performed with the Illumina HumanMethylation450 BeadChip platform. These arrays<br />

provide a genome-wide coverage of more than 485,000 CpG methylation sites per sample<br />

at single-nucleotide resolution. To evaluate the correlation between methylation patterns<br />

and gene expression, we used the Affymetrix U133P platform.<br />

We observed that both, hyper- and hypomethylation of promoter regions of genes involved<br />

in cell differentiation correlate well with measured gene expression data. Additionally, we<br />

were able to identify a substantial number of common CpG methylation sites, which were<br />

differentially methylated between naive and memory states of T-(CD4, CD8) and B-cells<br />

(CD19).<br />

The impact of these alterations on inflammatory diseases will be a subject to further<br />

research which will include the analysis of autoreactive immune cells from rheumatoid<br />

arthritis patients.<br />

118<br />

Role of Histamine H4 receptors in eicosanoid biosynthesis.<br />

Dos Santos Capelo, R1; Kahnt, AS1; Steinhilber, D1 1 Institute of Pharmaceutical Chemistry, Max-von-Laue-Strasse. 9, 60438 Frankfurt am Main,<br />

Germany<br />

The H4 receptor is a G-protein-coupled receptor (GCPR) expressed in many cells of the<br />

immune system, including monocytes, mast cells, dendritic cells, natural killer cells,<br />

eosinophils and T cells. So far, not much is known about its physiological role, but the<br />

receptor is implicated in the pathology of various diseases such as allergy, autoimmune<br />

disorders, bronchial asthma, atopic dermatitis and pruritus. Upon activation the receptor<br />

couples to Gi/o proteins. This leads to a decrease in cAMP production [1,2,3,4], activates<br />

downstream mitogen-activated protein (MAP) kinase pathways [4] and mobilizes<br />

intracellular Ca2+ [1,5]. 5-lipoxygenase (5-LO) catalyzes the first two steps in leukotriene<br />

(LT) biosynthesis. Upon stimulation (e.g. MAP kinases and Ca2+ influx) the enzyme<br />

translocates to perinuclear membrane compartments where it is activated and starts the<br />

synthesis of LTA4 [6]. LTA4 can then be further metabolized by two terminal synthases.<br />

Soluble LTA4 hydrolase produces the potent chemoattractant LTB4, whereas membrane<br />

bound LTC4 synthase conjugates LTA4 with glutathione (GSH) to form the corresponding<br />

cysteinyl leukotrienes (CysLTs), LTC4, LTD4 and LTE4. Like the H4 receptor, 5-LO is<br />

primarily found in mature leukocytes such as granulocytes, mast cells, dendritic cells,<br />

monocytes / macrophages and B lymphocytes [7]. T cells and erythrocytes are 5-LO<br />

negative. 5-LO derived LTs are lipid mediators which were shown to primarily mediate<br />

inflammatory and allergic reactions. Besides their well studied role in asthma, 5-LO derived<br />

LTs have also been implicated to play a role in cardiovascular diseases and cancer [8].<br />

Recently, a connection between LT biosnythesis and histamine receptors has been drawn.<br />

Flamand et al. have published that histamine is able to inhibit LT formation in human PMN<br />

in a dose-dependent manner via Gs coupled H2 receptor activation. This inhibition was<br />

characterized by cAMP dependent protein kinase A (PKA) activation which in turn led to a<br />

decrease in arachidonic acid release and 5-LO translocation [9]. Based on this, we<br />

postulate that the human histamine H4 receptor might control LT biosynthesis in activated<br />

human leukocytes such as macrophages and eosinophils. Using western blot and<br />

polymerase chain reaction (PCR) technique we have investigated the H4 receptor<br />

expression in various cell lines and primary cells and have chosen suitable cell lines. Next,<br />

we plan to treat these cells with different cytokines plus histamine and monitor 5-LO<br />

activity. After this, we plan to elucidate the underlying mechanism to provide the basis for<br />

Poster 121


the development of potential new anti-leuktriene drugs.<br />

References:<br />

1. Oda, T. et al.: J. Biol. Chem. 2000, 275: 36781–36786.<br />

2. Nakamura, T. et al.: Biochem. Biophys. Res. Commun. 2000, 279: 615–620.<br />

3. Zhu, Y. et al.: Mol. Pharmacol. 2001, 59: 434–441.<br />

4. Liu, C. et al.: Mol. Pharmacol. 2001, 59: 420–426.<br />

5. Morse, K.L. et al.: J. Pharmacol. Exp. Ther. 2001, 296: 1058–1066.<br />

6. Rouzer C.A. et al.: J. Biol. Chem. 1988, 263(22): 10980-10988.<br />

7. Heaggstrom J.Z. et al.: Cell. Mol. Life. Sci. 2002, 59(5):742-753.<br />

8. Werz O. et al.: Pharmacol. Ther. 2006, 112(3):701-718.<br />

9. Flamand N. et al.: Br. J. Pharmacol. 2004, 141(4): 552-561.<br />

119<br />

Quantitative Analysis of Isolated EGFR Adaptor Protein Pattern by NSAF and<br />

SILAC Reveals Cellular Dynamics of Receptor Signaling and Internalization<br />

Foerster, S. 1, Hammer, E. 2; Kacprowski, T. 3, Albrecht, M. 3, Völker, U. 2, Ritter, C.A. 1<br />

1 Clinical Pharmacy, Institute of Pharmacy, <strong>Ernst</strong>-<strong>Moritz</strong>-<strong>Arndt</strong>-University, Friedrich-Ludwig-Jahn-<br />

Str. 17, 17487 <strong>Greifswald</strong>, Germany<br />

2 Department of Functional Genomics, Interfaculty Institute of Genetics and Functional Genomics,<br />

<strong>Ernst</strong>-<strong>Moritz</strong>-<strong>Arndt</strong>-University, Friedrich-Ludwig-Jahn-Str. 15, 17487 <strong>Greifswald</strong>, Germany<br />

3 Department of Bioinformatics, Institute of Biometrics and Medical Informatics, University Medicine<br />

<strong>Greifswald</strong>, Ferdinand-Sauerbruch-Str. 1, 17475 <strong>Greifswald</strong>, Germany<br />

Overexpression, aberrant or enhanced activation of the epidermal growth factor receptor<br />

(EGFR) are frequent events in human cancers. By recruiting specific adaptor proteins to the<br />

activated intracellular receptor tyrosine kinase domain, EGFR signaling regulates<br />

proliferation, growth and differentiation [1]. We hypothesize that cellular responses are<br />

realized by characteristic patterns of adaptor proteins, which switch respective signal<br />

cascades and therefore developed a method to analyze the EGFR adaptor protein pattern<br />

in relation to its physical state.<br />

The EGFR-adaptor protein-complex was isolated by immunoprecipitation using the specific<br />

EGFR antibody cetuximab,which was biotinylated and bound to magnetic beads. A431 cells<br />

were stimulated with 100 ng/ml EGF for 30 minutes or left untreated and EGFR-associated<br />

protein complexes were precipitated from respective cell lysates. For stable isotope labeling<br />

in cell culture (SILAC), the stimulated cells were first adapted to heavy isotopes ( 13C) of Llysine<br />

and L-arginine and light cells were left untreated or incubated with 10 µM Tyrphostin<br />

AG1478 for 1 h before stimulation with EGF. Lysates of heavy ( 13C) and light ( 12C) cultures<br />

were mixed in a 1:1 ratio for precipitation. After pre-fractionation by 1D-SDS-PAGE and ingel-digestion,<br />

protein complexes were characterized by LC-ESI-tandem high resolution<br />

mass spectrometry (LC-MS/MS). SILAC provides a ratio of heavy/light peptides for<br />

quantification of proteins in stimulated and control samples while the label free complexes<br />

were analyzed with the normalized spectral abundance factor (NSAF), which overcomes<br />

sample to sample variation and enables a semi-quantitative determination of proteins from<br />

different samples [2].<br />

A set of 58 proteins was reproducibly identified in three LC-MS/MS replicates. The proteinprotein<br />

interactions within this set were obtained from the iRefIndex database and the<br />

corresponding protein interaction network was visualized in Cytoscape. The Gene Ontology<br />

annotations of the proteins were clustered and visualized in the network as well.. Most<br />

proteins are involved in intracellular localization and protein transport (27 proteins) or cell<br />

death (13 proteins). The first domain contained structural proteins such as actin and tubulin<br />

subunits as well as importins, exportins and transportins. The cell death regulating category<br />

included serine/threonine-protein phosphatase 2A and heat shock protein-ß1. Furthermore,<br />

several proteins were found which associations to the EGFR are yet unknown, such as<br />

Hook homolog 2 and cancerous inhibitor protein 2A. Quantification by SILAC identified<br />

subunits of the clathrin related adaptor protein complex, which were markedly pronounced<br />

upon EGF stimulation, suggesting increased receptor internalization. These data were<br />

confirmed by Western Blotting and immunofluorescence microscopy. EGFR signaling<br />

related proteins like growth factor receptor bound protein 2 (Grb2) and signal transducer<br />

and activator of transcription 3 (STAT3) were not subjected to stimulation dependent<br />

changes. Further dynamic changes revealed proteins which were affected by stimulation<br />

after inhibition of tyrosine kinase activity. This might indicate that alternative EGFR<br />

signaling pathways occurred. For instance, SHC-transforming protein 1 decreased upon<br />

stimulation and even more when EGFR activity was inhibited. Nevertheless, adaptor<br />

proteins are low abundant proteins, which is why we will work out a protein cross linking<br />

strategy to enrich for specific signaling molecules.<br />

As proof of principle, we were able to identify proteins that are part of the EGFR signaling<br />

pathway as well as proteins that relate to cellular localization and protein trafficking and<br />

whose abundances changed upon EGFR stimulation. Using this approach we will provide<br />

further insights into mechanisms of EGFR signal activation from a systems biology point of<br />

view.<br />

References:<br />

1. Oda, K. et al.: Mol.Syst.Biol. 2005, 1:2005.0010.<br />

2. Gokce, E. et al.: J.Am.Soc.Mass Spectrom. 2011, 22: 2199-2208.<br />

120<br />

Metabolism studies of Ifenprodil, a potent NR2B-selective NMDA receptor<br />

antagonist<br />

Falck, E.; Wünsch, B.<br />

Institut für Pharmazeutische und Medizinische Chemie der Westfälischen Wilhelms-<strong>Universität</strong>,<br />

Hittorfstr. 58-62, 48153 Münster, Germany<br />

Ifenprodil is known as an antagonist for a special binding site at the NMDA receptor subunit<br />

NR2B, called ifenprodil binding site [1]. NMDA receptors change their concentration and<br />

composition with the development of the brain during the lifetime period [2]. Furthermore,<br />

NMDA receptors containing the NR2B subunit are restricted in specific sites of the human<br />

brain [3]. This makes the NR2B subunit an interesting target for the therapy of<br />

neurodegenerative diseases such as schizophrenia, epilepsy or Parkinson’s disease.<br />

Epilepsy for example causes neuronal cell death which leads to an increased glutamate<br />

concentration. This induces an overactivation of the NMDA receptor inducing further cell<br />

death. Ifenprodil as an antagonist can reduce NMDA receptor activation and protect the<br />

brain against further cell damage. The ifenprodil binding site is located on the surface of the<br />

NR2B subunit and can interact with other binding sites located at the receptor. Ifenprodil<br />

has a high affinity to the receptor but a rather low selectivity. The interaction with other<br />

receptors leads to undesired side effects, e. g. impaired motor function. The ifenprodil<br />

binding site at the NMDA receptor represents an interesting target and ifenprodil a<br />

promising lead compound for the development of innovative NMDA receptor antagonists<br />

with comparable affinity but increased selectivity.<br />

In addition to the poor receptor selectivity fast metabolic degradation and low bioavailability<br />

of ifenprodil have been reported in literature. The precise knowledge about the<br />

biotransformation of ifenprodil is important for the design of metabolically more stable<br />

analogues. However, the metabolism of this compound has not been studied in detail.<br />

Herein we report the metabolism of ifenprodil, regarding the phase I and phase II<br />

metabolites resulting from hydroxylation and conjugation reactions.<br />

Transformations were carried out with rat liver microsomes and the essential co-factors<br />

NADPH/H +, UDPGA, PAPS and SAM. The metabolites were identified by fragmentation<br />

steps carried out using HPLC-ESI-MS n. The supposed structures of the resulting fragments<br />

were confirmed by recording the exact mass yielding from time-of-flight detection. All<br />

metabolites were identified by comparing their fragmentation pattern with the typical<br />

fragmentation pattern observed with the parent compound ifenprodil.<br />

References:<br />

1. Williams, K.: Curr. Drug Targets 2001, 2: 285-298.<br />

2. Chazot, P. L.: Curr. Med. Chem. 2004, 11: 389-396.<br />

3. Childers, W. E., Baudy, R. B.: J. Med. Chem. 2007, 20: 2557-2562.<br />

122 Poster<br />

1<br />

121<br />

11-Keto-β-boswellic acids prevent insulitis in 2 animal models with<br />

autoimmune diabetes<br />

Ammon, H.P.T. 1, Shehata, A.M. 2, Jauch, J. 3, Bettio S. 4, Quintanilla-Fend, L. 4<br />

1Department of Pharmacology, Institute of Pharmaceutical Sciences, University of Tuebingen,<br />

2Department of Pharmacology, Faculty of Pharmacy, University of Beni-Sueif, Beni-Sueif, Egypt,<br />

3Institute of Organic Chemistry II, University of Saarland, Saarbrücken, Germany,<br />

4Institute of Pathology, University of Tuebingen, Germany<br />

Background and aims: Type 1 diabetes and Late onset Autoimmune Diabetes of the Adult<br />

(LADA) are autoimmune diseases where a chronic inflammatory process destroys insulin<br />

producing β-cells. Boswellic acids which derive from gum resin of Boswellia species<br />

(Olibanum) have been shown to possess immunmodulatory properties and suppress<br />

proinflammatory cytokines including NFκB (1). In the model of Multiple Low Dose-<br />

Streptozotocin ((MLD-STZ) diabetes and the genetic Non Obese Diabetic (NOD)mouse,<br />

which are thougt to correspond to human one type diabetes, it was tested whether or not<br />

an extract of the gum resin of boswellia serrata (BE), 11-Keto-β-boswellic acid (KBA) and<br />

Acetyl-O-11-keto-β-boswellic acid (AKBA) could prevent developement of insulitis and<br />

appearance of apoptotic cells in the pancreatic islets, increase of proinflammatory<br />

cytokines in the blood and increase of blood glucose level.<br />

Methods: BK +/+ male mice were i.p. injected with 40 mg/kg STZ for 5 days. A second<br />

group received 150 mg/kg BE, a third group 15 mg/kg AKBA and a fourth group 7.5 mg/kg<br />

KBA simultaneously over a period of 10 days. Infiltration of lymphocytes into pancreatic<br />

islets and apoptosis of islet cells were determined histochemically (CD-3 antibodies and<br />

anticaspase-3) after day 10. In addition, proinflammatory cytokines were determined in<br />

serum. Blood glucose was measured over a period of 35 days.<br />

Four weeks old NOD-mice received daily 7.5 mg/kg KBA i.p. over a period of three weeks.<br />

Thereafter the pancreatica have been taken for histochemical studies.<br />

Results: 10 days after first STZ injection there was infiltration of lymphocytes into<br />

pancreatic islets and appearance of apoptotic cells. Moreover, all tested proinflammatory<br />

cytokines in the serum were significantly increased. There was also a continuous increase<br />

of blood glucose. Simultaneous i.p. administration of BE, AKBA and KBA significantly<br />

reduced cytokines in the serum, infiltration of lymphocytes and apoptosis of islet cells. After<br />

21 days blood glucose levels showed no increase.<br />

In NOD-mice which develope insulitis and first apoptotic cells in the pancreas after four<br />

weeks of age administration of KBA prevented both to a large extent.<br />

Conclusion: An extract of the gum resin of Boswellia serrata (olibanum) and two of its<br />

constituents i. e. AKBA and KBA can prevent developement of insulitis probably through<br />

their inhibitory action on proinflammatory cytokines. 11-Keto-β-boswellic-acids seem to be<br />

promising compounds for the prevention and treatment of autoimmune diabetes.<br />

Supported by: The authors are grateful for support for the Dr. h. c. Bürger-Büsing Foundation and the<br />

German Diabetes Foundation.<br />

References:<br />

(1) Ammon, HPT: Phymed. 2010, 17: 862-867.<br />

122<br />

The Thermal Behavior of Histidine in Frozen Aqueous Solutions: The Effect of<br />

pH, Annealing, Histidine Concentration and Formulation Composition.<br />

Alhussein, A. 1 ;Gieseler, H. 1 2<br />

1Department of Pharmaceutics-University of Erlangen, Cauerstrasse 4, 91058 Erlangen, Germany<br />

2 GILYOS GmbH, Friedrich-Bergius-Ring 15, 97076 Wuerzburg, Germany<br />

Therapeutic proteins are a major focus of research and development activities in the<br />

pharmaceutical industry. Due to their limited stability in aqueous solutions, proteins often<br />

need to be converted into solid state to achieve an acceptable shelf life as pharmaceutical<br />

products [1]. The most commonly used method for manufacture of solid protein<br />

pharmaceuticals is freeze drying [1,2]. In turn, freeze drying process (lyophilization)<br />

generates many stresses during both freezing and drying which may cause the loss of<br />

protein bio-activity. Therefore many excipients are added to the protein formulation to<br />

overcome these stresses and to improve protein stability during freeze drying.<br />

Disaccharides, such as sucrose and trehalose, are widely used as protein stabilizers (cryoand<br />

lyoprotectant), and these sugars have been extensively studied in the literature to<br />

investigate thier stabilizing effect on proteins during the lyophilization. However, formulation<br />

development to achieve the best stabilizing effect on protein is still a great challenge for the<br />

experts of freeze drying.


In the last few years, amino acids are attracting increased attention in the field of the freeze<br />

drying of proteins, and many amino acids were used as protein stabilizer in the freeze dried<br />

products [3,4]. In contrast to the sugars, amino acids can also function as buffers in protein<br />

formulations. Therefore amino acids provide more choices for the design of proteins<br />

formulations. Histidine is one of the amino acids that can be added to the protein<br />

formulations to function as both buffer and protein stabilizer [5].<br />

It is well known that amorphism is an essential property for stabilization of proteins during<br />

freeze drying. Protein stabilizer should remain amorphous (in the same phase with protein)<br />

to be able to interact with the protein molecule by the formation of hydrogen bonds, leading<br />

thereby to the preservation of protein native structure [6]. Since the stabilizing effect of an<br />

amino acid (or other stabilizer) during lyophilization is directly correlated with its physical<br />

state, it is important to evaluate the stabilizing efficiency by studying the changes in the<br />

physical state of the amino acid upon cooling and heating.<br />

The aim of this study was to investigate the thermal behavior of histidine in frozen aqueous<br />

solutions at different conditions using DSC technique (Differential Scanning Calorimetry).<br />

This research focused on tow thermal events: Histidine crystallization and the glass<br />

transition of histidine. The effect of histidine concentration and pH of histidine solution on<br />

the physical state of histidine was investigated. Furthermore, some of popular excipients<br />

which are widely used in the freeze dried protein formulations (such as sucrose, mannitol,<br />

and Tween 80) were added into histidine solutions to evaluate thier effect on histidine<br />

during the DSC measurement. Additionally, the effect of annealing (which is a common<br />

practice applied during the freeze drying to enhance the crystallization of the bulking<br />

agents) was investigated, where all DSC scans of the studied formulations were performed<br />

with/without annealing to detect the possible effect of this step on the thermal behavior of<br />

histidine.<br />

The results showed that histidine crystallization may occur during the heating phase of the<br />

DSC-scan and this event depends strongly on both pH and histidine concentration.<br />

However, the enhancement of histidine crystallization by application of annealing seems<br />

also to be pH-dependent. Furthermore, the glass transition temperature of histidine (Tg )<br />

was greatly varied according to the pH of solution. The addition of sucrose to the histidine<br />

solution resulted in an inhibition of histidine crystallization, while the addition of mannitol or<br />

Tween 80 seems to have no inhibitory effect. The interpretation of histidine thermal<br />

behavior in the systems containing both mannitol and sucrose is more complex and<br />

depends on several factors including the weight ratio of mannitol to sucrose in the<br />

formulation.<br />

References:<br />

1. Wang, W.: Int. J. Pharm. 2000, 203(1): 1-60.<br />

2. Pikal, MJ.: Encyclopedia of Pharmaceutical Technology (Marcel Dekker Publishing Co. New York) 2002.<br />

3. Österberg, T.: Pharm. Res. 1997, 14(7): 892-898.<br />

4. Izutsu, K. et al.: Int. J. Pharm. 2005, 301(1-2): 161-169.<br />

5. Österberg, T.: Eur. J. Pharm. Sci. 1999, 8(4): 301-308.<br />

6. Carpenter, JF. et al.: J. Dairy. Sci. 1990, 73(12): 3627–3636.<br />

123<br />

Biomimetic catecholates for the chemical modification of metal surfaces:<br />

antifouling properties<br />

Khalil, F. 1; Maison, W. 1<br />

1 Pharmaceutical and Medicinal Chemistry, Bundesstraße 45, 20146 Hamburg, Germany<br />

Biofouling is the non-specific attachment of biological material (proteins, carbohydrates and<br />

prokaryotic cells) to surfaces upon their exposure to any biological milieu [1]. Biofouling is<br />

of great concern in many areas ranging from marine technology, biosensors, dentistry to<br />

biomedical implants and devices. The most common approach to prepare non-fouling<br />

surfaces is through the immobilization of polymers such as poly(ethylene glycol) (PEG),<br />

which are able to repel the adhesion of proteins and cells [2].<br />

Methods for the covalent immobilization of molecular monolayers rely on the use of<br />

functionalized anchor molecules, including thiols, silanols, phosphates or phosphonates. All<br />

of these immobilization techniques have drawbacks, such as limited stability, elaborate<br />

syntheses or a limited range of substrate materials. Catecholate, inspired by musseladhesive<br />

proteins (MAPs) [3] and bacterial siderophores [4,5], have received considerable<br />

interest because of their high binding affinity to various surfaces [6].<br />

By following a biomimetic approach, we have synthesized conjugates of catecholates and<br />

bifunctional tetravalent scaffolds based on a trisalkylmethyl core structure. Three<br />

catecholateamines have been conjugated to a central scaffold, resembling the tripodal<br />

metal binding motif of natural metal binders, such as MAPs and siderophores [7]. The<br />

resulting trimeric catecholates are easily coupled to different effector molecules for the<br />

stabile immobilization of effector molecules such as PEG on metal surfaces.<br />

Futher, the immobilization of these multivalent catecholate derivatives on TiO2 surface have<br />

been performed and showed promising antifouling properties.<br />

References:<br />

1. Hall-Stoodley, L., Costerton, J. W., Stoodley, P., Nat. Rev. 2004, 2(2), 95-108.<br />

2. Banerjee, I., Pangule, R. C., Kane, R. S., Adv. Mater. 2011, 23(26), 690-718.<br />

3. Waite, J. H., Tanzer, M. L., Science 1981, 212(4498), 1038-1040.<br />

4. Loomis, L. D., Raymond, K. N., Inorg. Chem. 1991, 30(5), 906-911.<br />

5. Gademann, K., Chimia 2007, 61(6), 373-377<br />

6. Lee, H., Dellatore, S. M., Miller, W. M., Messersmith, P. B., Science 2007, 318(5849), 426-430.<br />

7. Franzmann, E., Khalil, F., Weidmann, C., Schröder, M., Rohnke, M., Janek, J., Smarsly, B., Maison, W.,<br />

Chem. Eur. J. 2011, 17(31), 8596-8603.<br />

124<br />

Quantification of impurities with NMR spectroscopy: Comparison of different<br />

approaches to quantify small signals overlapped with large signals using the<br />

example topiramate.<br />

Wiest , J.; Ilko , D.; Holzgrabe, U.<br />

Institute of Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg,<br />

Germany<br />

Molecules without chromophores like topiramate (TOP) pose a challenge to analysts as<br />

they cannot easily be quantified via common photometric detection regimes like UV/VIS or<br />

fluorometry. Our aim was to develop an HPLC-CAD method for the determination of purity<br />

and content of TOP and to compare the results to the existing HPLC refractive index<br />

detection (HPLC-RI) method which has a poor sensitivity and a limited performance. The<br />

newly developed HPLC-CAD method obtained good results for quantification of TOP and<br />

the other related substances, however the main impurity diacetone fructose (DAF) could<br />

not be determined due to its high vapour pressure. To solve this problem, we tried to apply<br />

NMR spectroscopy. Prerequisite of qNMR is a clear separation of the signals of all<br />

components used for quantification. Variation of the solvent resulted in pyridine-d5 which<br />

gave the best signal separation.The spectra were acquired with inverse gated 13C<br />

decoupling using globally optimized alternating phase rectangular pulse (GARP) to remove<br />

disturbing 13C satellites. To avoid rotation sidebands the spectra were measured without<br />

spinning. Processing the spectra has a great influence on the results. Thus we compared<br />

the results of four Topiramate batches with an internal standard using a standard addition<br />

method. The results were obtained with three different approaches: (i) Deconvolution of the<br />

overlapping signals executed with TopSpin 3.0; (ii) Integration after baseline adjustment (iii)<br />

Quantification by signal height. An experimental protocol was found which is particularly<br />

suitable for quantification of DAF residues in TOP samples.<br />

References:<br />

Almeling, S.; Ilko, D.; Holzgrabe, U.: J. Pharm. Biomed. Anal. <strong>2012</strong>, Epub ahead of print<br />

www.elsevier.com/locate/jpba , 23.05.<strong>2012</strong><br />

Pauli, G.F.: Phytochem. Anal. 2001, 12: 28-42<br />

125<br />

Impact of drug-drug interactions on the INR values of patients treated with<br />

Marcumar ® in the ambulatory care setting<br />

Bertram, L. 1; Krämer, I. 1<br />

1 Department of Pharmacy, University Medical Center of Johannes Gutenberg-University,<br />

Langenbeckstr.1, 55131 Mainz, Germany<br />

PURPOSE: Treatment with the oral vitamin K antagonist phenprocoumon (Marcumar ®) is<br />

associated with a high risk of interactions with additionally prescribed medication and selfmedication.<br />

In consequence the efficacy of Marcumar ® can be increased or reduced and<br />

the resulting INR-values may drift out of the therapeutic range. Thus the risk of thromboembolic<br />

events or bleeding increases.<br />

The aim of this study was to investigate whether patients’ INR-values in the ambulatory<br />

health-care sector in Germany are influenced by additionally prescribed potentially<br />

interacting drugs [1].<br />

METHODS: 50 patients treated with Marcumar ® were evaluated in a retrospective study<br />

over three years (1800 months in total). The INR-values and type of comedication of ten<br />

patients each recruited in five physician offices in Rhineland-Palatinate were retrieved from<br />

the patients’ medical records. The percentage rate of INR values lying in the target range<br />

(e.g. INR 2-3) and extended target range (target range +/- 0.2) was evaluated for individual<br />

patients. Time in therapeutic range (TTR) was calculated by using the Rosendaal-Method<br />

[2]. Potentially interacting drugs associated with serious interactions were chosen from the<br />

literature [1]. The Mann-Whitney-U-test was used to determine any significant relationship.<br />

RESULTS: Patients taking potentially interacting drugs in addition to a permanent<br />

phenprocoumon therapy achieved lower rates of adequate INR values than patients not<br />

taking potentially interacting drugs. The median percentage rate of INR values within the<br />

therapeutic range amounted to 69% without interacting comedication and 53% with using<br />

interacting comedication (p= 0.008) as well as 83% versus 67% (p= 0.076) for the median<br />

extended therapeutic range, and 75% versus 61% (p= 0.002) for median TTR respectively.<br />

CONCLUSION: Results document that the concomitant intake of interacting drugs<br />

significantly affects the rate of INR-values in therapeutic range and TTR. With regard to the<br />

extended target INR range the difference is not significant. However study results clearly<br />

show that the efficacy of phenprocoumon in routine clinical practice is depending on the<br />

potentially interacting concomitant drug therapy. These combinations should be avoided<br />

whenever possible because additional monitoring is necessary and safety of the<br />

anticoagulant therapy is compromised.<br />

References:<br />

1. S. Isstas, „Pharmakovigilanz bei Patienten mit Phenprocoumon-Therapie- Untersuchungen zu<br />

potentiellen Interaktionen und Betreuungsqualität“, dissertation, Johannes Gutenberg-UniversityMainz,<br />

2011.<br />

2. Rosendaal, F.R. et al.: Thromb. Haemost.1993, 69: 236-239.<br />

126<br />

A novel combinative particle size reduction technology for drug nanocrystals<br />

production and further nanosuspension transfer to tablets<br />

Poster 123


Salazar J1, Müller R H1, Möschwitzer J P1 2<br />

1 Institute of Pharmacy, Dept. of Pharmaceutics, Biopharmaceutics and NutriCosmetics, Freie<br />

<strong>Universität</strong> Berlin, Kelchstrasse 31, 12169 Berlin, Germany<br />

2 Pharmaceutical Development, Abbott GmbH & Co. KG, Knollstrasse 50, 67061 Ludwigshafen am<br />

Rhein, Germany<br />

The aim of this research was to systematically analyze the process parameters affecting<br />

the comminution effectiveness of a novel particle size reduction method. This combinative<br />

method employs spray-drying (SD) as bottom-up step and drug pre-treatment followed by<br />

high pressure homogenization (HPH) as top-down process for nanosuspension production<br />

1 2. The liquid nanosuspensions were then transferred to immediate release (IR) tablets by<br />

applying downstream techniques such as wet granulation (WG), freeze-drying (FD) or SD.<br />

Finally, a dissolution test was performed for drug release assessment.<br />

The poorly water-soluble compound glibenclamide (BCS Class II) was used as model drug.<br />

The influence of drug and surfactant concentrations during SD on the particle size reduction<br />

effectiveness of the HPH was analyzed. The drug was dissolved in ethanol to prepare<br />

solution concentrations of 1%, 2% or 3% (w/v). The amounts of docusate sodium salt<br />

(DSS) in the ethanolic solution were 0%, 0.1%, 0.2%, 0.3% or 0.4% (w/v). The ethanolic<br />

solutions were then spray-dried using a Mini Spray Dryer B-290 coupled to an Inert Loop B-<br />

295 (Büchi Labortechnik AG, Switzerland). The morphology and solid state of the spraydried<br />

powders were analyzed by SEM, DSC and PXRD. Suspensions were produced by<br />

dispersing 1% (w/w) drug (treated/untreated) in demineralized water using an Ultra-Turrax<br />

T-25 (IKA Werke GmbH & Co. KG, Germany). In the case of the drug powders processed<br />

without surfactant DSS 0.2% (w/v) was added as dispersant. The different suspensions<br />

were then homogenized by employing the HPH technique (1500 bar, 20 cycles) using a<br />

Micron LAB 40 homogenizer (APV Systems, Germany). The particle size of the<br />

nanosuspensions was analyzed using photon correlation spectroscopy and laser<br />

diffractometry. The liquid nanosuspensions were down-streamed by WG, FD or SD and<br />

then processed to tablets using lactose monohydrate as filler. Mannitol was used as matrix<br />

former for FD and SD (0%-6% w/v). A dissolution test was performed to determine the<br />

immediate drug release performance of the tablets.<br />

It was found that high drug concentrations and middle DSS concentration resulted in<br />

porous, flowable and amorphous drug powders after SD. These characteristics resulted in<br />

smaller particle sizes after reduction. The drug pre-treatment can significantly improve the<br />

particle size reduction effectiveness of the HPH. The best nanosuspension produced with<br />

the spray-dried powders (2% drug, 0.2% DSS during SD) showed a very small particle size<br />

(236 nm mean particle size, d50% of 0.131 µm and d90% of 0.285 µm) and a narrow size<br />

distribution. The best drug release was 90% after 10 min and it was delivered by SD of the<br />

best nanosuspension with 2% mannitol before tableting. This drug release was markedly<br />

better compared to the one delivered by the tablets processed with micronized drug (26%<br />

drug release after 10 min). The rank order of the tablet release rate employing the<br />

downstream techniques was: SD tablets > FD tablets > WG tablets. The surfactant and<br />

drug concentrations used for the SD process are critical and need to be selected carefully.<br />

The transfer of the drug nanocrystals to tablets was successfully achieved.<br />

References:<br />

[1]: Möschwitzer, J., Method for producing ultrafine submicronic suspensions, WO/002006094808A3, 2006<br />

[2]: Möschwitzer, J., Müller R.H., New method for the effective production of ultrafine drug nanocrystals,<br />

Journal of Nanoscience and Nanotechnology 6(9-10): 3145-3153, 2006<br />

127<br />

Differential scanning calorimetry as a supportive tool for the analysis of<br />

nanomilled fenofibrate suspensions<br />

Komoß, C. 1; Bitterlich, A. 2; Kwade, A. 2; Bunjes, H. 1<br />

1Institute of Pharmaceutical Technology, TU Braunschweig, Mendelssohnstraße 1, 38106<br />

Braunschweig, Germany<br />

2Institute for Particle Technology, TU Braunschweig, Volkmaroder Straße 5, 38104 Braunschweig,<br />

Germany<br />

An important approach to overcome poor dissolution properties and the often resulting bad<br />

biopharmaceutical profile of active pharmaceutical ingredients (API) is to reduce the API´s<br />

particle size, a process also known as “nanosizing” [1]. This leads to a strongly increased<br />

surface-to-volume ratio and, thus, to an increased dissolution rate. However, the large<br />

specific surface area creates a positive gain of free energy. Hence, there is a tendency<br />

towards crystal growth, (re-)agglomeration of the particles and, thus, instability of the<br />

nanosuspensions.<br />

A reduction of particle size can also lead to altered solid state properties like a reduced<br />

melting point [2]. Therefore, in this study, differential scanning calorimetry (DSC) analysis of<br />

nanosuspensions of the lipid lowering agent fenofibrate (kindly provided by Novartis<br />

Pharma AG) was conducted. The suspensions were produced by wet media milling using a<br />

modified planetary ball mill [3] (PM400, Retsch GmbH) or a stirred media mill (Dispermat<br />

SL 5 C nano, VMA Getzmann GmbH) and different polymers as stabilizers.<br />

Nanosuspensions were additionally analyzed using dynamic light scattering (DLS) and<br />

optical methods like transmission electron microscopy (TEM) and polarization microscopy.<br />

The evolution of onset and peak maximum temperature of PVA-stabilized fenofibrate<br />

nanosuspensions over grinding time was investigated via DSC indicating that both values<br />

decreased with longer grinding times [fig.1]. However, particle size determined via dynamic<br />

light scattering passed through a minimum and increased again with extended grinding<br />

times. TEM micrographs revealed that this phenomenon resulted from the formation of<br />

agglomerates containing extremly small particles after very long grinding times. Thus, the<br />

decrease of onset and peak maximum temperature was correlated with decreasing primary<br />

particle size.<br />

Furthermore, DSC was applied to differentiate between different types of instability of the<br />

fenofibrate nanosuspensions: particle growth or agglomeration of very small crystals,<br />

respectively. For this purpose, the onset and peak maximum temperatures of a HPMCstabilized<br />

nanosuspension at day 0 and after 1 week of storage under different conditions<br />

were compared. In combination with TEM investigations, the data clearly showed an<br />

increase in melting temperature for the samples with a pronounced crystal growth. DSC<br />

analysis can thus be a helpful approach not only for getting information about the real<br />

primary particle size but also for differentiating between different kinds of instabilities.<br />

124 Poster<br />

Temperature [ C]<br />

78<br />

77<br />

76<br />

75<br />

74<br />

73<br />

72<br />

71<br />

70<br />

69<br />

68<br />

Onset<br />

Peak<br />

z-avg<br />

4 6 8 10 20 40 60 100 200 400 800<br />

Grinding time [min]<br />

Fig. 1: Onset and peak maximum temperature determined via DSC and particle size (zaverage)<br />

determined via DLS of a PVA-stabilized fenofibrate nanosuspension at different<br />

points of the milling process<br />

References:<br />

1. Merisko-Liversidge, E., Liversidge, G.G.: Adv. Drug Deliv. Rev. 2011, 63(6): 427-440.<br />

2. Bunjes, H., Koch, M. H. J., Westesen, K.: Langmuir 2000, 16(12): 5234–5241.<br />

3. Juhnke, M., Berghausen, J., Timpe, C.: Chem. Eng. Technol. 2010, 33(9): 1412-1418.<br />

128<br />

Hydrogels for drug delivery: hydrolytically cleavable carbamate linkers for time<br />

controlled protein release<br />

Hammer N. 1,Brandl F.P. 1, Kirchhof S. 1 , Meßmann V. 1, Teßmar J.K.V. 1, Göpferich A. 1<br />

1 University of Regensburg, Department of Pharmaceutical Technology, <strong>Universität</strong>sstr.31<br />

Regensburg, 93053, Germany<br />

In the past decades, the significance of macromolecular drugs of biological origin has<br />

increased enormously. However, their application is still hampered by short biological halflives.<br />

Hydrogels are one possibility to protect these delicate molecules against<br />

physicochemical instability and proteolytic degradation. Furthermore, hydrogels are<br />

considered biocompatible due to their physicochemical similarity to native extracellular<br />

matrix. Despite these advantages, one frequently observed problem is the rapid release of<br />

incorporated proteins due to the high water content of the hydrogels. To overcome this<br />

problem, proteins can be reversibly attached to the hydrogel backbone by suitable linker<br />

molecules.<br />

We used non-degradable hydrogels to characterize carbamate-mediated protein release.<br />

The gels were formed from star-shaped poly(ethylene glycol) (PEG) alkyne and starshaped<br />

PEG azide; the cross-linking reaction was catalyzed by 0.5 µmol CuSO4 and 2.5<br />

µmol ascorbic acid. For the attachment of proteins to the hydrogel structure, we<br />

synthesized different star-shaped PEG linkers. Two to three of the four end-groups were<br />

modified with an alkynyl groups to allow cross-linking with star-shaped PEG azides. The<br />

remaining end groups were modified with hydrolytically cleavable carbamates for reversible<br />

protein binding.<br />

Gel properties were studied by oscillatory rheology. Swelling and stability of the gel<br />

cylinders in phosphate buffered saline at 37 °C were analyzed by weighing the cylinders at<br />

regular time points. We compared protein release from hydrogels with and without<br />

carbamate linkers. For protein binding, lysozyme was incubated with the carbamate linker<br />

for 2h at room temperature and mixed with additional polymer to form hydrogels. The gel<br />

cylinders were incubated in phosphate buffered saline at 37 °C. The amount of released<br />

protein was determined at regular time points by Bradford assay.<br />

Gelation started after about 16 min depending on the copper concentration.Oscillatory<br />

rheology showed a gel strength of 7600 Pa, which indicates an almost complete<br />

crosslinking reaction. Hydrogels with attached lysozyme still had a gel strength of 5400 Pa.<br />

We determined swelling ratios of about 1.2 (without lysozyme) and 1.28 (with lysozyme),<br />

respectively. The gel cylinders were stable for more than 2 weeks. Without binding,<br />

incorporated lysozyme was released within the first 24 h. Attachment of the protein to the<br />

hydrogel backbone decelerated the release significantly. After cleavage of the carbamate<br />

linker, 22 % of lysozyme was released within 14 days. The study is not completed yet;<br />

further time points will follow.<br />

lysozyme conc. [%]<br />

20%<br />

release of attached lysozyme<br />

0%<br />

0 5 10 15<br />

time [d]<br />

Altogether, covalent attachment of proteins to the gel structure by degradable carbamate<br />

linkers seems to be a promising strategy to control protein release.<br />

Acknowledgments: DFG, grant GO 565/16-1<br />

lysozyme conc. [%]<br />

50%<br />

1000<br />

900<br />

800<br />

700<br />

600<br />

500<br />

400<br />

300<br />

200<br />

100<br />

0<br />

Particle size [nm]<br />

release of lysoyzme without linker<br />

100%<br />

0%<br />

0 20<br />

time [h]<br />

40


129<br />

Production of Resveratrol Nanocrystals by a Novel Combinative Process (H 42<br />

Technology)<br />

Liu, T. 1; Müller, R. H. 1; Möschwitzer, J. P. 1 2<br />

1 Institute of Pharmacy, Dept. of Pharmaceutics, Biopharmaceutics and NutriCosmetics, Freie<br />

<strong>Universität</strong> Berlin, Kelchstr.31 Berlin, 12169,Germany<br />

2 Pharmaceutical Development, Abbott GmbH & Co. KG, Knollstr. 50 Ludwigshafen am Rhein,<br />

67061, Germany<br />

In order to improve the manufacturing efficiency of drug nanocrystal, a new combinative<br />

particle size reduction technology (H 42) has been developed recently. Spray-drying as a<br />

bottom-up step is applied to modify the properties (e.g. crystallinity and morphology) of the<br />

raw drug particles. The subsequent high pressure homogenization step is used to obtain<br />

the final nanosuspensions. According to the design of experiment (DoE) methodology,<br />

different compositions of resveratrol (API) and sodium cholate (surfactant) were dissolved<br />

in ethanol and the solutions were spray-dried using a mini spray dryer B-290 with inert loop<br />

B-295 (Büchi Labortechnik AG, Switzerland). The crystallinity of spray-dried powders was<br />

analyzed by differential scanning calorimetry (DSC, Mettler Toledo AG, Germany) and<br />

powder X-ray diffraction (PXRD) with Philips PW 1710 diffractometer control unit (Philips<br />

Industrial & Electro-Acoustic Systems Division, The Netherlands). High pressure<br />

homogenization (HPH) for 20 cycles at 1500 bar using a Micron Lab 40 homogenizer (APV<br />

Deutschland GmbH, Germany) was employed to produce nanosuspensions. Laser<br />

diffractometry (LD, Malvern Instruments, UK) and photon correlation spectroscopy (PCS,<br />

Malvern Instruments, UK) were applied to determine the particle size. The PXRD and DSC<br />

results showed that 6 DoE points of spray-dried powder resulted in amorphous drug, which<br />

evidently indicated a relationship between formulation of the feed and final API crystallinity.<br />

After only 1 homogenization cycle at 1500 bar, a particle size of around 200nm (d 0.5) was<br />

achieved by using modified API, which was comparable with the result of unmodified API<br />

after 20 cycles. The production time for the nanocrystals had been significantly reduced. On<br />

the other hand, the largest particle size (d 0.9) obtained from modified API (736 nm) was<br />

much smaller than the unmodified drug (2.2 µm) after 20 cycles. However, no direct<br />

correlation was found between the degree of crystallinity and the particle size of the drug<br />

nanocrystals after HPH. To sum up, due to the short production time and low energy input<br />

the new combination technology (H 42) was confirmed to be an effective method to<br />

produce resveratrol nanocrystals. Further more detailed study will be focused on the<br />

mechanism of this process and influencing factors of particle size reduction.<br />

130<br />

Differentiation of several identically prepared batches of tablets using near<br />

infrared spectroscopy<br />

Kaminski, D; Baumann, K<br />

1 Technische <strong>Universität</strong> Braunschweig, Institute for Medical and Pharmaceutical Chemistry,<br />

Beethovenstr. 55, 38106 Braunschweig, Germany<br />

Near infrared spectroscopy (NIR) is the first choice method in pharmaceutical industry for<br />

rapid inspection of raw materials. It is also suitable for online process control of the<br />

products as the NIR is a nondestructive analytical technique. Chemical composition as well<br />

as physical properties can be monitored. Even small variations, which are unavoidable<br />

during the production process, lead to significant changes in NIR spectra. This allows for a<br />

close monitoring of the physical properties, such as compaction pressure [1], humidity [2]<br />

and the homogeneity of active pharmaceutical ingredient (API) distribution [3] during the<br />

production process and the storage of tablets.<br />

This leads to the question whether it is possible to differentiate several batches of directtabletted<br />

ibuprofen with exactly the same manufacturing process by means of chemometric<br />

methods. Different methods of data pre-treatment are compared and an outlier detection is<br />

performed by principal component analysis (PCA). Those samples that show outlying<br />

spectra are scrutinized with respect to their content of the AIP which is independently<br />

determined by titration. The tabletting material and the production process are deliberately<br />

varied in order to study the sensitivity of the developed chemometric methods.<br />

The approach presented here attempts to separate the random error and deterministic<br />

variations in the spectral signal. The goal of this work is to define confidence limits based<br />

on a comprehensive characterization of all sources of spectral variations in order to detect<br />

tablets which are likely out of specification for further analysis by orthogonal analytical<br />

techniques.<br />

References:<br />

1. Kirsch JD, Drennen JK: J. Pharmaceut. Biomed. 1999, 19: 351-362<br />

2. Last IR, Prebble KA: J. Pharm. Biomed. Anal. 1993, 11: 1071-1076<br />

3. Plugge W, Van der Vlies C: J. Pharm. Biomed. Anal. 1993, 11: 435-442<br />

131<br />

Development of a medicated chewing gum containing riboflavin phosphate<br />

Wilde L. 1, Wollatz U. 1, Kordaß B. 2, Glöckl G. 1, Weitschies W. 1<br />

1Institute of Pharmacy, <strong>Ernst</strong>-<strong>Moritz</strong>-<strong>Arndt</strong>-University, Department of Biopharmaceutics &<br />

Pharmaceutical Technology, Center of Drug Absorption and Transport (C_DAT), Felix-Hausdorff-<br />

Straße 3, D-17487 <strong>Greifswald</strong>, Germany<br />

2<strong>Greifswald</strong> University Hospital, D-17475 <strong>Greifswald</strong>, Germany<br />

Medicated chewing gums are most often used for the treatment of travel-sickness or for the<br />

dehabituation of smokers. It was our idea to test, whether they might also be applicable to<br />

characterise the chewing motion of different patients by dentists. Therefore, a medicated<br />

chewing gum was developed, which contains an active agent, Riboflavin-5’-phosphate<br />

sodium, that can be dissolved by the saliva. Two different chewing gum matrices were<br />

investigated: Cafosa Gum S/A (Spain) and Gumlink A/S (Denmark). Both products are<br />

lipophilic mixtures consisting of elastomers for elasticity, resins for a cohesive body, waxes<br />

as softening agents and fats as plasticizers. Both gum bases can be compressed directly<br />

with the help of an eccentric tablet punch. For the preparation of the Gumlink gums, 19.9 g<br />

gum base and 100 mg lecithin were mixed in a cube blender for 10 minutes. After addition<br />

of 4 mg riboflavin phosphate and again blending for 10 minutes, the punches had to be<br />

lubricated with magnesium stearate to prevent sticking. The Cafosa gums were prepared in<br />

the same way, but with 19.8 g gum base, 200 mg lecithin and 20 mg riboflavin phosphate.<br />

In this way, we produced biconvex gums with a diameter of 12 mm and a weight of 500 mg.<br />

For the determination of content, a gum was given into an Erlenmeyer flask and 6 mL of<br />

heptane was added. After complete dissolution of the gum 20.0 mL purified water was<br />

added, the two phases were mixed intensely for 2 minutes and then separated by standing<br />

for 1 minute without stirring. For the Cafosa gums, two millilitres of the water phase were<br />

diluted and analysed at a wavelength of 267 and 600 nm (background correction) using a<br />

Cary 50 Scan (Varian). The water phase of the Gumlink gums was analysed without<br />

dilution. The determination of content show a content uniformity for the Gumlink chewing<br />

gums of 0.091 mg ± 0.0006 mg (n = 7) and for the Cafosa gums of 0.886 mg ± 0.070 mg (n<br />

= 7).<br />

For the chewing trials, the gums were chewed 5, 7 or 10 times (n = 6) by one volunteer,<br />

removed with a forceps and the saliva was rinsed off with purified water. Afterwards the<br />

determination of content followed as mentioned above.<br />

The results for the first chewing tests (figure 1) showed that chewing five times dissolved<br />

about 25 % of active agent, while seven chews increased this value to nearly 35 %. Ten<br />

chews released even more than 50 % of riboflavin phosphate out of the chewing gum<br />

matrix.<br />

Figure 1: Amount of riboflavin phosphate (Gumlink) after chewing 5, 7, 10 times; nominal<br />

quantity in each gum: 0.1 mg riboflavin phosphate (n = 6, mean ± SD).<br />

The Cafosa gum base was less suitable for spectrometric analysis because of the opacity<br />

of the solution even after filtration and centrifugation. That was the reason for higher<br />

absorption and thus incorrect content readings.<br />

We succeeded in producing uniform chewing gums with a small amount of hydrophilic<br />

active agent by direct compression and developed a method for the determination of<br />

content. We showed that the number of chews affects the dissolution of this agent, so that<br />

conclusions can be drawn from the chewing motion.<br />

Acknowledgments: The gum bases were a gift donated by Cafosa Gum S/A (Spain) and Gumlink A/S<br />

(Denmark). Financial support by the Federal Ministry of Education and Research (FKZ: 03IP612) is<br />

gratefully acknowledged.<br />

132<br />

Test purchase of slimming products containing unapproved active ingredients<br />

that may cause harmful side effects<br />

Parr MK1 2; Geyer H1; Paul M3, Smolka K4, Schänzer W1 1 Freie <strong>Universität</strong> Berlin, Pharmaceutical Chemistry, Königin-Luise-Str. 2+4, 14195 Berlin,<br />

Germany<br />

2 German Sport University Cologne, Biochemistry, Am Sportpark Müngersdorf, 50933 Cologne,<br />

Germany<br />

3 Stadt Köln, Gesundheitsamt, Neumarkt 15-21, 50667 Cologne, Germany<br />

4 Zollkriminalamt, Bergisch-Gladbacher-Str. 837, 51069 Cologne, Germany<br />

An increasing trend for attractive body appearance, stronger muscles and slender shape,<br />

coupled with a maximum of convenience is recognized in broad parts of society.<br />

Suggestions from advertisements and the easy access of image enhancing products,<br />

combined with the implication that such products cannot be harmful, make people believe<br />

that they can simply take “pills for everything” [1].<br />

Thus a huge market of dietary supplements offers numerous products containing a variety<br />

of unapproved and partially also harmful ingredients.<br />

Herein we present the results of products obtained by test purchases on the Internet. All<br />

products promised to cause extreme weight losses in a very short time period. The<br />

products were delivered by regular mail after credit card payment. No cautionary note was<br />

found on the label of the products.<br />

They were tested by GC-MS for their active ingredients. One of the products, named DNP<br />

Burn, brand name Body Advance Performance, was found to contain high amounts of 2,4dinitrophenol<br />

(Figure 1(1)). Identity was confirmed by comparison with authentic reference<br />

material. Semi-quantitation yielded an amount of about 200 mg. DNP uncouples the<br />

oxidative phosphorylation in the mitochondria resulting in a drastically increased metabolic<br />

rate. Several fatalities have been recorded after its intake [2].<br />

Other products were tested positive for the presence of methylhexanamine (Figure 1(2)).<br />

Due to its indirect sympathomimetic effects it was tested as locally administered<br />

vasocontrictor for the relief of hyperplasic nasal mucosa. Currently no pharmaceutical<br />

containing methylhexanamine is approved. Nowadays lots of preparations with<br />

methylhexanamine are marketed as supplements for slimming purposes. Increasing<br />

numbers of adverse analytical findings with methylhexanamine in doping control [3] indicate<br />

a growing market. Recently two fatal casualties in physical stress under the influence of<br />

methylhexanamine are investigated. This resulted in a recall of such products by the US<br />

FDA [4].<br />

Labelling of weight loss supplements uses several substitutes such as geranium root<br />

extract, geranium oil extract, Geranamin(e), 4-methyl-2-hexanamin(e), Forthan(e), 4methyl-2-hexylamin(e),<br />

2-hexanamine-4-methyl-, 2-methyl-4-methylhexan, Floradrene, 1,3dimethylamylamin(e),<br />

DMAA, 1,3-dimethylpentylamin(e), 2-amino-4-methylhexan(e), or<br />

pentylamine-1, 3-dimethyl- making the inference almost impossible for the consumers.<br />

Poster 125<br />

OH<br />

NO 2<br />

NO 2<br />

NH 2<br />

(1) (2)<br />

Figure 1: Chemical structure of ingredients of weight loss supplements<br />

(1) 2,4-dinitrophenol, DNP, (2) 4-methylhexan-2-amine<br />

References:<br />

1 Graham MR, Ryan P, Baker JS, Davies B, Thomas NE, Cooper SM, Evans P, Easmon S, Walker CJ,<br />

Cowan D, Kicman AT. Counterfeiting in performance- and image-enhancing drugs. Drug Test Anal,<br />

2009;1(3):135-42.


2 U.S. Dept Health & Human Services/Agency for Toxic Substances & Disease Registry. Hazardous<br />

Substances Data Bank HSDB. Toxicological Profile for Dinitrophenols, 1995, online at<br />

http://www.atsdr.cdc.gov/ToxProfiles/tp.asp?id=729&tid=132, accessed 16.05.<strong>2012</strong><br />

3 World Anti-Doping Agency. Adverse analytical findings reported by accredited laboratories, online at<br />

http://www.wada-ama.org/en/Science-Medicine/Anti-Doping-Laboratories/Laboratory-Statistics, accessed<br />

16.05.<strong>2012</strong><br />

4 DMAA News: FDA challenges marketing of DMAA products for lack of safety evidence, <strong>2012</strong>, online at<br />

http://fdarecalls.wordpress.com, accessed 16.05.<strong>2012</strong><br />

133<br />

Coating of collars via fluidised-bed technology: Influence of process<br />

parameters on coating homogeneity<br />

Wentzlaff, M. 1; Seidlitz, A. 1;Nagel, S. 1; Harder, C. 2; Schnittker, C. 3; Trip, E. 3; Bock, R. 3;<br />

Grabow, N. 4; Sternberg, K. 4;Weitschies, W. 1<br />

1Biopharmaceutics and Pharmaceutical Technology, University of <strong>Greifswald</strong>, Felix-Hausdorff-<br />

Str.3, 17487 <strong>Greifswald</strong>, Germany<br />

2Biotronik SE & Co.KG, Hartmannstr. 65, 91052 Erlangen, Germany<br />

3Biotronik SE & Co.KG, Woermannkehre 1, 12359 Berlin, Germany<br />

4Institute for Biomedical Engineering, University of Rostock, Friedrich-Barnewitz-Str.4, 18119<br />

Rostock, Germany<br />

Introduction<br />

Collars are polymeric tubes mounted on cardiac pacemaker leads containing an active<br />

substance to reduce inflammatory and proliferative reaction of myocardia after implantation<br />

[1,2]. Currently the active pharmaceutical ingredient is incorporated in collars during its<br />

manufacturing via injection moulding technology. However, this often results in high drug<br />

content variability. The fluidised-bed process is an established method used in galenics for<br />

coating of a large amount of small dosage forms with high uniformity.<br />

The purpose of this research was to adapt the process parameters of fluidized-bed process<br />

for coating of collars.<br />

Materials and Methods<br />

The fluidised-bed apparatus Mini-Glatt (Glatt GmbH, Germany) was used for coating of<br />

placebo-collars made of polyurethane. An aqueous dispersion of Eudragit RS ® 30D<br />

containing the water-soluble model substance fluorescein sodium was usedas the coating<br />

lacquer. Different coating processes parameters like spraying pressure, varied nozzle<br />

inserts and spray rate were investigated for optimisation coating homogeneity. Coating<br />

homogeneity was studied by scanning electron microscopy.<br />

Results<br />

By varying the spraying pressure it is possible to influence the surface morphology. While a<br />

spraying pressure of 0.7 bar created a rough surface morphology the surface became<br />

smoother when increasing the spraying pressure to 1.1 bar. A smoother surface was also<br />

created by reducing the nozzle orifice. It was shown that a nozzle orifice with a diameter of<br />

0.3 mm reduced the surface roughness compared to an orifice with a diameter of 0.5 mm.<br />

A reduction of the spray rate from 2.35 g/min to 0.77 g/min also resulted in a smoother<br />

surface.<br />

Conclusion<br />

Our findings provide a basis for coating of collars via fluidized-bed technology. By adapting<br />

the process parameters it was possible to produce coatings with desired morphologic<br />

characteristics.In order to obtain a sufficient coating thickness with a uniformdrug content,<br />

coating conditions have to be defined precisely. Therefore, further trials with optimization of<br />

coating dispersion concentrations and composition are necessary to ensure minimum<br />

mechanical stress acting on the collars and coatings.<br />

Acknowledgments: This project was partially funded by the Federal Ministry of Education and Research<br />

(BMBF) within the research project REMEDIS<br />

References:<br />

1. A. Wilson et al.: PACE1990, 13: 1876-1878.<br />

2. G. Brewer et al.: PACE 1988, 11: 1760-1769.<br />

134<br />

Evaluation of Interactions between Extractables of Elastomeric Closures and<br />

Protein Therapeutics<br />

Richter, C. 1 2, Lipke, U. 1, Zapf, T. 1, Lamprecht, A. 2, Lipperheide, C. 1<br />

1 Federal Institute for Drugs and Medical Devices, Kurt-Georg-Kiesinger-Allee 3, 53175 Bonn,<br />

Germany<br />

2 Institute of Pharmacy, University of Bonn, Gerhard-Domagk-Str.3, 53121 Bonn, Germany<br />

Parenteral liquid medicinal products are commonly offered in glass containers sealed with<br />

stoppers, which are made of elastomeric materials by vulcanization with the utilisation of<br />

various additives. These additives are predominantly small molecules. As they remain<br />

unlinked in the rubber material after the production process they are capable to migrate into<br />

the product formulation (“leachables”) during product shelf life and to react with the<br />

ingredients of the formulation. Particularly, medicinal products containing proteins as active<br />

ingredients are susceptible to react with substances released from the elastomeric material.<br />

In order to confirm compatibility between container and the product migration studies are<br />

requested prior to marketing authorisation. They should give evidence that quality, safety<br />

and efficacy of the protein containing medicinal product remains unaffected by leachables<br />

until end of shelf-life.<br />

The aim of the present study is to establish an appropriate method for evaluating the impact<br />

of leachables from elastomeric closures on protein stability without time-consuming<br />

migration studies.<br />

For the study various rubber stoppers were purchased from different vendors from the<br />

market. These rubber stoppers were extracted by applying worst case conditions<br />

(extraction with 2-propanol for 3 hours under reflux conditions) in order to receive the<br />

maximum amount of extractable substances (“extractables”) which is equivalent to the<br />

maximum amount of potential leachables. The extraction profiles of the different rubber<br />

stoppers were examined by RP-HPLC-DAD with a method, especially developed for the<br />

simultaneous analysis of polar and rather non-polar extractables. For further investigations,<br />

a chlorobutyl rubber stopper was chosen which showed the highest amount of extractables.<br />

Extractables stock solution was prepared as follows: 16 samples of the chlorobutyl rubber<br />

stopper were extracted in 200ml of 2-propanol for 6h under reflux conditions. After filtration<br />

(PE filter 0.45 µm) and solvent evaporation an oily residue was obtained. The residue was<br />

dissolved in 20ml of medium (20% (w/w) 1.2-propanediol and 0.5% (w/v) polysorbate 80 in<br />

water) for 1h in water bath (60-70°C). The resulting stock solution was filtrated and the<br />

amount of extractables was analyzed by RP-HPLC-DAD to confirm all extractables solved<br />

in the stock solution.<br />

The extractables stock solution was added to solutions of human serum albumin and<br />

different protein therapeutics and stored at 5°C. Samples were taken at several time points<br />

and examined by size-exclusion chromatography and capillary electrophoresis in order to<br />

detect physical and chemical alterations in protein structure as a result of the contact with<br />

the extractables.<br />

The proposed test approach turned out to be a suitable alternative to time consuming<br />

migration studies to investigate potential interactions between rubber stoppers and protein<br />

therapeutics.<br />

135<br />

In vitro dissolution testing of drug-eluting stents: Impact of implantation<br />

procedure<br />

Seidlitz, A. 1; Nagel, S. 1; Semmling, B. 1; Grabow, N. 2; Sternberg, K. 2; Weitschies, W. 1<br />

1 Institute of Pharmacy, EMA University of <strong>Greifswald</strong>, Felix-Hausdorff-Straße 3, 17487 <strong>Greifswald</strong>,<br />

Germany<br />

2 Institute for Biomedical Engineering, University of Rostock, Friedrich-Barnewitz-Straße 4, 18119<br />

Rostock, Germany<br />

Drug-eluting stents (DES) are drug/device combination products that are used to physically<br />

and pharmacologically prevent the renarrowing of a previously occluded natural passage.<br />

In the case of coronary DES the drug is intended to suppress intimal hyperplasia which<br />

often occurs after balloon angioplasty and stenting. Due to the placement of the DES in<br />

contact with the vessel wall, which is the site of action for the released drug, and the flowing<br />

blood, not only drug release but also its distribution between these compartments is of<br />

greatest importance. In vitro drug release and distribution can be examined using the<br />

vessel-simulating flow-through cell [1], a method adapted from the compendial flow-through<br />

cell (USP 4) for DES. In this setup the DES is implanted into a hydrogel compartment<br />

simulating the vessel wall and is perfused by PBS pH 7.4 representing the blood which is<br />

recirculated through the system (closed loop). For the studies presented here a<br />

haemostatic valve (Y-connector with rotating adapter and Tuohy-Borst valve) was added to<br />

the setup allowing for the placement of the DES in the gel via balloon catheter under flow<br />

conditions. Release and distribution of fluorescent model substances from coated stents<br />

(hydrophilic fluorescein sodium or hydrophobic triamterene embedded in a<br />

polymethacrylate blend, Eudragit RL/RS ratio 3/7) were examined using this setup. Stents<br />

were introduced via the haemostatic valve and the placement in the gel lumen under media<br />

flow (flow rate 35 mL/min, perfusion time prior to stent expansion 1 min) was compared to<br />

expansion into the air-filled gel lumen and subsequent perfusion. The experiments were<br />

terminated after different perfusion times up to 60 min and drug content in the perfusion<br />

media, in the hydrogel and the amount remaining in the stent coating were determined<br />

fluorimetrically. The results are presented in figure 1.<br />

Figure 1: Normalized amount of fluorescein sodium (A) and triamterene (B) in the<br />

compartment media, hydrogel or stent coating over time as a function of implantation<br />

procedure (without media present in the lumen or with media flowing through hydrogel<br />

during stent placement at 35 mL/min), n = 3 for each data point, mean ± SD.<br />

The initial distribution of released fluorescein sodium between the gel and media was<br />

greatly influenced by the implantation procedure. A much lower concentration in the<br />

hydrogel was observed when implanting under media perfusion of the lumen. This must be<br />

attributed to the fact that a certain amount of the fast released model substance is released<br />

exclusively into the media prior to contacting the hydrogel. When implanting without flow on<br />

the other hand, the initial release occurs solely into the gel for the first seconds. For<br />

triamterene, which is released much slower from the stent coating, this initial difference<br />

concerning the available acceptor compartments does not seem to have an impact on<br />

model substance release and distribution. Based on these results, the implantation<br />

procedure can be expected to have a great influence on release and distribution from fast<br />

releasing stent systems. The methods is also promising for other drug/device combination<br />

products applied in the vascular system such as drug-eluting balloons, which are intended<br />

to deliver their drug load to the vessel wall within very short contact times of approximately<br />

1 min.<br />

Acknowledgments: Financial support by the European Regional Development Fund (ERDF) and the<br />

European Social Fund (ESF) within the collaborative research between economy and science of the state<br />

Mecklenburg-Vorpommern is gratefully acknowledged. The authors thank Biotronik SE & Co. KG, B. Braun<br />

Melsungen AG and Rhoem GmbH for generously supplying raw materials.<br />

References:<br />

1. Seidlitz, A. et al.: J. Control. Release 2008, 130(1): 2-8.<br />

136<br />

A Novel Method to Characterize Screw-Elements in Pharmaceutical Twin-<br />

Screw-Extrusion<br />

Kiene, F. 1; Eeckman, L. 2; Thommes, M. 1<br />

1 Institut für Pharmazeutische Technologieund Biopharmazie, Heinrich-Heine-<strong>Universität</strong><br />

Düsseldorf, Germany<br />

2Department of PharmaceuticsLaboratory of Pharmaceutical Technology, Ghent University,<br />

Belgium<br />

Twin screw-extrusion is a commonly used technique in pharmaceutical applications. One of<br />

the relevant parameters in pharmaceutical extrusion is the screw configuration, because it<br />

can be adjusted to achieve desired product properties[1].The aim of this study was to<br />

characterize different types of conveying and kneading elements by their specific mechanic<br />

power and their transportation capacity, using high viscous silicon oil as model compound.<br />

For this, a part (4D) of the barrel of the lab scale extruder Micro 27GL-28D (Leistritz,<br />

Nuremberg, Germany) was separated with a front- and back-plate in order to investigate a<br />

smaller section of the barrel. A pressure sensor,I31-S-6-M-B01C-1-4-D (Gefran,<br />

126 Poster


Provagliod’Iseo Brescia, Italy),was attached to the front-plate. The torque was measured by<br />

a torque-gaugeT22/50 (HBM Messtechnik, Darmstadt, Germany)[2].The barrel load was set<br />

to 75% (Wacker ® AK 100000 Silicon oil, WackerChemie AG, Burghausen, Germany) and<br />

the rotation speed of the screws was varied from 50 to 200 rpm.<br />

Figure 1: Schematic drawing of the extruder setup; A: Back-plate with assembled screw; B:<br />

Isolated Extruder-Barrel<br />

Depending on the screw geometry different torqueand pressure were obtained and showed<br />

a linear correlation to rotation speed. This could be explained by the newtonian behaviour<br />

of silicon oil. Furthermore the specific mechanic power as a function of rotation speed (f)<br />

and torque (τ) for different loadings (m) was calculated as:<br />

. Equation 1<br />

Kneading blocks and conveying elements applied similar specific mechanic power to the<br />

material while differences within the subtypes can be observed. For example a high<br />

staggering angle of the kneading blocks led to lower values for the specific mechanic<br />

power. This could be explained by a higher transportation capacity of elements with lower<br />

staggering angle and can be determined by the pressure. Considering the conveying<br />

elements, a lower pitch resulted in a higher pressure andconsequently in a higher<br />

transportation capacity.<br />

In conclusion the setup was able to measure the specific mechanic power and the<br />

transportation capacity of different screw elements. Based on this, it will be possible to get<br />

deeper insight into the extrusion process.<br />

1. Djuric, D. and P. Kleinebudde, J Pharm Sci 2008, 97(11): 4934-4942.<br />

2. Köster, M. and M. Thommes, Chemical Engineering Journal 2010, 164(2-3): 371-375.<br />

137<br />

Rational Design of Nanoparticle Dissolution Testing for Parenterals: The<br />

Challenge of In Vitro Drug Release<br />

Janas,C. 1; Wacker, M. 1<br />

1Institute of Pharmaceutical Technology, Goethe-University, D-60438 Frankfurt, Germany<br />

Over the past decade significant research efforts have focused on a new generation of<br />

nanocarrier systems. These parenteral drug delivery devices are well-suited for the delivery<br />

of drugs with poor aqueous solubility, unfavourable body distribution and/or toxicity after<br />

intravenous injection. Due to the modified drug release of the active moieties from the<br />

nanocarrier matrix, physiology-based drug release models are needed in order to assure<br />

the quality and safety of these products. Moreover, they provide information about the drug<br />

release under in vivo conditions and are therefore an important tool in formulation<br />

development.<br />

There are several approaches which aimed to adopt standard methods from the European<br />

Pharmacopeia to the dissolution testing of parenteral nanoparticles and liposomes. Most of<br />

these studies showed a number of limitations due to the special requirements of the<br />

nanosized dosage form [1].<br />

In particular the isolation of the dissolved drug from particle bound drug molecules in the<br />

polymeric matrix is one major challenge in the development of an efficient dissolution<br />

method. Further aspects are the selection of dissolution media with regards to physiological<br />

parameters such as a solubility enhancement by human plasma, the colloid stability under<br />

in vivo conditions and the highly specific changes in surface properties by degradation and<br />

decoration with different plasma proteins. In order to ascertain anexperimental setup, that<br />

covers a wide range of different drug substances, the developed model has to be validated<br />

by using drugs with various physico-chemical properties.<br />

In our survey PLGA nanoparticles (NPs) loaded with model compounds intended for<br />

parenteral application are investigated in different release studies using dialysis-based<br />

methods. Buffers for dissolution media are prepared with various additives in order to not<br />

only bridge the gap between drug transport through the dialysis membrane but also to<br />

create conditions related to the in vivo situation.<br />

Initial experiments with the poorly soluble photosensitizer mTHPP were run with several<br />

membrane types to identify the transport rate of the API assuring that the dissolution of the<br />

drug is followed by direct passage through the membrane. The analysis shows acceptable<br />

transport rates in hydro-alcoholic solution and in complex with β-Methylcyclodextrin (0,1%<br />

in phosphate buffer saline, PBS). Based on these results the dissolution behaviour of<br />

mTHPP incorporated into PLGA NPs in PBS pH 7,2 is studied and compared with<br />

dissolution in buffer solutions composed of increasing amounts of foetal calf serum (FCS)<br />

and cyclodextrins (CD) respectively [2].<br />

Furthermore the colloid stability of investigated nanocarriers in phosphate buffer in the<br />

presence and absence of FCS is analysed as a function of time. There was no effect on<br />

particle size and polydispersity index (measured with photon correlation spectroscopy)<br />

during 24 h of incubation at 37°C which might predict no/delayed aggregation in vivo. The<br />

results of dissolution testing of mTHPP demonstrate a release of 100 % in both media (see<br />

Fig.1) and hence the sustained release of the NP formulation depending on media<br />

composition is investigatedin our experiments.<br />

Fig.1: Particle size, polydispersity index, and dissolution of mTHPPNP (n=3)<br />

Acknowledgments: Biolitec research GmbH, Dr. Arno Wiehe and Dr. Volker Albrecht<br />

References:<br />

1. D Souza S, DeLuca P,:Pharm. Res.2006, 23 (3): 460-474.<br />

2. DesRoches M-C et al.: J.Photochem.Photobiol. B. 2006, 85 (1): 56-64.<br />

138<br />

Chemically selective imaging of structured surfaces – a novel complementary<br />

analytical approach for pharmaceutical investigations<br />

Kann, B. 1;Gantzsch, S. 1; Lehr, C.M. 1 2 3; Schaefer, U. F. 1; Windbergs, M. 1 2 3<br />

1Saarland University, Department of Biopharmaceutics and Pharmaceutical Technology, Campus<br />

A4.1, 66123 Saarbruecken, Germany<br />

2PharmBioTec GmbH, Campus C2.2, 66123 Saarbruecken, Germany<br />

3Helmholtz-Institute for Pharmaceutical Research Saarland, Campus C2.3, 66123 Saarbruecken,<br />

Germany<br />

Confocal Raman microscopy is an upcoming analytical technique in the field of<br />

pharmaceutics. It is well suited for the contactless and chemically selective characterization<br />

of drug delivery systems and in vitro test systems.However, confocal Raman microscopy<br />

requires a smooth sample surface as spectra are exclusively recorded from the focal plane.<br />

As pharmaceutical samples often exhibit a structured surface such as lyophilisates and<br />

porous materials, confocal Raman analysis is impaired.<br />

We overcome the pitfalls of a confocal setup by combining Raman microscopy with optical<br />

profilometry as complementary analytical tools gaining a full-fledged characterization of the<br />

sample surface independent of its structure.<br />

To evaluate our novel approach, we analyze a protein-loaded lyophilisate. The therapeutic<br />

efficiency of proteins highly depends on their physical structure and molecular<br />

conformation, thus changes can have severe side effects on drug dosing for the patient.<br />

However, maintaining their therapeutically active conformation throughout manufacturing<br />

and within the final dosage form during shelf-life is still a major challenge. We freeze dry an<br />

aqueous solution containing a mixture of native and heat denaturated bovine serum<br />

albumin (BSA) for 48 h with a final drying step for 1 h gaining a solid lyophilisate. Optical<br />

profilometry of the surface is performed prior to Raman analysis (WITec alpha 300/500) as<br />

visualized in Figure 1a. As structural changes within the protein get along with a change in<br />

their Raman spectra, Raman microscopy is a valuable method to detect protein<br />

deconformation. The shift of the amide I peak in the Raman spectrum of the protein<br />

occurring between 1500-1800 cm-1 is used to discriminate between active and heat<br />

denaturated BSA within the dosage form. By overlaying the topography map with the<br />

respective Raman spectra at each measuring point we gain a spatially resolved false color<br />

image of the lyophilisate indicating each protein conformation in a different color.<br />

Furthermore, we investigate a Transwell-based permeation model. Polymeric membranes<br />

provide a suitable alternative to cell culture models overcoming the drawbacks of timeconsuming<br />

preparation and high expenses. For simulation of the intestinal mucosa, the<br />

apical side of the membrane is coated with lipids. Due to its non-destructive and chemically<br />

selective nature, Raman microscopy is a well suited technique for the analysis of chemical<br />

composition and three dimensional morphology of these systems.Raman spectra are<br />

recorded and subsequently converted into chemically resolved images (WITec alpha<br />

300/500). A comprehensive physical characterization of the transwell-based intestinal<br />

model is performed of the pure membrane as well as of the coated membrane before and<br />

after experiments. The pore morphology of the pure membrane is investigated with Raman<br />

spectroscopy in xz and xy-direction resulting in virtual cross-section images and three<br />

dimensional presentations (Figure 1b), respectively. After the coating, the pore morphology<br />

is verified again. Coating itself is achieved by centrifuging the lipid onto the membrane and<br />

a subsequent drying step. To monitor the coating formation on the membrane, a<br />

topography map is created after each coating step followed by subsequent Raman<br />

analysis.<br />

Figure 1. a) Topography map of a lyophilisate. b) Three dimensional image of the pore<br />

morphology of a polymeric membrane.<br />

We successfully introduce chemically selective imaging of structured surfaces for<br />

pharmaceutical applications by combining confocal Raman microscopy and optical<br />

profilometry as complementary analytical techniques.<br />

139<br />

Custom-tailoring microsystems for production intensification of lipid based<br />

colloidal drug delivery systems<br />

Finke, J. H. 1, Niemann, S. 1,Richter, C. 2, Gothsch, T. 3, Kwade, A. 3, Büttgenbach, S. 2, Müller-<br />

Goymann, C. C. 2<br />

1 Technische <strong>Universität</strong> Braunschweig, Institut für Pharmazeutische Technologie, Mendelssohnstr.<br />

1, 38106 Braunschweig, Germany<br />

2 Technische <strong>Universität</strong> Braunschweig, Institut für Mikrotechnik, 3 Technische <strong>Universität</strong><br />

Braunschweig, Institut für Partikeltechnik<br />

The manufacturing of colloidal drug delivery systems poses big challenges to the<br />

Poster 127


production process. It comprises a variety of different process steps, especially if solid lipid<br />

nanoparticles (SLN) are produced. Commonly, the whole process is broken down to<br />

separate process steps in different devices and performed batch-wise. In terms of process<br />

intensification, a change towards substantially smaller, cleaner and more efficient<br />

equipment or methods is required. Therefore, a continuously working microsystem (MS)<br />

approach was applied for SLN manufacturing. It comprises: (i) dispersion of solid active in<br />

the lipid phase, (ii) initial formation of lipid droplets in the aqueous phase, (iii)<br />

homogenization of these to colloidal, narrowly distributed droplets and (iv) crystallization of<br />

the latter in a defined way (Fig. 1). Additionally, this approach reduces educt batch sizes<br />

and product loss, thus facilitating its application as an instrument for formulation screening.<br />

In this study, the progress of process intensification is presented with regard to the<br />

homogenization step.<br />

MSs for high pressure homogenization were primarily fabricated in silicon/glass [1].<br />

Different geometric design approaches were tested and ranked regarding their droplet<br />

breakup efficiency in emulsions: straight channels < Z-channels < Y-channel ≈ orifice.<br />

Furthermore, various minor structural changes within the main geometric principles were<br />

analysed [2].<br />

According to silicone MS results, orifice channels fabricated in stainless steel [3] were<br />

investigated in detail as the most promising design principle. In general, steel MS supply<br />

lower fragility, higher process pressures and elevated temperatures, as necessary for SLN<br />

production. To elucidate the design parameters which influence the colloidal dispersion<br />

efficiency, orifice widths, number of consecutive orifices, and the distance between orifices<br />

were varied. It was shown that double orifices yield lower particle sizes compared to single<br />

orifices. Nevertheless, triple orifices do not provide further decrease in particle size, but<br />

rank between double and single orifices. These phenomena are attributed to the<br />

development of counter pressure towards the first orifice by a second one causing a more<br />

efficient breakup, on the one hand. On the other hand, the pressure drop is distributed over<br />

a higher number of orifices reducing the stress intensity at each orifice and, thus, the<br />

overall efficiency in case of three orifices. A longer distance between orifices appears more<br />

efficient in droplet breakup likely due to a reduction of counter pressure effects because a<br />

stable flow pattern is not able to recover due to the proximity of two orifices at a low<br />

distance (300 µm, equal to orifice length). For the combination of 80 and 120 µm wide<br />

orifices in one MS, it has been demonstrated more efficient if the smaller orifice is exposed<br />

to the product flow first, yielding the higher pressure drop over the size determining, first<br />

distinct energy input. These findings were confirmed by applying a pressure regulating<br />

valve, facilitating the arbitrary variation of counter pressure within the experimental setup. It<br />

was proved that a moderate counter pressure drastically decreases resulting particle sizes<br />

for a single orifice MS. In double orifices no further decrease in particle size was detected<br />

due to external counter pressure.<br />

Regarding the producibility of SLN, it was shown that particles can be produced in custom<br />

tailored MS with comparable or even better properties (particle size and distribution, melting<br />

behaviour) within one cycle instead of 20 cycles in a conventional homogenization setup.<br />

Conclusively, the effectiveness of MSs for high pressure homogenization in a continuously<br />

perfused microsystem for SLN production was systematically improved and crucial<br />

parameters were identified. This demonstrates a successful process intensification<br />

compared to conventional colloidal dispersion production.<br />

Fig. 1. Schematic representation of SLN production process in an microsystem approach<br />

Acknowledgments: The present study was performed in the context of the Research Group 856<br />

funded by the German Research Foundation (DFG). One of the authors (S. B.) gratefully<br />

acknowledges the financial support of the Volkswagen Foundation.<br />

References:<br />

1. Lesche, C., Büttgenbach, S.: MST Kongress 2009<br />

2. Finke, J. H. et al.: 8th CESPT, Austria, Graz 2010<br />

3. Richter, C., Krah, T., Büttgenbach, S.: Microsyst. Technol. <strong>2012</strong>: in press<br />

140<br />

Air drying of drug nanosuspensions as a simple small scale process for<br />

screening and feasibility studies<br />

Laabs, F.; Bunjes, H.<br />

Institute of Pharmaceutical Technology, Mendelssohnstraße 1, 38106 Braunschweig, Germany<br />

The conversion of poorly water soluble pharmaceutical drug substances into<br />

nanosuspensions is a common approach to overcome their poor dissolution and absorption<br />

properties. However, solid dosage forms are often preferred with regard to improved<br />

physicochemical stability and better patient convenience. Established unit operations like<br />

spray drying or freeze drying can be applied to convert nanosuspensions into solid dosage<br />

forms. However, preservation of the particle size during the drying process can be<br />

challenging [1]. Furthermore, the conversion step is often time consuming (e. g. freeze<br />

drying) and/or only a single sample can be processed at a time (e. g. spray drying). This is<br />

especially impedimental for screening studies where processing of many samples is<br />

required.<br />

In order to circumvent the above mentioned issues, a simple small scale air drying process<br />

was established. In this study, the applicability of the air drying process on screening and<br />

feasibility studies was investigated. In particular, the influence of the type and concentration<br />

of different carrier materials and the transferability of the air drying results onto those<br />

obtained by spray drying were assessed for two different API nanosuspensions.<br />

A naproxen nanosuspension (5 %) stabilized with Kollidon VA64 (2.5 %) and a griseofulvin<br />

nanosuspension (5 %) stabilized with Kollidon VA64 (2.5 %) and sodium dodecyl sulfate<br />

(0.25 %) were prepared by wet media milling for 4 h using a stirred media mill (Dispermat<br />

SL 5 C nano). Air drying of the nanosuspensions was performed on a polystyrene well plate<br />

or using an evaporator (EVA EC S) and samples were compared to those obtained by<br />

spray drying (Büchi B-191). After processing, the dried samples were resuspended and the<br />

particle size was determined via photon correlation spectroscopy.<br />

As exemplified in Figure 1 and 2, air drying of the nanosuspensions could successfully be<br />

performed and the results were similar to those obtained by spray drying. As already<br />

observed in previous spray drying studies [2], drying of the griseofulvin nanosuspension<br />

was shown to be less critical compared to the naproxen nanosuspension.<br />

In conclusion, the air drying process seems promising for screening and feasibility studies<br />

and can provide first estimates of results that can be achieved using other drying<br />

processes.<br />

Figure 1: Particle size values<br />

(PCS) of a naproxen<br />

nanosuspension before and after<br />

spray and air drying with Kollidon<br />

VA64 as carrier material at<br />

different carrier:API ratios<br />

Figure 2: Particle size values (PCS)<br />

of a griseofulvin nanosuspension<br />

before and after spray and air<br />

drying with different carrier<br />

materials at a carrier:API ratio of<br />

0.5:1<br />

Acknowledgments: We would like to thank A. Bitterlich and Prof. Dr. A. Kwade (TU Braunschweig, Institute<br />

for Particle Technology) for the cooperation concerning the development and the production of the<br />

nanosuspensions.<br />

References:<br />

Van Eerdenbrugh, B. et al.: Eur. J. Pharm. Sci. 2008, 35(1-2): 127-135<br />

Laabs, F., Komoß, C., Bunjes, H.: 8 th PBP World Meeting <strong>2012</strong>, Istanbul, Turkey<br />

141<br />

Tablet disintegration – design of an apparatus to measure the disintegration<br />

force and water uptake of tablets simultaneously<br />

Quodbach J1, Kleinebudde P1 1 Institute of Pharmaceutics and Biopharmaceutics, Heinrich-Heine-University, Universitaetsstrasse<br />

1, 40225 Duesseldorf, Germany<br />

The underlying mechanisms of action of tablet disintegrants are still mostly unknown. Even<br />

though tablet disintegration is often the first step in the bioavailability cascade, only few<br />

attempts have been made recently to understand how disintegration takes place or is<br />

influenced [1, 2, 3]. An apparatus was built to gain a more profound knowledge about the<br />

disintegration force kinetics, water uptake kinetics, and differences of tablets containing<br />

various super disintegrants. The measuring principle is based on a design from the<br />

University of Pavia [4] and was altered to allow for a higher sample throughput and more<br />

precise results. The experimental setup is schematically shown in Figure .<br />

One beaker is placed on a balance (Sartorius CP224s) another one is placed under the<br />

measuring probe of a Texture Analyser (TA.XTplus). The beakers are connected with a<br />

tube allowing water to flow from one beaker to the other. A tablet holder made of aluminium<br />

with 19 holes (r = 0.5 mm) is placed in the beaker on the Texture Analyser. An additional<br />

filter paper is placed on the tablet holder. The tablet itself is attached to the measuring<br />

probe with adhesive tape. Water is filled into the beakers until the tablet holder is in contact<br />

with the surface. Then the measuring probe with the tablets is slowly moved downwards<br />

until a trigger threshold is reached (0.25 N) when the tablet touches the tablet holder and<br />

the data acquisition begins. The Texture Analyser registers the force the tablet exerts onto<br />

the measuring probe, whereas the balance records the water uptake of the tablets as the<br />

negative mass change. Adhesive tape is fixed around the circumference of the tablet to<br />

ensure that the complete amount of force is transmitted to the Texture Analyser.<br />

As example, three measurements are presented in Figure . The measured tablets consist<br />

of 2 % Kyron T-314 (Corel Pharma Chem), a polacrilin potassium ion-exchange resin also<br />

used as super disintegrant, 97.5 % dibasic calcium phosphate (Chemische Fabrik<br />

Budenheim KG) and 0.5 % magnesium stearate (Merck AG). Initially, little water uptake<br />

results in a strong force development. Afterwards, the water uptake and force behave<br />

almost linear, which is followed by a decrease of absolute force. This can be due to a<br />

collapse of the internal structure after the maximum force is reached.<br />

Further improvements of the apparatus are still necessary to get a more precise resolution<br />

of the mass change, especially at the beginning. Yet, it proves to be an easy to use tool<br />

which allows insights into the relationship between water uptake and force development but<br />

also into kinetic processes during tablet disintegration.<br />

References:<br />

1. Desai P M, Liew C V, Heng P W S, J. Pharm. Sci. <strong>2012</strong>, 101(6): 2155-2164.<br />

2. Krausbauer E, et. al. J. Pharm. Sci. 2007, 97(1): 529-541.<br />

3. Massimo G, et. al.: Pharm. Dev. Technol. 2000, 5(2): 163-169.<br />

4. Caramella C: Pharm. Tech. Int. September 1990, (9):30-42.<br />

128 Poster<br />

z-average diame er (nm)<br />

1950<br />

1900<br />

1850<br />

250<br />

200<br />

150<br />

100<br />

50<br />

0<br />

be ore drying<br />

after spray drying<br />

after air drying (well plate)<br />

PdI<br />

10 1 5 1 2:1 1:1 0.5:1 0 1<br />

carrier:API ra io<br />

1.0<br />

0.9<br />

0.35<br />

0.30<br />

0.25<br />

0.20<br />

0.15<br />

0.10<br />

0.05<br />

0.00<br />

PdI<br />

z average d ameter (nm)<br />

300<br />

250<br />

200<br />

150<br />

100<br />

50<br />

0<br />

before drying<br />

after sp ay drying<br />

after air drying (well plate)<br />

after air drying (evaporator)<br />

PdI<br />

mann tol lac ose KVA 6 K30<br />

carr er material<br />

Figure 1: Experimental setup Figure 2: Water uptake versus force<br />

development<br />

0. 0<br />

0.35<br />

0.30<br />

0.25<br />

0.20<br />

0.15<br />

0.10<br />

0.05<br />

0.00<br />

PdI


142<br />

Selection of solvent type affects nanocrystal particle size during precipitation<br />

combined with high pressure homogenization process<br />

Sinha, B. 1; Möschwitzer, J. P. 2; Müller, R. H. 1<br />

1 Institute of Pharmacy, Dept. Pharmaceutics, Biopharmaceutics&Nutricosmetics, Freie University<br />

of Berlin, Berlin, 12169, Germany;<br />

2 Pharmaceutical Development, Abbott GmbH &Co.KG, Ludwigshafen, 67061, Germany<br />

Precipitation combined with high pressure homogenization could be a feasible technique for<br />

production of small size drug nanocrystal. Solvent type plays a critical role in nucleation,<br />

stabilization and particle growth during precipitation process. The aim of this study was to<br />

investigate how various solvents affect the final particle size of drug nanocrystal during this<br />

combination technique.<br />

Precipitation step was combined with high pressure homogenization process to prepare<br />

drug nanocrystal in this study. Ibuprofen (IBP, BASF, Germany) and resveratrol<br />

(E.denkFineChemie GmbH, Germany) were used as two model compounds. Solvent phase<br />

(S) was prepared by solubilizing drug in a water miscible solvent such as alcohol (Ethanol,<br />

Methanol, Isopropanol), acetone, tertrahydrofuran (THF), dimethylformamide (DMF),<br />

dimethylsulfoxide (DMSO) and ethylacetate. Aqueous solution of SDS (0.2%) was mixed<br />

with either 0.1% HPMC (viscosity 5 cps) or with 0.5% PVP (K40) to constitute the antisolvent<br />

phase (AS) for Ibuprofen and Resveratrol, respectively. Using a needle and a pump<br />

system, S phase was injected in the AS phase just before homogenization zone in<br />

Emulsiflex C5 (Avestin Europe GmbH, Mannheim, Germany) operated at 1000-1200 bar.<br />

Particle size of the nanocrystal was measured by laser diffractometry using Mastersizer<br />

2000 (Malvern Instruments, UK). Solvents were rank ordered based on the minimum<br />

particle size obtained using a solvent.<br />

Depending on the solvent type, particle size of drug nanocrystals was found to change.<br />

When isopropanol was used for making ibuprofen nanocrystal, the smallest particle size<br />

obtained was 306 nm [d(0.5)]. It was only possible to reduce particle size further to less<br />

than 200 nm when solvent was changed to acetone, methanol, ethanol, THF or DMSO.<br />

Mean particle size was increased when ethyl acetate and DMF was used. Therefore, the<br />

solvent preference rank order for preparing IBP nanocrystal was found to be<br />

THF~Ethanol>Methanol~DMSO>Acetone>IPA>Ethyl acetate. On the other hand, use of<br />

DMSO produced the smallest size resveratrol nanocrystal, i.e. 153 nm [LD (50%)]. Hence,<br />

the solvent preference rank order for resveratrol was found to be<br />

DMSO~Ethanol~IPA~Acetone>Methanol>DMF>THF. Other factors such as<br />

solvent/antisolvent ratio, drug concentration in the solvent phase and stabilizer composition<br />

was also found to be critical to control the particle size.<br />

Beside other factors, selection of the right solvent for precipitation study is necessary to<br />

achieve smaller particle size. Further optimization of other parameters leads to the smallest<br />

size drug nanocrystal in precipitation coupled with homogenization process.<br />

143<br />

Identification of factors affecting the sorption isotherms of spray dried binary<br />

carbohydrate mixtures containing maltodextrin and sucrose<br />

Tackenberg, M. W. 1 2; Horvat, M. 1;Thommes, M. 2; Kleinebudde, P. 2; Schuchmann, H. P. 1<br />

1 KIT – Karlsruhe Institute of Technology, Institute of Process Engineering in Life Sciences, Food<br />

Process Engineering (LVT), Kaiserstr. 12, 76131 Karlsruhe<br />

2 Heinrich-Heine-University, Institute of Pharmaceutics and Biopharmaceutics, Universitaetsstr. 1,<br />

40225 Duesseldorf, Germany<br />

Maltodextrins, amorphous starch degradation products, are widely used as excipients for<br />

solid drug formulations. The different types of Maltodextrins are characterized by their<br />

dextrose equivalent (DE), which is correlated to the average molecular size (DE 0: ~ starch;<br />

DE 100: glucose). The DE of common maltodextrins varies between 2 and 20.<br />

Furthermore, maltodextrins can be distinguished by their ratio of amylose to amylopectin<br />

content and carbohydrate composition with respect to the origin. These differences can be<br />

used to tailor-made new matrix formulations with outstanding properties for drug<br />

administration like inclusion compounds and storage stability. This study elucidates the<br />

effect of type and content of the maltodextrins on the sorption isotherms of spray dried<br />

binary carbohydrate matrices.<br />

Various maltodextrins (Glucidex ® with DE 6, Waxy Maize based; Glucidex ® with DE 12 and<br />

17, Maize based, Roquette Frères; Kleptose ® Linecaps, Pea based with DE 12 and 17,<br />

Roquette Frères) were coprocessed with sucrose (Compri-Zucker O, Südzucker AG) using<br />

spray drying (Büchi Minispray Dryer B-191, Büchi Labortechnik AG). Afterwards the<br />

sorption isotherms were determined at 25.0 °C (SPS 11, PMT Analytical GmbH & Co. KG).<br />

Therefore 0.5 g sample were dried to a water activity (aW) < 0.005, followed by a increasing<br />

of the water activity in steps of 0.1 to aW 0.9. The obtained experimental data points<br />

between aW 0.1 and 0.8 were fitted with the Guggenheim-Anderson-de Boer Equation<br />

(GAB) (1). For all isotherms R² > 0.99 was observed.<br />

m����K C a� m=<br />

(1−Ka� )(1 − K a� +CKa�) (1)<br />

• m = water content; aW = water activity<br />

• C = Guggenheim constant; K = correcting factor<br />

• mmono = water content of the saturated molecule monolayer of the surface<br />

The sucrose content increases the water adsorption at aW ≥ 0.4, as shown for the<br />

Glucidex ® DE 17 formulations (Fig. 1). This could be correlated to the highly hygroscopic<br />

amorphous sucrose molecules. At aW ≤ 0.3 the water uptake decreases with increasing<br />

sucrose content, which could be caused by the dominant properties of the high molecular<br />

weight fraction at low aW [1].<br />

Furthermore an effect of the DE to the water adsorption was found, as shown for the<br />

Glucidex ® DE 6 and 17 formulations with 20 % sucrose (Fig. 2). At low aW (< 0.5) the water<br />

uptake decreases with increasing DE. At aW > 0.5 it increases with increasing DE. This<br />

could be explained with the dominant properties of the high molecular weight compounds at<br />

low aW and the increasing lower molecular weight compounds (higher DE) at higher aW, too.<br />

Finally, an influence of the starch source (Maize or Pea) on the sorption isotherm of the<br />

maltodextrin sucrose mixtures was not detected, as shown for the Glucidex ® DE 17 and<br />

Kleptose ® Linecaps (KL) DE 17 (Fig. 2).<br />

Fig. 1: Influence of sucrose content on<br />

sorption behaviour.<br />

Fig. 2: Influence of DE and starch basis on<br />

sorption behaviour.<br />

From the results, it can be assumed that the average molecular size is the most important<br />

factor regarding the sorption behaviour of carbohydrates. Hence, this behaviour can be<br />

customized by a combination of maltodextrin and sucrose.<br />

Acknowledgments: Roquette Frères; Südzucker AG, Max-Buchner-Forschungsstiftung; AIF-ZIM Project<br />

KF2256805<br />

References:<br />

1. Radosta, S. et al.: Starch/Stärke 1989, 41(10): 395-401<br />

144<br />

Improved test on emulsifying properties of lanolin forcommunity pharmacies<br />

Koeck J; Kortmann C; Pein M; Breitkreutz J<br />

Institute of Pharmaceutics and Biopharmaceutics, Heinrich-Heine-University, Universitaetsstraße 1,<br />

40225 Duesseldorf, Germany<br />

Lanolin is a commonly used ointment base in dermal preparations. However, several<br />

problems are known like its sensitization potential, discussed controversially [1, 2],<br />

insufficient emulsification and stability properties [3]. As of its natural origin the emulsifying<br />

properties (EP) change depending on batch and age.<br />

For evaluating EP there have been two tests established: Addition of water and coldemulsifying<br />

according to German Pharmacopoeia [4] and titration, which was introduced by<br />

Reimannusing a solution containing Octoxynol-10 [5]. The first test only differentiates<br />

between “pass” and “fail”, the latter provides the possibility to check the EP more precisely<br />

and faster.As Octoxynol-10 is rarely used in community pharmacies, there is a need to<br />

improve the Reimann method.<br />

Lanolin of two qualities was used: Eucerinumanhydricum (EA) (Beiersdorf, D-<br />

Hamburg,Release date: 2004) and UnguentumAlcoholumLanae (UAL) (Caesar &Loretz, D-<br />

Hilden, Release date: 2010).<br />

Test samples were prepared of cold-emulsifying with stirring by hand 10 g of lanolin with<br />

15 g of water.<br />

Titration solutions (TS) were prepared, containing 4 g Methylene Blue, 1 g Octoxynol-10<br />

and water to 100 ml as prescribed by Reimann (TS1). The other test was performed<br />

exchanging Octoxynol-10 (HLB=13.6, Triton® X-100, Roth, D-Karlsruhe) byPolysorbate 80<br />

(HLB=15.0, Casear&Loretz, D-Hilden) (TS2).<br />

After titration in small steps (approximately 0.1 ml) test sample was stirred by hand until a<br />

blue coloured homogenous semisolid substance resulted. Endpoint was specified when TS<br />

was not further incorporable when stirring.<br />

It was possible to formulate a titration solution with Polysorbate 80 (TS2) which can be<br />

used with comparable results to Octoxynol-10 (Amount of TS to instability of aqueous<br />

lanolin (UAL): TS1 0.9 ml and TS2 1.0 ml). With this optimized method it is possible to use<br />

the proposed test on EP in community pharmacies with conventional and available<br />

excipients.<br />

Further, it was possible to differentiate between lanolin of different qualities and ages (EA<br />

and UAL) with TS1, according to its EP(Amount of TS1 to instability of EA: 0.4 ml and UAL:<br />

0.9 ml). Reimann specified an amount of at least 0.5 ml TS1 to use lanolin for dermal<br />

preparations.<br />

Concluding, it becomes feasible for community pharmacies to easily test EP of lanolin<br />

before preparing drug formulations. Other water-in-oil emulsions will be investigated by this<br />

test in future.<br />

References<br />

1. Kligman, A.M.: Contact Dermatitis.1998, 39(3): 103-107.<br />

2. Aberer, W.: Dermatologie in Beruf und Umwelt.2006, 54(4): 135-139.<br />

3. Eifler-Bollen, R.: Pharmazeutische Zeitung.2002, 147(17): 1898.<br />

4. Deutsches Arzneibuch: Wollwachsalkoholsalbe(Deutscher Apotheker Verlag).1999.<br />

5. Reimann, H.: PharmazeutischeZeitung.1993, 138(2): 114-115.<br />

145<br />

Tailor-made production of quercetin and rutin nanocrystals for investigating<br />

the correlation between the size and effect to cell<br />

Chen,R. 1, Wszelaki, N. 2, Melzig, M. F. 2, Müller, R. H. 1, Keck, C. M. 1 3<br />

1Institute of Pharmacy, Free University of Berlin, Kelchstrasse 31, 12169, Berlin<br />

2Institute of Pharmacy, Free University of Berlin, Königin-Luise-Strasse 2+4, 14195 Berlin<br />

3Applied Pharmacy Division,University of Applied Sciences Kaiserslautern, Campus Pirmasens,<br />

Carl-Schurz-Strasse 10-16, 66953 Pirmasens<br />

Flavonoids are believed having potential to protect neuro cell from neurogenerative<br />

damages caused by endogenous toxins. However, most flavonoids are poorly soluble,<br />

which lead to a low bioavailability and/or cell uptake when they are applied in aqueous<br />

solution form. To formulate the flavonoids into nanocrystals is an effective method to<br />

increase the solubility and to enable the cell uptake. As the solubility enhancing effect and<br />

the ability to be taken up by cells is size dependent, tailor-made production of nanocrystals<br />

is the first required step for investigating the correlation between size and effect to cell in<br />

future.<br />

In this study, quercetin and rutin nanocrystals were produced by high pressure<br />

homogenization (HPH) (LAB 40, APV Deutschland) in sterile condition. For the further<br />

investigation about the influence of size on the performance of nanocrystals in cell culture,<br />

a series of nanocrystals with different sizes were tailor-made with Poloxamer 188 as a nontoxic<br />

surfactant (0.2%, w/w) and glycerol as a osmotic adjuster (2.5%, w/w) by applying<br />

different HPH pressure (up to 1500 bar) and cycles (up to 20 cycles) and choosing different<br />

active concentrations (2%, 0.2%, 0.02%). Particles size was characterized by photon<br />

Poster 129


correlation spectroscopy (PCS), laser diffractometry (LD) and light microscopy. Zetapotential<br />

and short-term stability were examined. For correct size analysis the<br />

measurement method of PCS was optimized by choosing different measuring cuvettes and<br />

using different dilution media and sample concentrations. The ways to adding glycerol in<br />

nanosuspension was also compared.<br />

Via controlling production condition, a series of nanocrystals (from 172 nm to 1500 nm)<br />

were produced. Results showed that an increase in cycles of HPH could lead to a<br />

significant smaller size for quercetin but not for rutin. Lower active concentration in<br />

quercetin had no influence on size but in rutin it provided a smaller size. Zeta-potential in<br />

50μS/cm NaCl solution and original dispersion medium of quercetin nanocrystals were both<br />

above 30 mV, which indicated a better stability compared to rutin nanocrystals holding a<br />

zeta-potential of around 25 mV. The short-term stability also confirmed that the quercetin<br />

nanocrystals in 4 oC had a good stability. Through investigating PCS cuvettes and dilution<br />

methods, measurement condition was fixed as using a low volume cuvette, active powder<br />

saturated diluting medium and 0.02% measuring concentration. Adding glycerol in<br />

suspension before HPH gave a slightly smaller size compared to HPH without glycerol.<br />

However, for precise controlling the concentration of glycerol in final product, it was added<br />

in after HPH.<br />

In conclusion, different sizes of quercetin and rutin nanocrystals for cell culture test can be<br />

tailor-made by applying different pressures and cycles as well as using different active<br />

concentrations. The preliminary requirement for further cell culture research was achieved.<br />

Acknowledgements: The authors would like to thank China Scholarship Council for financial support.<br />

146<br />

Hydrogels for drug delivery: influence of macromer branching on<br />

characteristics of poly(ethylene glycol) based hydrogels<br />

Kirchhof, S. 1, Brandl F. 1, Hammer N. 1, Meßmann V. 1, Teßmar J. 1 and Göpferich A. 1<br />

1 University of Regensburg, Department of Pharmaceutical Technology, <strong>Universität</strong>sstr.31, 93053<br />

Regensburg, Germany<br />

Poly(ethylene glycol) (PEG) based hydrogels are interesting materials for a variety of<br />

biomedical applications. In the field of controlled drug delivery, the use of PEG gels has<br />

been promoted due to their excellent biocompatibility. Nevertheless, most of the existing<br />

cross-linking mechanisms are associated with disadvantages, such as the formation of<br />

potentially harmful radicals. A relatively unknown cross-linking mechanism for hydrogels is<br />

the Diels-Alder click reaction. Previously, we have shown the suitability of this reaction for<br />

the formation of suitable PEG based hydrogels. Here, we report the influences of the<br />

macromer branching factor on the characteristics of the resulting hydrogels.<br />

Gelation time / min<br />

250<br />

200<br />

150<br />

100<br />

50<br />

0<br />

4armPEG-hydrogel<br />

8armPEG-hydrogel<br />

5 10 15<br />

Total polymer concentration / w/v<br />

Figure 1: Gelation times of different PEGhydrogels;<br />

gelation time depends on<br />

branching factor and total polymer<br />

concentration<br />

Rea tive mass increase Mt/M0<br />

18<br />

16<br />

14<br />

12<br />

10<br />

8<br />

6<br />

4<br />

2<br />

15% 4armPEG-hydrogel<br />

5% 8armPEG-hydrogel<br />

0<br />

0 1 2 3 4 15 16 17 18 19 20 21<br />

Time / days<br />

Figure 2: Swelling behaviour of different<br />

PEG-hydrogels; 4armPEG-hydrogels were<br />

dissolved within 4 days, whereas<br />

8armPEG-hydrogels swelled on a lower<br />

level even after 3 weeks<br />

PEG with branching factors of four and eight (4armPEG10k-OH and 8armPEG10k-OH)<br />

were used as raw materials. 4armPEG10k-furan, 8armPEG10k-furan and 4armPEG10kmaleimide<br />

were synthesized according to previously established protocols [1]. For the<br />

synthesis of 8armPEG10k-maleimide, 8armPEG10k-amine and Nmethoxycarbonylmaleimide<br />

were reacted to yield the desired product. All compounds were<br />

characterized by 1H-NMR spectroscopy. For hydrogel preperation, defined amounts of<br />

4armPEG10k-maleimide and 4armPEG10k-furan or 8armPEG10k-maleimide and<br />

8armPEG10k-furan were separately dissolved in Millipore water. The two precursor<br />

solutions were mixed to initiate gel formation. Gelation kinetics and gel strength were<br />

studied by performing oscillatory shear experiments on a TA Instruments AR 2000<br />

rheometer with parallel plate geometry. For swelling studies, gel cylinders (Ø 7 mm) were<br />

incubated with 10 ml of phosphate buffer solution (pH 7.4).<br />

Rheological studies showed decreasing gelation times with increasing polymer<br />

concentrations (Fig. 1). This was observed for both degrees of branching. Moreover, the<br />

gelation time dramatically decreased by increasing the branching factor from four to eight.<br />

This is explained by the higher cross-linking density of the resulting hydrogels. 4armPEGhydrogels<br />

(15 % polymer) showed very high swelling and dissolved within one week (Fig.<br />

2). This could be explained by the reversibility of the Diels-Alder reaction. In contrast to<br />

that, 8armPEG-hydrogels showed almost no swelling even at lower polymer concentrations<br />

(5 % polymer). Until now, it is not completely clear why 8armPEG-hydrogels show this<br />

extraordinary stability. Either the retro-Diels-Alder reaction could be unfavorable or the gels<br />

could show self-healing properties, a phenomenon which has been described for other<br />

materials cross-linked by Diels-Alder reactions. Further studies are under way.<br />

In summary, our studies showed a clear influence of the branching factor on gelation time,<br />

gel strength, swelling and degradation. Given their favorable gelation time and long-term<br />

stability, 8armPEG-hydrogels seem to be a promising biomaterial for biomedical<br />

applications.<br />

Acknowledgments: This work has been supported by Deutsche Forschungsgemeinschaft (DFG), grant<br />

number GO565/16-1.<br />

References:<br />

1. Kirchhof, S. et al.: Application of the Diels-Alder Reaction as a New Cross-Linking Mechanism for<br />

Poly(ethylene glycol) Based Hydrogels, ACS Meeting Philadelphia, <strong>2012</strong><br />

147<br />

Chemical stability studies on the antileishmanial drug candidate KuRei300 in<br />

lipid based nanocarrier formulations as important prerequisite for in vivo<br />

experiments<br />

Kupetz, E. 1; Preu, L. 2; Bunjes, H. 1<br />

1 Institute of Pharmaceutical Technology, Technische <strong>Universität</strong> Braunschweig,<br />

Mendelssohnstraße 1, 38106 Braunschweig, Germany<br />

2 Institute of Medicinal and Pharmaceutical Chemistry, Technische <strong>Universität</strong> Braunschweig,<br />

Beethovenstraße 55, 38106 Braunschweig, Germany<br />

Visceral leishmaniasis is an infection caused by Leishmania donovani parasites. The WHO<br />

assigned it the status of a neglected tropical disease thus encouraging research for new<br />

active compounds. In a directed synthesis program the paullon chalcon derivative<br />

KuRei300 emerged as a promising drug candidate. It exhibited good antileishmanial activity<br />

in experiments with axenic amastigotes and human macrophages [1]. In vivo studies in<br />

mice are intended and a new parenteral formulation for KuRei300 was developed for this<br />

purpose [2]. This formulation is a mixed micellar solution based on phospholipids and a bile<br />

salt. It meets important quality criteria for parenteral preparations such as the use of<br />

approved excipients, physical stability and the possibility for sterile filtration. Another<br />

fundamental aspect that is, however, often overlooked is the possible decomposition of a<br />

formulated agent during storage. The present study therefore addressed the chemical<br />

stability of KuRei300 in liquid formulations.<br />

Since DMSO is a common solvent for in vivo / in vitro tests, a preliminary experiment was<br />

performed in which KuRei300 containing solutions were stored either under the influence or<br />

protection from light and monitored for signs of decay by HPLC. In the light exposed<br />

solution the original drug content declined rapidly and one predominant new peak<br />

appeared. The corresponding compound was identified as the Z-isomer of KuRei300 by 1H<br />

NMR spectroscopy. As a result of these findings all samples were stored protected from<br />

light in subsequent experiments. In a second set of experiments the stability of KuRei300 in<br />

the mixed micellar carrier solution was assessed under different conditions. To obtain the<br />

mixed micellar solution Lipoid S100 (13.4% w/w) was dispersed in an aqueous phase that<br />

contained sodium glycocholate (7.4% w/w overall) and was buffered to pH 7.4 to avoid<br />

phospholipid hydrolysis. This turbid phospholipid dispersion was then vigorously shaken<br />

until a translucent solution had formed. A KuRei300 solution in ethanol was pipetted into a<br />

vial and the ethanol was evaporated thus forming a substance coat on the walls of the vial.<br />

The micellar solution was then added and the vial placed on a rotator. During this passive<br />

loading procedure the major part of the substance film dissolved in the carrier. After<br />

loading, excess KuRei300 was filtered off and the loaded carrier was transferred into fresh<br />

vials which were crimped [2]. The samples were produced without and with 0.1% EDTA in<br />

the aqueous phase and were or were not flushed with nitrogen. At regular time intervals the<br />

chemical stability of KuRei300 was evaluated by HPLC. In the non-stabilized preparation<br />

(no N2, no EDTA) the substance underwent drastic and quick decomposition. The signals in<br />

the chromatogram differed, however, very much from those in the DMSO solution. Z-<br />

Kurei300 was not formed in significant amounts. Nitrogen flushing of the vial´s head space<br />

considerably slowed down the decay and after 10 weeks additional EDTA demonstrated a<br />

further benefit.<br />

In conclusion, the chemical stability of new drug candidates needs to be given the<br />

appropriate attention from the earliest stages of formulation. Photoisomerization and its<br />

impact on pharmacodynamics have been reported for anti-tumorigenic chalcones<br />

previously [3]. The relevance for the biological activity of KuRei300 has not been studied<br />

yet. Our study demonstrated that simple techniques such as light exclusion, storage under<br />

an inert gas and addition of a metal complexing agent (EDTA) strongly improved the<br />

chemical stability of the drug candidate. Such measures can prevent hard-to-interpret<br />

results from in vitro / in vivo experiments that may result if a potential chemical<br />

decomposition is disregarded.<br />

Acknowledgements: We would like to thank J. Ryczak (Institute of Medicinal and Pharmaceutical Chemistry,<br />

Beethovenstraße 55, 38106 Braunschweig) for the synthesis of KuRei300 and assistance in the<br />

interpretation of NMR spectra.<br />

References:<br />

1. Reichwald, C. et al.: J. Med. Chem. 2008, 51(3): 659-665<br />

2. Kupetz, E. Et al.: 8 th PBP World Meeting <strong>2012</strong>, Istanbul, Turkey<br />

3. Iwata, S. et al.: Biol. Pharm. Bull. 1997, 20(12): 1266-1270<br />

148<br />

Development of a microfluidic system for screening of colloidal drug<br />

formulations under flow conditions<br />

Pretor, S. 1; Finke, J.H. 1; Al-Halhouli, A.T. 2; Schmolke, H. 3; Busker, M. 4;Dietzel,<br />

A. 2;Büttgenbach, S. 2;Klages, C.-P. 3;Behrends, S. 4; Reichl, S. 1;Müller-Goymann, C.C. 1<br />

1Institut für Pharmazeutische Technologie, TU Braunschweig, Mendelssohnstraße 1, 38106<br />

Braunschweig, Deutschland<br />

2Institut für Mikrotechnik, TU Braunschweig,<br />

3Institut für Oberflächentechnik, TU Braunschweig;<br />

4Institut für Pharmakologie und Toxikologie, TU Braunschweig<br />

In recent years, research and drug development have focused on microfluidic systems<br />

because they offer miniaturization, parallelization and automation of screening processes<br />

[1]. In biotechnology, these systems are used as microbioreactors, whichhave already<br />

shown to be versatile tools for screening of microorganisms and optimal production<br />

parameters in small scale [2]. Additionally, cultivation of mammalian cell lines is<br />

alsopossible in microscale under flow conditions[3].<br />

Amicrofluidic systemconsisting of apolydimethylsiloxane (PDMS) fluidic channel bonded on<br />

a glass surface, a piezo driven micropump (Bartels Mikrosysteme) and a culture medium<br />

reservoir has been developed for the cultivation of mammalian cells. In a first series of<br />

experiments, immortalized human corneal epithelial cells (HCE-T) and primary human<br />

dermal fibroblasts (HDF)were cultivated in themicrochannel of the microfluidic system and<br />

examined according to their attachment and viability on surface-treated PDMS.<br />

In a second approach, the cellular uptake of fluorescence-labeled solid lipid nanoparticles<br />

was investigated. The aim was to checkthe suitability ofthe microfluidic system for tracing of<br />

nanoparticles, because it maysimulate physiological conditions by applying defined<br />

amounts of shear stress to the cells.<br />

Methods:<br />

Theattachment behavior of HDF on hydrophilizedPDMS plates with positively and<br />

negatively charged surfaces was estimated. The surface modification was done by layerby-layer<br />

deposition of polyelectrolytes.Positively charged surfaces were coated with<br />

130 Poster


fibronectin in addition. Cells were seeded on these plates and the ratio of attached cells<br />

was calculated by counting suspended cells after certain periods of time. Afterwards, the<br />

remaining cells were microscopically observed. Untreated PDMS served as negative<br />

control.<br />

For flow experiments, a constant medium flow of 0.2-1.0 ml/min was applied to the cells for<br />

24 hours. Cell staining was performed with Hoechst 33342, Annexin V and<br />

Ethidiumhomodimer to indicate viability, apoptosis and necrosis, respectively.<br />

Fluorescence labeled solid lipid nanoparticleswere prepared by using a microchannel<br />

emulsification method according to [4].HCE-T and HDF-cells were incubated with<br />

nanoparticles bothon a standard glass well plate and in the flow system.The<br />

attachednanoparticleswerevisualizedwith a confocal laser scanning microscope equipped<br />

with a 60x oil immersion objective.<br />

Results:<br />

Optimal cell growth wasachieved via treatment of PDMS surfaces with a positively charged<br />

coating and an additional fibronectindeposition.This coatingresulted in short attachment<br />

times after cell seeding tomicrochannel surfaces.<br />

Neither PDMS and glass, nor shear stress of 10 dyn/cm² affected growth of HCE-T and<br />

HDF cells in microchannels negatively with respect to cell viability.<br />

Solid lipid nanoparticles were able to transport the fluorescent dyecoumarin-6 as a model<br />

drug to epithelial cells.Its fluorescence intensity was investigated by CLSM both on special<br />

culture plates and in themicrochannels.<br />

Acknowledgement:The present study was performed in the context of the research group 856 mikroPART<br />

funded by the German Research Foundation (DFG). One of the authors (S. B.) gratefully acknowledges the<br />

financial support of the Volkswagen Foundation.<br />

References:<br />

[1] Dittrich, P.S.; Manz, A.: Nature20065: 210-218<br />

[2] Demming et al.: Biomicrofluidics2011 5: 014104<br />

[3] Kim et al.: Lab on a Chip20077: 681-694<br />

[4] Finke et al.:2nd Symposium on Phospholipids in Pharmaceutical Research Heidelberg2011(poster)<br />

149<br />

Optimizing the PCS measurement methods for nanoemulsions to see minimal<br />

changes in the z-average<br />

Harden, D. 1; Müller, R. H. 1; Keck, C. M. 1 2<br />

1 Institute of Pharmacy, Department of Pharmaceutics, Biopharmaceutics & NutriCosmetics, Freie<br />

<strong>Universität</strong> Berlin, Kelchstraße 31, 12169 Berlin, Germany<br />

2 Applied Pharmacy Division, University of Applied Sciences Kaiserslautern, Campus Pirmasens,<br />

Carl-Schurz-Straße 10-16, 66953 Pirmasens, Germany<br />

The first instrument for the photon correlation spectroscopy (PCS) was introduced to<br />

market in the end of 1970 (Malvern Instruments). Today the PCS is one of the most<br />

frequently used instruments to measure the mean particle size (z-average) of<br />

nanoparticles. PCS is known as a very accurate method with high reproducibility. However,<br />

reproducibility not only depends on the method, but can also be influenced by the sample.<br />

An example is particle interaction, i.e. repulsive or attractive forces, which can lead to faster<br />

or slower diffusion and thus to smaller or larger size results. Particle interaction depends on<br />

the particle concentration within the sample. Hence, if the concentration varies between<br />

different measurements of one sample, the size results might not be reproducible. The aim<br />

of this study was to investigate the reproducibility of PCS measurements (Zetasizer Nano<br />

ZS, Malvern Instruments, UK) using different concentrations of a nanoemulsion (Lipofundin<br />

MCT 20%, B.Braun Melsungen AG, Germany) for the measurements.<br />

Today, modern instruments can analyze samples containing high concentrations of<br />

nanoparticles. In this method (1), the distance of the laser beam in the measuring cell is<br />

automatically adjusted to the particle concentration of the sample. This is done to avoid<br />

multiple scattering effects and to obtain an optimal intensity of the scattered light on the<br />

detector. The results of the PCS measurements using this method did not lead to similar<br />

results. Results show that there was a variation of 8 nm for a particle concentration of<br />

0.005% to 1.0% (228 nm to 236 nm). The variation was random, i.e. with increasing the zaverage<br />

went up and down.<br />

In case of the older PCS instruments, there was no changing in the distance of the laser<br />

beam of measuring point to the cuvette wall, therefore for these measurements a lower<br />

particle concentration must be chosen/ selected, if not there would be multi scattering and<br />

this will cause a smaller z-average. This method (2) was transferred to the Zetasizer Nano<br />

ZS with a constant distance of 4.65 mm from the wall to the measurement point. The same<br />

particle concentrations as in method 1 (1% to 0.005%) were chosen. The results showed<br />

that there was an concentration dependent increase in the z-average from 184 nm (1%) to<br />

236 nm (0.005%), however, the differences in the size when analyzing low concentrations<br />

(0.05% to 0.005%) was only 2nm, whereas it was 8nm with method 1.<br />

The results indicate that the reproducibility the PCS analysis should be investigated for<br />

each sample, which could possess particle interactions. For this different concentrations<br />

should be analysed using either the automatic method 1 or method 2 with the fixed laser<br />

beam position. In case of the nanoemulsion it is best to analyze the samples with method 2,<br />

e.g. using lower concentrations of the nanoemulsion and no automatic adjustment to obtain<br />

reproducible size results. The concentration range for good reproducibility was found be<br />

0.02% to 0.005%.<br />

150<br />

Efficiency increase by transfer of a two-pot to a single-pot fluid bed<br />

granulation<br />

Germer, K. 1; Rockmann, Th. 2; Wolf, B. 1<br />

1 Anhalt University of Applied Sciences, Pharmaceutical Engineering, Strenzfelder Allee 28, 06406<br />

Bernburg, Germany<br />

2 Salutas Pharma Corp., Teamleader Galenics, Otto-von-Guericke Allee 1, 39197 Barleben,<br />

Germany<br />

Traditionally, pharmaceutical granulates are manufactured by spray agglomeration in a<br />

mixing apparatus. In the past, the wet granulates were dried by time consuming tray drying.<br />

Nowadays, fluid bed drying is the preferred procedure. The application of two consecutive<br />

steps in different plants is still connected with material loss and contamination risk for which<br />

reason both agglomeration and drying step should be performed in consecutive steps in<br />

one and the same apparatus [1, 2].<br />

The aim of the investigation was the transfer of a two-step process consisting of spray<br />

mixing agglomeration followed by fluid bed drying of a commercial drug product to a singlepot<br />

procedure in a batch laboratory fluid bed apparatus. Therefore, a fluid bed granulator<br />

and dryer GPCG 1.1 (Glatt Corp., Binzen, Germany) was used (Figure 1). To optimize the<br />

process parameters of fluid bed agglomeration, the spray rate was varied between 20 and<br />

40 g/min, all other process parameters were kept constant. Parameters of the laboratory<br />

fluid bed drying process referred to the production process. Dry granulates were<br />

compressed to tablets. Evaluation criteria were the maintenance of stable and reproducible<br />

fluid bed process, furthermore acceptable yields and high granulate and tablet quality<br />

according to market product specification and pharmacopoeia.<br />

Figure1: Fluid bed granulator and dryer GPCG 1.1<br />

Best products were received with high inlet air temperature (80°C), high spray rate (30-40<br />

g/min) and additional water spraying. Without interruption of the fluid bed process the wet<br />

agglomerates were dried. The whole granulation process was stable and reproducible.<br />

Product yield was above 90%. Granulate and tablet properties fulfilled the specified quality<br />

criteria. Particle size was slightly increased with increasing spray rate compared to the<br />

production process, but there was no significant influence neither on granulate nor on tablet<br />

properties. The two-step granulation process with two different plants was successful<br />

transferred to a single-pot fluid bed process saving process time and equipment and<br />

reducing material loss and contamination risk.<br />

References:<br />

1. K. Giry et al.: Chemical Engineerig and Processing 2009, 48: 1293-1301.<br />

2. J.Z. Gao et al.: International Journal of Pharmaceutics 2002, 237: 1-14.<br />

151<br />

Determination of residence time distribution in hot melt extrusion via image<br />

analysis<br />

Reitz E; Brock D; Thommes M<br />

Institute of Pharmaceutics and Biopharmaceutics, Heinrich-Heine University, Universitaetsstrasse<br />

1, 40225 Duesseldorf, Germany<br />

Hot melt extrusion is an important and modern technology in pharmaceutical applications<br />

and is gaining more and more interest because of its versatile capabilities. One<br />

fundamental parameter for process understanding and process control is the residence<br />

time (RT). This parameter plays an important role especially in hot melt extrusion<br />

processes. The knowledge of the RT is important in critical hot melt extrusion processes,<br />

e.g. in processing of heat sensitive products. For such processes a short RT is desired to<br />

prevent degradation. The knowledge of the RT is also important for those types of solid<br />

dispersions, where a drug shall be grinded or dispersed and for which a long RT is<br />

beneficial.<br />

The aim of this study was to develop a method for an on-line RT measurement. The idea<br />

was to determine the grey value of the extrudates by adding carbon black as tracer to white<br />

sugar alcohol extrudates (figure 1).<br />

Figure 1: colouring of the white sugar alcohol extrudate by carbon black<br />

0 200 00 600 800<br />

time [s]<br />

1000 1200 1 00<br />

Poster 131<br />

black<br />

grey<br />

white<br />

Xy itol 0. mm<br />

Xy itol 0. mm<br />

Manni ol 0. mm<br />

Manni ol 0. mm<br />

Figure 2: residence time distribution for xylitol and mannitol, measured by taking pictures of<br />

the extrudates leaving the die (n=2)


Hot melt extrusion (Mikro 27GL-28D, Leistritz, Nuremberg) was performed with xylitol and<br />

mannitol (n=2). Carbon black in a defined amount (0.5 g carbon black as bolus by 40 g/min<br />

powder feed rate) was added and a series of pictures (Nikon D300, Nikon Corp. Japan)<br />

was taken from the extrudate in the die region in two-second intervals. A software algorithm<br />

(Matlab version R2009a, Mathworks) was developed which enabled the calculation of a<br />

grey value of a defined piece of the extrudate over the time resulting in a residence time<br />

distribution (RTD).<br />

It was possible to calculate the grey value for the extrudate emerging from the die plate at<br />

each time point, describing the load of the tracer. An adjustment of the grey values to a<br />

black and white standard was necessary to exclude any light effects affecting the colour of<br />

the extrudate on the photograph. Therefore on each photograph black and white standards<br />

were recorded as well. The camera software cut a piece of the extrudate as well as of the<br />

black and white standard out of the photographs for each time point. For these three parts<br />

of the photographs, the grey values were calculated and the value for the extrudates was<br />

adjusted by the standard values.<br />

Table 1: important parameters of the RTD<br />

onset [s] midpoint [s] offset [s]<br />

xylitol 317 435<br />

690<br />

328 430<br />

706<br />

mannitol 233 324<br />

523<br />

249 325<br />

525<br />

With this calculation, the RT as well as the RTD were determined (figure 2). This process<br />

was reproducible for both carriers used in this study (table 1). Due to the high sampling rate<br />

of the photos, the residence time could be determined very precisely.<br />

In conclusion, a tool for residence time measurement was developed, which helps to<br />

understand and to control hot melt extrusion processes.<br />

152<br />

Systematic studies of process parameters for wet granulation in a high-shear<br />

mixer<br />

Hoffmann, B. 1; Mosig, J. 1; Kleinebudde, P. 1<br />

1 Institute of Pharmaceutics and Biopharmaceutics, Heinrich-Heine-University, Universitaetsstr.1,<br />

40225 Duesseldorf, Germany<br />

The aim of this study was to investigate the influence of different process parameters on<br />

wet granulation processes in a high-shear mixer. The statistical evaluation was arranged by<br />

the programme MODDE 9.0 (Umetrics, Sweden). The DOE had a full factorial design of 4<br />

factors with an interaction model and three repetitions of the center point.<br />

Lactose monohydrate (Granulac 200, Meggle, Germany) was used as excipient. For the<br />

granulation polyvinylpyrrolidone (Kollidon 30, BASF, Germany) was added as solid binder<br />

and water as granulation liquid.<br />

It was granulated in a 10 L-bowl of a high-shear mixer (Diosna, Germany). Water addition<br />

time was kept constant at 200 s, the chopper was started every minute for 10 s. Varied<br />

parameters of the process were the impeller speed (imp; 150 rpm to 300 rpm), the liquid<br />

content (liq; 10 % to 12 % (w/w) related to solid mass), the binder content (bin; 4 % to 6 %<br />

(w/w) of total solid mass) and the wet massing time (wmt; 0 to 300 s). Response<br />

parameters were the particle size distribution (x50, x25 and x75), the interquartil range, the<br />

yield (particles < 1250 µm) and the particle size distribution of the yield (x50 of the yield<br />

y50). Further response parameters were bulk density, tapped density and the Hausner factor<br />

(according to Ph. Eur. 2.9.34).<br />

The mean particle size (x50) ranged between 150 µm – 1700 µm. Although most granules<br />

had an x50 about 200 - 300 µm, there were coarse granules obtained in every batch. This<br />

became apparent in the results of the yield, as 94 % in the best and only 10 % in the worst<br />

case could be obtained. Decreasing binder content had the biggest influence on the<br />

increase of the yield. Contrary to expectations a lower influence of the impeller speed in<br />

contrast to the other factors was observed. After backward regression, it became only<br />

significant for the x50 and the y50. This should be exemplified by the coefficient plot of the y50<br />

(fig. 1). The effect of the liquid content and the wet massing time was much higher (imp<br />

106.5 µm; liq 236.7 µm; bin 177.6 µm; wmt 240.2 µm). In contrast to literature1. a<br />

significant effect of the impeller speed on the interquartil range as parameter for the width<br />

of particle size distribution could not be found. For densities and the Hausner factor no<br />

parameter seemed to have a significant effect.<br />

In conclusion, liquid content, wet massing time and binder content had the greatest effect<br />

on analyzed granule properties. Further studies shall investigate if these findings can be<br />

transferred to high-shear granulation processes with further filler-binder combinations.<br />

[µm]<br />

140<br />

120<br />

100<br />

80<br />

60<br />

40<br />

20<br />

0<br />

-20<br />

-40<br />

imp<br />

iq<br />

Figure 1 – Coefficient plot of y50 (R² Adj. = 0.936; Q² = 0.828)<br />

References:<br />

1. Oulahna, D. et al., Powder Technology 2003 130: 238-246.<br />

bin<br />

wmt<br />

imp*bin<br />

imp*wmt<br />

liq*wmt<br />

bin*wmt<br />

153<br />

Fluorescent Nanoparticles capped with Octreotide for Targeting of<br />

Somatostatin Receptors<br />

Abd-Ellatif. A, Hennig. R, Pollinger. K, Tessmar. J .and Goepferich. A<br />

Department of Pharmaceutical Technology, University of Regensburg, Regensburg, Germany<br />

Somatostatin receptor subtype 2 (SSTR2) is widely expressed in normal and tumor cells,<br />

which is clinically used for diagnostic purposes [1].Targeting therapeutic nanoparticles to<br />

the SSTR2 using somatostatin analogue octreotide (OCT), would be a promising approach<br />

for certain cancer cells, since this would reduce side effects and increase drug delivery<br />

efficacy to the target site, highly over-expressing the respective receptors.<br />

To attach OCT to nanoparticles, it was succinylated with succinic anhydride at low pH<br />

selectively at its N-terminus [2]. Succinylated OCT was then conjugated using a watersoluble<br />

carbodiimide (EDC) and N-hydroxy succinimide to photo-stable fluorescent<br />

Quantum dot nanoparticles. These have an emission wavelength of 655 nm and are<br />

functionalized with amine-derivatized polyethylene glycol (PEG).These are known to have a<br />

very low non-specific binding to cellular membranes.<br />

Succinylated OCT, was characterized by mass spectrometry and the formulated<br />

nanoparticles were characterized by size exclusion chromatography (SEC), fluorescence<br />

spectrometry, 1H-NMR,FTIR spectroscopy and for the resulting zeta-potential using<br />

dynamic light scattering. Finally, cellular uptake was studied for SSTR2-positive pancreatic<br />

carcinoid tumor cells (BON1) using confocal microscopy and flow cytometry.<br />

Mass spectrometry indicated mono-succinylation of OCT by succinic anhydride in aqueous<br />

media, as the result showed an increase in molecular mass of about 100 Da. A slight peak<br />

shift was observed in SEC for OCT-Qdots compared to the native nanoparticles. In addition<br />

a slight red shift was observed in the fluorescence emission spectra of Qdots after<br />

conjugation to OCT. The zeta-potentials showed an inversion to positive charge of Qdots<br />

after the conjugation to OCT. FTIR spectroscopy and 1H-NMR confirmed the attachment of<br />

OCT to Qdots, which served as proof for the binding of the peptide to Qdots.<br />

When BON1 cells incubated with the ligand modified particles, confocal microscopy and<br />

flow cytometry showed high binding to BON1cells. Flow cytometry revealed the<br />

displacement of Qdots-OCT, when high concentrations of free OCT were used to displace<br />

it.<br />

We concluded that nanoparticles modified with OCT may serve as promising tool to target<br />

different cells that over-express SSTR2 receptors.<br />

This work has been financially supported by a scholarsh p from the Arab Republic of Egypt for Ahmed<br />

Abdellatif.<br />

References:<br />

1. Reubi, J.C. et al., J. Clin. Endocrinol. Metab. 2010, 95(5): 2343-50.<br />

2. Kinstler, O.B. et al., Pharm Res. 1996, 13(7): 996-1002.<br />

3. Updegrove, T.B. et al., RNA 2011, 17(3): 489-500.<br />

4. Na, D.H. et al., AAPS Pharm. Sci.Tech. 2003, 4(4): E72.<br />

154<br />

An accelerated release method for fast assessment of real time release from<br />

an intravaginal ring<br />

Externbrink A1, Klein S1 1 <strong>Ernst</strong>-<strong>Moritz</strong>-<strong>Arndt</strong> University <strong>Greifswald</strong>, Department of Pharmacy, Institute of Biopharmacy and<br />

Pharmaceutical Technology, C_DAT, Felix-Hausdorff.Str. 3, 17489 <strong>Greifswald</strong><br />

Purpose<br />

Intravaginal rings (IVR’s) are usually designed to deliver the active drug over long periods<br />

of time. Accelerated drug release testing would allow for fast assessment of real time<br />

release during product development and quality control. Therefore, the objective of the<br />

present study was to develop a discriminative accelerated release method for an IVR and<br />

to establish a relationship between real time and accelerated release profiles.<br />

Methods<br />

NuvaRing ®, an approved hormonal contraceptive IVR which releases 120 µg etonogestrel<br />

(ENG) and 15 µg ethinylestradiol per day over 21 days, was selected as a model<br />

formulation. Release experiments were performed with ring segments and the release from<br />

one IVR was calculated with respect to the mass ratio. Prior to the release experiments,<br />

rings were cut into 10 segments which were endcapped with acrylate glue to prevent<br />

uncontrolled diffusion which would affect the overall release. USP apparatus 7 (400-DS)<br />

was used to determine the daily etonogestrel release. 10 mL of a vaginal fluid simulant<br />

were used as the release medium. The first set of experiments was performed at 37 °C<br />

(real time conditions) and automated sampling as well as media replacement was<br />

performed every 12 hours. Subsequent experiments were performed at elevated<br />

temperatures, i.e. 44, 50 and 55 °C (accelerated conditions). Sampling and media<br />

replacement was performed in appropriate intervals.<br />

Results<br />

For all temperatures the zero order release constants were calculated from the slopes of<br />

the cumulative release versus time graph and increased with increase in temperature. The<br />

elevated temperature release constants were plotted according to the arrhenius equation<br />

and a linear correlation between rate constants and temperature was found. The predicted<br />

rate constant at 37 °C was in agreement with the experimental value.<br />

The real time and accelerated release profiles at 50 °C were directly compared by plotting<br />

the cumulative real time release versus the accelerated release and a strong linear<br />

correlation was obtained with a correlation coefficient of 0.999. Additionally, the cumulative<br />

real time and accelerated release profiles at 50 °C were plotted on the same axis after time<br />

scaling. Applying the scaling factor which was determined as the ratio between the release<br />

constants at elevated temperature and 37 °C real time and accelerated release profiles at<br />

50 °C were superimposable.<br />

Reservoir type IVR’s like NuvaRing ® typically show an initial burst release phase followed<br />

by near zero order release kinetics with a slight decrease in daily drug release over time.<br />

Drug release is usually expressed as daily release versus time. In order to be able to<br />

monitor the changes in the daily real time release profile over time with accelerated test<br />

conditions the sampling frequency was adjusted to reflect daily real time release. The<br />

required sampling interval at 50 °C was determined from the ratio between zero order<br />

release constants at 37 °C and 50 °C. A good agreement between the daily real time<br />

release profiles and the elevated temperature release profiles with adjusted sampling<br />

intervals was observed.<br />

Conclusion<br />

An accelerated release method that is predictive of real time release was developed.<br />

132 Poster


Temperature appears to be a valid parameter to accelerate ENG release from NuvaRing ®.<br />

An Arrhenius relationship between temperature and release constants indicates that the<br />

integrity of the dosage form and the release medium is maintained under accelerated<br />

conditions. Comparison of real time and elevated temperature release profiles at 50 °C<br />

revealed a strong correlation between real time and accelerated release. In order to assess<br />

the changes in the daily release profiles over time the sampling intervals at elevated<br />

temperatures can be adjusted to reflect daily real time release. With the proposed method<br />

and a temperature of 50 °C the overall duration of drug release from Nuvaring segments<br />

could be reduced from 21 to less than five days.<br />

155<br />

Sartan-modified quantum dots targeting the Angiotensin II receptor type 1<br />

Hennig R1; Pollinger K1; Wenzel R1; Breunig M1; Tessmar J1; Göpferich A1 1 University of Regensburg, Dept. of Pharmaceutical Technology, <strong>Universität</strong>sstr. 31, 93053<br />

Regensburg, Germany<br />

The angiotensin II type 1 (AT1) receptor, which belongs to the class of G protein-coupled<br />

receptors, is a pharmacological target of great interest. Although it is expressed<br />

ubiquitously throughout the body, it is known that this receptor is overexpressed in several<br />

tissues due to pathological conditions and, therefore, represents a promising target for<br />

nanoparticulate drug delivery [1, 2].<br />

To address nanoparticles to AT1 receptors we labelled commercially available fluorescent<br />

quantum dots (Qdots) with a metabolite of losartan, EXP3174, which is largely responsible<br />

for the actions of losartan in vivo [3]. In addition to the increased receptor affinity, EXP3174<br />

has a carboxylic function that allows relatively easy conjugation to nanoparticles.<br />

Furthermore, amides of that carboxylic group are known to retain a high receptor blocking<br />

efficacy in similar derivatives [4]. First, quantitative real-time PCR showed that human<br />

adrenal corticocarcinoma NCI-H295R cells, which served as receptor-positive model cells,<br />

highly expressed the AT1 receptor. Flow cytometry (FACS) experiments with these ontarget<br />

and HeLa cells as off-target cells revealed a strong binding of EXP3174-modified<br />

Qdots to AT1-positive but not to AT1-negative HeLa cells. To further investigate the<br />

distribution pattern of the modified Qdots we performed binding experiments with a confocal<br />

laser scanning microscope (CLSM). The EXP3174-Qdots showed a membrane-associated<br />

staining of the NCI-H295R cells, indicating a binding to the cellular surface. This was<br />

expected for antagonist-modified nanoparticles which do not provoke receptor<br />

internalization upon ligand binding. In competitive displacement experiments a great excess<br />

of free ligand was needed to displace the EXP3174-modified Qdots from the cells, which is<br />

characteristic for nanoparticles that exhibit multivalent binding [5].<br />

In conclusion, we could show that a non-peptide antagonist for the AT1 receptor can be<br />

bound to quantum dots in order to target AT1-positive cells in a multivalent manner.<br />

Although the chemical character is significantly altered by conjugation, a sufficient receptor<br />

affinity remains. This allows for potential targeting of AT1 receptor overexpressing sites<br />

within the body.<br />

Acknowledgements: This work was supported by the Deutsche Forschungsgemeinschaft: DFG grant No<br />

GO565/17-1.<br />

References:<br />

1. Dvir, T. et al.: Nano Lett. 2011, 11(10): 4411–4414.<br />

2. Suzuki, K. et al.: Am. J. Pathol. 2007, 170(6): 1841-1853.<br />

3. Fabiani, M. et al.: Am. J. Hypertens. 2000, 13(9): 1005–1013.<br />

4. Santella, J. B. et al.: Bioorg. Med. Chem. Lett. 1994, 4(18): 2235–2240.<br />

5. Montet, X. et al.: J. Med. Chem. 2006, 49(20): 6087-6093.<br />

156<br />

Biphasic drug release from coated pellets<br />

Priese, F., Wolf, B.<br />

Anhalt University of Applied Sciences, Pharmaceutical Engineering, Neues Labor, Strenzfelder<br />

Allee 28, D-06406 Bernburg, Germany<br />

Pellets used for multiparticulate drug delivery systems may contain the API (active<br />

pharmaceutical ingredient) incorporated into the pellet core or as a coating layer on the<br />

pellet surface. Rapid release occurs when the API is readily soluble. With the help of further<br />

excipients the API release can be varied in a very broad range. Retarded release is<br />

achieved with functional polymers showing swelling capacity but no solubility under<br />

physiological conditions [1]. Additional to the advantage of homogeneous drug release of<br />

multiparticulate systems a biphasic release may be achieved by incorporation of a fast as<br />

well as a slow or retarded released dosage of the API. The therapeutic effect is reached<br />

immediately after drug administration and the patient is supplied with drug over a long time<br />

period by the slow released maintenance dosage.<br />

Inert microcrystalline cellulose pellets (Cellets ® 200, IPC GmbH, Dresden, Germany) were<br />

coated with the model API sodium benzoate as maintenance dosage (Figure) in a first step.<br />

The coating liquid consists of sodium benzoate and furthermore of polyvinylpyrrolidone for<br />

film stability increase and talcum to prevent the pellets from sticking during the coating<br />

process. For retarded release of that dosage the pellets were coated with Surelease ®<br />

dispersion (ethylcellulose, Colorcon Corp.) in a second step. At least the pellets were<br />

coated with the initial sodium benzoate dosage. The coating was performed in a batch<br />

laboratory fluid bed apparatus with Wurster technique (GPCG 1.1, Glatt Corp., Binzen,<br />

Germany). The weight ratio of initial to maintenance dosage was varied (1:2.2 and 1:6).<br />

Evaluation criteria were the coating process stability, the product yield and the pellet<br />

quality. Mean pellet size, sphericity, bulk density and drug release were measured.<br />

Figure: Scheme of threefold coated pellets<br />

The fluid bed coating processes were stable. At the end of the first and the second coating<br />

step the coating liquids were changed so that the process was performed without<br />

interruption. Yield was above 90%, significant material loss at the inner wall of the chamber<br />

or at the partition was never observed. The pellets were received as particles with smooth<br />

surface and sphericity of 0.95-0.96 indicating homogeneous growth of the several coating<br />

layers. The initial sodium benzoate dosage from the external layer was released<br />

immediately after 1 minute. The maintenance dosage was retarded released due to<br />

ethylcellulose film swelling (90% release after 17-25 minutes). In comparison, the release of<br />

sodium benzoate coated pellets without polymer film occurs completely within 1 minute due<br />

to readily solubility and high dissolution rate. The retarding effect of Surelease is improved<br />

with increasing layer thickness. By variation of the ratio of initial to maintenance dosage<br />

and polymer layer thickness the release kinetics can be varied in a wide range. The final<br />

pellets are filled into hard gelatin capsules or compressed into tablets. The influence of the<br />

compression process on the integrity of the pellets and the maintenance of the release<br />

kinetics is under investigation.<br />

Acknowledgement: The technical assistance of the student Markus Seifert in the experimental trials is<br />

grateful acknowledged.<br />

References:<br />

[1] Siepmann, F., Siepmann J., Walther, M., MacRae, R.J., Bodmeier, R.: Polymer blends for controlled<br />

release coatings, Journal of Controlled Release 2008, 125: 1–15.<br />

157<br />

Influence of fluid bed rotor pelletization parameters on pellet properties<br />

Wolf B, Ren X<br />

Anhalt University of Applied Sciences, Pharmaceutical Engineering, Neues Labor, Strenzfelder<br />

Allee 28, D-06406 Bernburg, Germany<br />

API (active pharmaceutical ingredient) loaded pellets are used for the preparation of<br />

multiparticulate drug delivery systems. Actual market products are pellet filled hard gelatine<br />

capsules, in some cases pellets are compressed into tablets. The API may be incorporated<br />

into the pellet or sprayed onto the surface of inert pellets forming an external layer. Enteric<br />

resistance or controlled release can be achieved in both cases with further coating layers.<br />

Pellets are predominantly manufactured by extrusion/spheronization, but fluid bed rotor<br />

agglomeration [1, 2] is of increasing interest because of the advantage of a single pot<br />

process, i.e. mixing of the initial solid components, agglomeration and drying are performed<br />

in one and the same apparatus.<br />

Preliminary studies were performed with a simple binary mixture of microcrystalline<br />

cellulose and lactose monohydrate 1:1 to evaluate the possible process parameter range.<br />

In a second trial, the highest possible content of ternary model substances (benzocaine,<br />

calcium hydrogen phosphate, talcum and maize starch) in the mixture was investigated.<br />

Evaluation criteria were process stability and pellet quality. Finally, by use of a 23 design of<br />

experiments (DoE) the influence of the process parameters spray rate, process air<br />

temperature and rotor disc rate and the influence of the model substances (content 10% in<br />

the formulation) on the pellet formation process and on the mean particle size d50 was<br />

investigated. The pellet manufacturing process was performed in a laboratory fluid bed<br />

granulator and dryer (GPCG 1.1 with rotor disc equipment, Glatt Corp., Binzen, Germany).<br />

Purified water was used as granulation liquid. At the end of the agglomeration process the<br />

product was dried to residual moisture below 5%.<br />

With the binary mixture, the pellet fluid bed manufacturing was possible in a broad range of<br />

spray rate, process air temperature and rotor disk rate. The pellets were received in<br />

satisfactory spherical shape. The maximum content of maize starch was 40%, higher<br />

amount led to overwetting and formation of inacceptable large agglomerates. The content<br />

of insoluble and not swellable calcium hydrogen phosphate was limited to 40%. Above this<br />

value agglomeration did not occur anymore, the fluid bed process broke down and the<br />

product precipitated onto the wall and at the spray nozzle. This was also the case with<br />

more than 15% of coarse-grained benzocaine as well as fine-grained talcum what is<br />

sometimes used in low concentration as anti-sticking agent in coating processes.<br />

Factor and interaction Effect Tendency<br />

Factor Pellet size<br />

A = Spray rate high positive A ↑ ↑<br />

B = Process air temperature high negative B ↑ ↓<br />

C = Rotor disk rate weak negative C ↑ ↓<br />

Interaction A/B and A/C negative A/B, A/C ↑ ↓<br />

Interaction B/C and A/B/C weak positive B/C, A/B/C ↑<br />

Table: Evaluation of DoE, factors, effects and tendency on pellet size<br />

Low and high level of the process parameters were spray rate 15/25 g/min, process air<br />

temperature 50/70°C and rotor disc rate 450/750 rpm. The evaluation of the DoE<br />

experiments is summarized in the table. The influence of spray rate on particle size is high<br />

positive and that of process air temperature high negative as expected. The rotor disk rate<br />

does not significant effect the particle size. Effects by interaction are proved but not<br />

significant. Fine-grained and insoluble calcium hydrogen phosphate does not influence the<br />

particle size. Vice versa coarse-grained benzocaine gives high effect. The flow properties of<br />

the products are influenced by the particle size and the shape. Angle of slope is diminished<br />

with increasing pellet size indicating improved flow properties. On the other hand flow time<br />

is raised with increasing particle size. Large pellets deviate from spherical shape more than<br />

smaller ones deteriorating the flow property. Fluid bed rotor granulation may be used for<br />

pellet manufacturing in a wide range of process parameters but the drug substance<br />

properties i.e. initial particle size, solubility and hydrophilic-lipophilic properties should be<br />

taken into consideration.<br />

References:<br />

[1] Parikh, D.M., Mogavero, M.: Batch fluid bed granulation. In: Parikh, D.M. (editor): Handbook of<br />

pharmaceutical granulation technology (Taylor&Francis) 2005.<br />

[2] Kristensen, J.,Hansen, V.W.: AAPS PharmSciTech 2006,7(1): E153-E162<br />

Poster 133


158<br />

Stability and cellular uptake of a model vaccine containing Resovist ® as<br />

marker for MRI tracking<br />

K. Thom1, K. Schulz1, K. Aurich2, G. Glöckl1, J.-P. Kühn3 and W. Weitschies1 1Institute of Pharmacy, EMA University of <strong>Greifswald</strong>, <strong>Greifswald</strong>, D-17489, Germany<br />

2Institute of Immunology and Transfusion Medicine, EMA University of <strong>Greifswald</strong>, <strong>Greifswald</strong>, D-<br />

17489, Germany<br />

3Institute of Diagnostic Radiology and Neuroradiology, EMA University of <strong>Greifswald</strong>, <strong>Greifswald</strong>,<br />

D-17489, Germany<br />

Aluminum containing adjuvants in vaccines applied for immunopotentiation have been<br />

considered as save and efficacious for decades. However, their mode of action could not<br />

be ascertained in detail to date. Tracking the adjuvant after injection using magnetic<br />

resonance imaging (MRI) may be a new method to investigate the fate of aluminum<br />

hydroxide (AH) adjuvant in vivo. For this purpose the MRI contrast agent Resovist ® (Bayer<br />

Schering Pharma AG) containing ferucarbotran nanoparticles, was combined with<br />

Aluminum hydroxide Gel (Sigma). Prior to MRI analysis this combination was investigated<br />

concerning stability and cellular interaction.<br />

Opposite surface charges (zeta potential Resovist ® -38 mV, Aluminum hydroxide Gel<br />

+13 mV) lead to electrostatic attraction and hence to adsorption by the adjuvant within<br />

30 minutes. Tracking the AH adjuvant requires a strong adsorption of the ferucarbotran<br />

nanoparticles even under in vivo conditions. Therefore the degree of adsorption was<br />

determined in simulated interstitial fluid (sIF) [1] and in plasma for a period of 24 h. At<br />

different timepoints unbound iron content was determined using atomic absorption<br />

spectrometry (AAS).<br />

Investigating the uptake of ferucarbotran and ferucarbotran adsorbed to AH adjuvant into<br />

peripheral mononuclear blood cells (PBMC) gave an indication of the influence of the<br />

nanoparticles on the adjuvants behavior after injection of the model vaccine. For isolating<br />

PMBC a density gradient centrifugation was used (OptiPrep ®, Sigma). Cells were incubated<br />

either with Resovist ®, Aluminum hydroxide Gel or the combination thereof at 37 °C and<br />

300 rpm. One fraction was incubated without additives as negative control. For cell dilution<br />

different media were used (RPMI with 10 %, 50 % or without fetal calf serum = FCS, 100 %<br />

FCS or sIF) to investigate media influence on the uptake into cells. After one hour cells with<br />

adsorbed or incorporated nanoparticles were separated utilizing their magnetic behaviour.<br />

Subsequently magnetic and non-magnetic cells were counted. The iron content of each<br />

separated fraction with sIF as dilution medium was determined using AAS.<br />

In our experiments sIF eluted twice as much iron than plasma during the first hour but after<br />

6 and 24 hours iron content in all samples was nearly the same. Agitation of the samples<br />

during incubation did not have any effect on the elution. Iron amounts in the supernatant<br />

below 7 % of the input indicate high adsorption strength after injection of the model vaccine.<br />

Resovist ® was used in this study to ensure visibility of the AH adjuvant for MRI. The<br />

contrast agent leads to signal loss on MR images if it is accumulated in tissues. Incubation<br />

with ferucarbotran particles led to an iron uptake into cells which allowed separation in a<br />

magnetic field. During this process the dilution medium had an obvious influence. With<br />

increasing FCS concentration in the medium the number of magnetic cells decreased due<br />

to the rising amount of adherent proteins which block the cell surface. The amount of<br />

ferucarbotran which was taken up within sIF medium was similar to the values for RPMI<br />

with 50 % FCS. This correlates with the protein content of 2.5 % in sIF which corresponds<br />

to 5 % proteins in serum respectively. When the Resovist ® particles were adsorbed to AH<br />

adjuvant prior to incubation the magnetic cell fraction in each dilution medium exceeded the<br />

one reached after pure Resovist ® treatment. The enhancement of the iron uptake may<br />

facilitate signal loss resulting in visibility.<br />

The non-magnetic fraction of Resovist ® treated cells contained more iron than the magnetic<br />

fraction propably due to an excess of unbound Resovist ®. Cells incubated with Resovist ®<br />

adsorbed to AH gave opposite results. Here the AH-ferucarbotran aggregates showed<br />

magnetic behaviour by themselves and thus increased the iron content of the magnetic cell<br />

fraction.<br />

In conclusion, adsorption of Resovist ® is most probably stable enough for tracking AH<br />

adjuvant in vivo. AH adjuvant enhanced the iron uptake into PMBC. This might facilitate the<br />

detection on MR images<br />

References:<br />

1. Wolff, L. et al.: Vaccine 2009, 27: 1834-1840.<br />

159<br />

Effects of non-thermal plasma on HaCaT keratinocytes is not mediated by<br />

ozone<br />

Blackert S1, Haertel B1, Talmann L2, Oehmigen K2, von Woedtke T2, Lindequist U1 1 Institute of Pharmacy, University of <strong>Greifswald</strong>, D-17489, Germany<br />

2 Leibniz-Institute for Plasma Science and Technology e.V. (INP), D-17489 <strong>Greifswald</strong>, Germany<br />

Non-thermal atmospheric-pressure plasma has been shown to influence as first target the<br />

cell membrane with its embedded proteins. Such cell surface molecules as integrins,<br />

cadherins or the epidermal growth factor receptor (EGFR) are of importance in wound<br />

healing and also for development of cancer metastasis. Cold plasma comprises of<br />

electrons, positive or negative ions, free radicals [e.g. reactive oxygen species (ROS),<br />

ozone] and other excited atoms and molecules. Further, plasma has an optical emission in<br />

the UV-region, especially UVB. Each of these components can affect the cells during<br />

treatment.<br />

Since it is known that during plasma treatment ozone is generated, we wanted to know as<br />

to whether this ozone concentration affects HaCaT keratinocytes.<br />

Adherent HaCaT keratinocytes were treated with plasma by a surface dielectric barrier<br />

discharge in air and argon (1 to 5 min) or with ozone (5 min). Ozone was generated by an<br />

Ozonisator and monitored by FT-IR (100, 400, 900 and 1800 ppm). Intracellular ROS,<br />

apoptosis (annexin V/propidium iodide staining) and cell surface molecules (α2-, α3-, α4-,<br />

α6-, αV-, β1-integrin, E-cadherin, EGFR) were analyzed by flow cytometry 24h after<br />

treatment.<br />

Besides a reduction of cell viability significant intracellular changes were observed. DBD/air<br />

plasma for 5 min caused an increased expression of α2- and β1-integrin whereas Ecadherin<br />

and EGFR expression was not influenced. The effects of DBD/argon plasma were<br />

less pronounced. Apoptosis was only increased by DBD/air plasma (5 min) although the<br />

proportion of apoptotic cells was rather low. Intracellular ROS detected by the fluorescent<br />

dye CM-H2DC-FDA increased from 6.6 ± 1.1% (untreated control cells) to 18.0 ± 3.2% (5<br />

min DBD/air) and 10.9 ± 1.3% (5 min DBD/argon). A concentration of about 100 ppm<br />

ozone was measured above the solid phase during a 5 min DBD/air treatment cycle which<br />

had no influence on integrin, E-cadherin or EGFR expression. 1800 ppm ozone caused an<br />

increase of α2- and β1-integrin whereas all other molecules measured were not affected.<br />

Taken together, the extent of effects depended on the nature of plasma (air vs. argon) and<br />

the exposure time of cells to the plasma. Short (≤ 1 min) treatment cycles did neither<br />

change cell surface protein expression nor induced apoptosis or intracellular ROS. The<br />

lowest concentration of ozone tested (100 ppm = about 200 mg/m 3), which is 1000 fold<br />

higher than the MAC had marginal effect on viability and no influence on integrin<br />

expression of HaCaT keratinocytes. The effects of plasma on cell membrane proteins<br />

observed are rather attributed to induction of intracellular ROS than to generation of ozone.<br />

* Work supported by the German Federal Ministry of Education and Research within the joint research<br />

project Campus PlasmaMed” (grant no. 13N11182)<br />

160<br />

Evaluation of German and International drug interaction databases<br />

Pauly A. 1; Wolf C. 1; Busse M. 2; Strauss A-C. 2; Krebs S. 2; Dörje, F. 2; Leuner, K. 1<br />

1 Professur für Molekulare und Klinische Pharmazie, Cauerstraße 4, 91058 Erlangen, Germany<br />

2 Apotheke des <strong>Universität</strong>sklinikums Erlangen, Palmsanlage 3, 91054 Erlangen, Germany<br />

Background<br />

Drug interactions are a common problem in drug therapy, especially if polypharmacy is<br />

needed [1]. There are a number of databases available helping to identify clinically relevant<br />

drug interactions. They vary in scope, accuracy, comprehensiveness, user-friendliness and<br />

cost. So far, there does not exist an evaluation which includes German databases.<br />

Therefore, we compared seven databases in German and English to determine the best<br />

database for daily clinical practice.<br />

Methods<br />

We identified more than 100 drug interaction pairs out of our daily routine and literature<br />

research. These were first validated using three reference databases (Stockley’s, DrugDex,<br />

iFacts). Out of the 100 selected pairs 50 were classified as clinically relevant and 50 as<br />

clinically non-relevant.<br />

A predefined scoring system was used to assess the scope, accuracy and<br />

comprehensiveness of the selected drug interaction databases. ABDA-database, MediQ,<br />

Pharmavista and MMI Pharmindex were selected as German drug interactions<br />

programmes. Lexi-Interact, Epocrates and drugs.com represented the international test<br />

databases.<br />

Results<br />

With its reliable monographs and high accuracy, Lexi-Interact scored 509 of 600 possible<br />

points and ranked first in our evaluation. Lexi-Interact was the most comprehensive<br />

database (180 of 200 possible points) with detailed monographs containing not only<br />

mechanism, severity and management of the drug interaction, but also onset, level of<br />

evidence and discussion of evidence, and literature references. These monograph<br />

components were not provided in all databases.<br />

The free of charge Swiss database Pharmavista (495 of 600 points) provided detailed and<br />

accurate monographs, but only recognized Swiss trade names. The German ABDA-<br />

Database ranked third in the overall score (453 of 600 possible points), because it omitted<br />

the level of evidence and discussion of evidence. The ABDA-Database scored best in<br />

accuracy with 335 out of 400 possible points.<br />

MMI Pharmindex (441 of 600 possible points), MediQ (413 of 600), drugs.com (382 of 600)<br />

and Epocrates (357 of 600) ranked fourth to seventh. These databases failed to distinguish<br />

between clinically relevant and clinically non-relevant interactions in a higher percentage,<br />

revealed more problems identifying clinically non-relevant interactions and suffer from<br />

limitations in the quality and comprehensiveness of their monographs.<br />

Discussion<br />

For the daily clinical routine, an easy to use and accurate system is needed, which<br />

identifies drug interactions without the tendency of overalerting. Whereas most databases<br />

were self-explanatory, MMI pharmindex was more complex to use. Drugs.com displayed<br />

the highest rate of excessive alerts. As frequent alerts of clinically irrelevant drug<br />

interactions lead users to ignore the warnings [2], this might be a major caveat to use this<br />

database in the daily clinical routine.<br />

Conclusion<br />

When used appropriately, drug interaction database can help to identify clinically relevant<br />

drug interactions and thereby increase patient safety. However, we should be aware of the<br />

limitations of drug interaction databases. In our evaluation Lexi-Interact proved to be the<br />

most reliable and comprehensive source of drug interaction information.<br />

References:<br />

1. Storka, A. et al, Wiener Medizinische Wochenschrift, 2009. 159(17-18): 462-469.<br />

2. Payne, T.H., et al., Proceedings / AMIA . Annual Symposium. 2002: 602-606.<br />

161<br />

Applicability of different omeprazole formulations via enteral feeding tubes<br />

Hanke, U. 1, Iber, S. 1, Wedemeyer, R.-S. 2, Blume, H. 2, Weitschies, W. 1<br />

1 University of <strong>Greifswald</strong>, Center of Drug Absorption and Transport, Institute of Pharmacy,<br />

Department of Biopharmaceutics and Pharmaceutical Technology, Felix-Hausdorff-Street 3, 17487<br />

<strong>Greifswald</strong>, Germany<br />

2 SocraTec R&D Concepts in Drug Research and Development GmbH, Im Setzling 35, 61440<br />

Oberursel, Germany<br />

For patients with swallowing disorders or other diseases that preclude the oral route of food<br />

intake the use of enteral feeding tubes is necessary. Often the administration of protonpump<br />

inhibitors (PPI) like omeprazole is required for these patients to avoid stress-related<br />

mucosal bleeding or acid pneumonitis [1]. Unfortunately, there is still a lack of knowledge<br />

regarding the handling of drug application via feeding tubes. The aim of this study was to<br />

investigate the ability of three different omeprazole formulations (Nexium ® mups 40 mg,<br />

enteric coated tablets containing micropellets (Astra Zeneca GmbH), Omeprazol Hennig ®<br />

40 mg (Hennig Arzneimittel GmbH & Co. KG) and OMEP ® 40 mg (Hexal AG), both<br />

capsules with enteric coated pellets) to pass enteral feeding tubes in order to obtain<br />

recommendations for an appropriate application of these formulations via this route of<br />

administration.<br />

The feasibility of application was tested using two different feeding tubes (representatively<br />

for percutaneous endoscopic gastrostomy (PEG): Freka ® PEG-Set gastral CH 9, length:<br />

30 cm, 1 terminal port; representatively for nasogastric tubes: Freka ® Intestinale Sonde<br />

134 Poster


CH 9 for PEG CH 15, length: 120 cm, 2 lateral, 1 terminal port; both Fresenius Kabi AG).<br />

As application media served tap water, boiled tap water and different commercially<br />

available still mineral waters. The used media were analysed regarding pH, buffer capacity,<br />

and electrical conductivity. Diameter, swelling behaviour, and stability of the pellets over<br />

480 min were also investigated. To study the application of the pellet formulations via<br />

feeding tubes an appropriate application protocol had to be established. Briefly, the tubes<br />

were washed with 30 mL test fluid before and after the application. Pellets from one opened<br />

capsule or one tablet were administered together with 30 mL of the test fluid by using<br />

50 mL catheter syringes with included Luer adaptors. The contents were slowly injected<br />

through the tubes while gently shaking the syringe to avoid obstructions. For a complete<br />

removal of the Omeprazol Hennig ® and OMEP ® pellets out of the syringe the use of further<br />

60 mL of additional test fluid was necessary. The contents of the syringes were injected into<br />

the tubes either directly or after 1 h of rest. After tube passage the administered<br />

suspensions were collected, decanted, solved in 100 mL 0.1 M NaOH and further diluted.<br />

The analyses of the diluted probes (1:20) and calibration measurements were performed at<br />

304 nm using an UV spectrophotometer (Varian Cary 50, Varian Inc., Palo Alto, USA).<br />

Pellets of Nexium ® mups were small enough to pass all tubes without any problems. Pellets<br />

of OMEP ® only were able to pass the PEG without using the Luer adaptor. The application<br />

through the nasogastric tube immediately resulted in obstructions. Omeprazol Hennig ®<br />

pellets passed the PEG without any difficulties, the nasogastric tube could only be passed<br />

immediately after preparation of the suspension without using the Luer adaptor. This may<br />

be attributed to the swelling of the pellets during 1 h of rest. The swollen pellets had a<br />

higher tendency to agglomerate. For all media comparable results were obtained. For the<br />

latter two formulations nasogastric tubes with CH > 9 need to be tested.<br />

In conclusion, the ability of omeprazole formulations to pass through a feeding tube is<br />

extremely dependent on the properties of the formulation and the used liquid as well as on<br />

the type of feeding tube. Micropellets as used in Nexium ® mups are small enough to pass<br />

even small feeding tubes like nasogastric tubes without any problems. The bigger the pellet<br />

diameter, the higher is the risk of obstruction. Also the shape of the pellets has an impact<br />

on the administration as non spheric formed pellets more often tend to agglomerate inside<br />

the tube than spheric ones. Short tubes with only on terminal port like PEGs showed less<br />

obstructions compared to long tubes with lateral ports like nasogastric tubes. Generally, all<br />

omeprazole formulations should be administered slowly and under continuous shaking of<br />

the syringe immediately after preparation of the solution. Tube washing should be<br />

performed before and after the administration of drugs.<br />

References:<br />

1. Johnson, J. L. et al.: Am. J. Health-Syst. Pharm. 2008, 65: 2324-2325.<br />

2. Devlin, J. W. et al.: Ann. Pharmacother 200, 39: 1667-1677.<br />

162<br />

Nanostructured Lipid Carriers (NLC): The optimum concentration and<br />

production condition<br />

Junmahasathien T. 1; Müller R. H. 1; Keck, C. M. 1 2<br />

1 Institute of Pharmacy, Department of Pharmaceutics, Biopharmaceutics & NutriCosmetics, Freie<br />

<strong>Universität</strong> Berlin, Kelchstrasse 31, 12169, Berlin, Germany<br />

2 Applied Pharmacy Division, University of Applied Sciences Kaiserslautern, Campus Pirmasens,<br />

Carl-Schurz-Strasse 10-16, 66953 Pirmasens, Germany<br />

It is interesting that there is a finished product (lipid matrix) which is the mixture of oils, fatty<br />

alcohol and vegetable waxes (Polyglyceryl-2 Dipolyhydroxystearate, Octyldodecanol,<br />

Carnauba Wax, Candellila Wax, Beeswax, Cetearyl Glucoside and Cetearyl Alcohol) It can<br />

be used as NLC.<br />

Nanostructured lipid carriers (NLC) have been developed to overcome the disadvantages<br />

of solid lipid nanoparticles (SLN). Solid lipid mixed with liquid lipid, to decrease the degree<br />

of perfect crystal of lipid matrix.[1,2] The aim of this study was to develop a new<br />

formulation, e.g. the NLC-preconcentrate, containing high amounts of small sized NLC. For<br />

this propose, a new lipid matrix was used and the influence of the lipid concentration (10-<br />

30%) including the number of high pressure homogenization cycles (3, 5 and 10 cycles) on<br />

the particle size and physical stability were investigated. The formulation consisted of a new<br />

lipid matrix, Plantacare® 2000UP 2% as a stabilizer and water. Each formulation was<br />

premixed by an Ultra-Turrax at 8,000 rpm for 30 seconds and produced at 80°C using high<br />

pressure homogenization (HPH) at 500 bar. The results showed that the increase in the<br />

lipid concentration led to a larger particle size. More cycles led to smaller particles. The<br />

smallest particle size (145 nm) was obtained from the formulation containing 10% lipid<br />

which produced by applying 5 cycles of HPH. The maximum lipid concentration was 25%.<br />

Samples containing higher concentration, e.g. 30% lipid content, solidified during the<br />

production. The physical stability was affected by the lipid content, e.g. a higher lipid<br />

content led to a faster increase in particle size over time. Formulations containing 10% lipid<br />

remained physically stable over time. In conclusion, a new NLC formulation was<br />

successfully developed and optimized in this study. The optimum lipid concentration for<br />

producing this NLC is 10%.The optimal production conditions are 5 cycles of HPH, 500 bar<br />

at 80°C.<br />

Acknowledgments: UPM Handelsgesellschaft mbH, Haupstrasse 47 D-21447 Handorf Germany, PharmaSol<br />

GmbH, Blohmstrasse 66A 12307 Berlin Germany<br />

References:<br />

1. Teeranachaideekul V., et al.: Eur. J. Pharm. Biopharm. 2007, 67: 141-148.<br />

2. Müller R. H., et al.: Adv Drug Deliv Rev. 2007, 59: 522-530.<br />

163<br />

Regulations of Preclinical Studies and Clinical Trials in the Former GDR<br />

Retzar A., Friedrich C. 1<br />

1 Institute for the History of Pharmacy, Philipps-<strong>Universität</strong> Marburg, Roter Graben 10, 35032<br />

Marburg, Germany<br />

The poster deals with the legal conditions of preclinical studies and clinical trials in the<br />

former German Democratic Republic.<br />

Since 1949 it was determined that introducing pharmaceuticals was not permitted without<br />

registration. The decision on registration was made by the Ministry of Health based on the<br />

recommendation of the so-called ‘Zentraler Gutachterausschuss für Arzneimittelverkehr<br />

(ZGA)’, a commission of clinicians, pharmacologists, pharmacists and representatives from<br />

industry.<br />

In 1960 a directive regulated the procedure of registration, which required the submission of<br />

pharmacological and clinical surveys.<br />

In 1964 the Medicinal Products Act was established containing the obligation to prove the<br />

effectiveness and inoffensiveness of new drugs in preclinical studies and clinical trials<br />

before they were launched. Under the new regulations approval by the ZGA was necessary<br />

to conduct clinical trials. Therefore, an application had to be filed, which provided<br />

information about extent, methods and responsibilities of the trial.<br />

Due to increasing requirements in the field of drug studies the regulations had been<br />

revised. Consequently, the 12th implementing rule of the Medicinal Products Act came into<br />

effect in 1976. From now on drug studies were subdivided into 4 levels. Measures of postmarketing<br />

surveillance in level no. 4 were determined to detect adverse drug reactions.<br />

Acknowledgments: Institute for the History of Pharmacy, Ariane Retzar.<br />

References:<br />

Richter, J., Keune, H. G.: Arzneimittelrecht der DDR. Kommentar Teil I (Akademie-Verlag Berlin) 1972.<br />

Hackenberger, F., Koch H.: medicamentum 1977, 18(5): 130–135.<br />

164<br />

Pharmacy under the sea – The medical supply of the German „U-Boot-Waffe“<br />

during the World Wars<br />

Vongehr, F. 1; Friedrich, C. 1;<br />

1 Institut für Geschichte der Pharmazie der Philipps-<strong>Universität</strong> Marburg, Roter Graben 10, 35032<br />

Marburg, Germany<br />

Despite the fact that German submarines have been a common subject of military-historical<br />

research and literature, the medical service aboard which was determined by the special<br />

environmental conditions is a rather neglected aspect. However, it is known that<br />

complicated surgery such as amputations could be performed even in dived submarines,<br />

although the crews had to deal with severe problems like climate, humidity and narrowness.<br />

The question arises how the medical supply was organized.<br />

Beginning with the first German submarine „U 1“ commissioned in 1906 the new boats<br />

were only equipped with a few remedies, such as disinfecting ointment and acetylsalicylic<br />

acid, but during Wolrd War I the experience lead to the conclusion to extend the selection<br />

of drugs on the boats. Therefore, the navy issued a compendium, the so-called „Ärztlicher<br />

Ratgeber für Unterseeboote“, containing a more detailed medical supply and several<br />

instructions for the commanders, who mostly had to operate their boat without a physician.<br />

During World War II the „Kriegsmarine“ had learned from the experiences of the<br />

„Kaiserliche Marine“ and published revised editions of the „Ärztlicher Ratgeber“ in the<br />

1940s. The medical supply was now significantly augmented and several boats even had<br />

physicians among the crew. Especially submarines designed for tactical operations in<br />

remote areas carried an extensive supply of drugs, including anti-malaria drugs and<br />

sulfonamides.<br />

The allies confiscated important documents regarding the German „U-Boot-Waffe“ including<br />

logfiles and medical data for their own use after victory. It had lasted several decades until<br />

the documents were returned and opened for research. The historical sources regarding<br />

pharmaceutical questions aboard the German submarines are now an object of detailed<br />

research.<br />

165<br />

Market surveillance study to medical products with pyridostigmine:<br />

tetramethylurea contamination in tablets and injections relevant to regulatory<br />

legislation for toxicological concerns<br />

Bogan R; Zimmermann T<br />

Central Institute of the Bundeswehr Medical Service, Ingolstädter Landstraße 102, 85748 Garching<br />

Hochbrück<br />

Pyridostigmine is a well known Active Pharmaceutical Ingredient (API) described in<br />

monographs of the European, the British and the United States Pharmacopeia.<br />

Nevertheless in previous studies we elucidated a hitherto new degradation pathway of<br />

pyridostigmine leading to formation of tetramethylurea (TMU) [1,2]. Based on these data we<br />

analysed marketed medical products with API pyridostigmine for the occurrence of TMU.<br />

The samples tested were within and after expiry date. Dosage forms of tablets and<br />

injections were considered. As shown before the compendial method of the European<br />

Pharmacopeia could be used for detection and quantification of TMU.<br />

We examined four batches of tablets in two dosage strengths and two batches of injections<br />

in one dosage strength from two marketing authorization holders. Samples were analysed<br />

two times in a distance of 24 months; in between they were stored at the recommended<br />

condition below 25°C. At the start of the study the remaining time to the expiry date of 5<br />

batches was between 5 and 48 months. For comparison reason one batch 7 months after<br />

expiry date was included. At the retest time 5 batches exceeded the expiry date for 4 to 31<br />

months whereas 1 batch was still 24 months before expiry date. At the first examination<br />

TMU was detected in one batch of tablets 9 months before expiry date and in the one batch<br />

of injections which had exceeded the expiry date for 7 months. The second examination<br />

revealed an additional batch with TMU and a marked increase of TMU content in those<br />

batches which were already contaminated. The highest contamination was found in one<br />

batch of tablets with 0.36 % (area percent related to API) 9 months before expiry date and<br />

0.51 % resp. 15 months after expiry date.<br />

The study proved that the predicted degradation pathway also occurs in market authorized<br />

medical products. TMU contamination seems to be in particular a problem at the end or<br />

after expiry date. Nevertheless the occurrence of TMU even within expiry date was shown<br />

and has to be considered. The values measured exceed the threshold values for<br />

degradation products of the ICH [3]. Toxicological evaluation does not suggest acute<br />

toxicity in the relevant dose range [4,5], but in terms of chronic toxicity the risk of a<br />

teratogenic effect of TMU can not be excluded [6,7].<br />

As a conclusion of our findings efforts on a revision of the relevant monograph<br />

pyridostigmine in the European Pharmacopeia as well as regulatory provisions are in<br />

process.<br />

References:<br />

1. Bogan, R. Loch, M.: Wehrmed. Mschr. 2010, 54: 242-247<br />

2. Bogan, R.: Joint meeting of the austrian and german pharmaceutical societies, abstract volume, 2011, PS<br />

001: 86<br />

3. International conference on harmonization of technical requirements for registration of pharmaceuticals<br />

for human use, ICH Harmonised Tripartite Guideline Q3B, Impurities in new drug products, 2006<br />

4. Dixon, R.L. et al.: Arch. Int. Pharmacodyn. Ther. 1966, 160: 333<br />

5. Edler, L. et al.: Food and chemical toxicology. 2002, 40: 283-326<br />

Poster 135


6. Munley, S.M. et al.: Drug. Chem. Toxicol. 2001, 24: 259-271<br />

7. Teramoto, S. et al.: Teratology. 1981, 23: 335-342<br />

166<br />

Improved taste evaluation of pharmaceutics using food sensors?<br />

Eckert, C., Pein, M.<br />

Institute of Pharmaceutics and Biopharmaceutics, Heinrich-Heine University, Duesseldorf,<br />

Germany<br />

Introduction: Two electronic taste sensing systems are commercially available. Whereas<br />

one sensor set with seven sensors related to specific gustatory stimuli is offered for the<br />

Japanese taste sensing system (Insent), Alphamos (France) offers two sensor sets<br />

containing seven cross-selective sensors for the αAstree. The two sensor sets are<br />

recommended for different applications. While sensor set 2 is marketed for pharmaceutical<br />

applications, sensor set 5 is recommended for taste evaluation of food. So far, this<br />

differentiation has not been questioned. Therefore, two active pharmaceutical ingredients<br />

(APIs) and two different lozenges were characterised using these two sensor sets.<br />

Materials and methods: Caffeine citrate (DAC) was purchased from Fagron (Barsbuettel,<br />

Germany), quinine hydrochloride (Ph. Eur.) from Caesar&Loretz (Hilden, Germany). Both<br />

APIs were determined in concentration ranges from 0.05 mM to 50 mM in 12 levels. Both<br />

lozenges were evaluated using different sample weights (0.5-12 g/100 ml).The samples<br />

were dissolved in aqua purificata and measured using the αAstree equipped with sensor<br />

sets 2 and 5. The measurement setup was used according to Pein [1].<br />

Results and discussion: Each sensor of both sensor sets fulfils the repeatability<br />

requirements [2] with RSD values


34 patients (BMI < 20: 4, 20-25: 17, > 25: 13). In 18 patient charts the documentation of<br />

bodyweight and height was missing. As an index for renal function the GFR was evaluated<br />

from serum creatinine. 25 (48.1%) patients showed a GFR > 60 ml/min/1.73 m², 21 (40.4%)<br />

patients between 30-59 ml/min/1.73 m² and 6 (11.5%) patients < 30 ml/min/1.73 m².<br />

Potassium levels as an important electrolyte for cardiac functions were for almost all<br />

patients in a normal range (3.5-5.1 mmol/l).<br />

CONCLUSION: Our results confirm the study of Groth-Tonberge et al. concerning fallpromoting<br />

drugs on a cardiological ward [2]. Average age, medication per patient and most<br />

prescribed medication groups (diuretics, beta blockers, angiotensin-converting-enzyme<br />

(ACE) inhibitors, antibiotics) were in good agreement and a potential relation between falls,<br />

polypharmacy and higher age could be shown. Additionally, considering the fact that more<br />

than 50% of the patients also had an impaired kidney function (GFR < 60 ml/min/1.73 m²)<br />

the importance of pharmaceutical care focused on both polypharmacy and adequate drug<br />

dosing is evident and could help to prevent fall events.<br />

References:<br />

[1] Pierobon, A., Funk, M. Sturzprävention bei älteren Menschen. Risiken–Folgen–Maßnahmen (Georg<br />

Thieme Verl.) 2007.<br />

[2] Groth-Tonberge, C., Feuchtinger, J., Strehl, E. Krankenhauspharmazie 2011, 32(11): 565.<br />

[3] Levey, A. S. et al. Ann Intern Med 2009, 150(9): 604.<br />

170<br />

Development of lipid micropellets with metformin hydrochloride using solventfree<br />

cold extrusion/spheronization<br />

Petrovick, G.F.; Breitkreutz, J.<br />

Institute of Pharmaceutics and Biopharmaceutics, Heinrich-Heine-University, 40225 Duesseldorf,<br />

Universitaetsstrasse 1, Germany<br />

The diabetes mellitus type 2 (DM2) constitutes 85 to 95% of all diabetes cases in<br />

developed countries and has reached epidemic proportions in several nations [1, 2].<br />

Metformin is the drug of choice to treat DM2 in many countries [3, 4]. But the relatively large<br />

size of available tablets (doses form 500 to 1000 mg) can cause difficulty to swallow<br />

(especially in elderly patients) which may lead to discontinuation of treatment. Considering<br />

this problem, microparticulate systems have advantages when compared to tablets or<br />

capsules [5]. Solid lipid extrusion in a twin-screw extruder is an innovative preparation<br />

method for matrices in which lipids are treated below their melting ranges in a thermomechanical<br />

process. The drug is dispersed in a lipid matrix in which the lipid does not melt<br />

but softens [5, 6].<br />

A hard fat (Witocan ® 42/44) was used as lipidic binder. The extrusion was performed at 33<br />

°C in a Micro 27GL-28D extruder (Leistritz, Nuremberg, Germany) with co-rotating twin<br />

screws, using a screw plate with 91 dies of 0.5 mm and 1.35 mm length, with a powder<br />

feed rate of 40 g/min. The extrudates were spheronized (Schlueter spheronizator RM 300,<br />

Schlueter, Neustadt am Ruebenberge, Germany) for 15 min at 1500 rpm and 33 ± 1 °C.<br />

Metformin hydrochloride lipid micropellets (figure 1) with a high drug load of 80% presenting<br />

narrow particle size distribution and aspect ratio under 1.2 could be obtained. The process<br />

parameters such as pressure and product final temperature were also evaluated (figure 2).<br />

Spheronization was difficult as the process temperature range was difficult to control.<br />

Friction and heat conveying were major issues as they influence processability and product<br />

quality.<br />

Solvent-free cold extrusion followed by spheronization present itself as a robust method to<br />

obtain high dosage micropellets of metformin with adequate size and shape characteristics<br />

which are mainly impacted by the spheronization step.<br />

Figure 1: Micropellets with 80%<br />

metformin hydrochloride<br />

Figure 2: Process parameters<br />

References:<br />

1. Zimmet, P.Z. et al., J Diabetes Complicat 1997, 11(2): 60-68.<br />

2. Giannella-Neto, D.; Gomes, M.B. Diabetol Metab Syndr 2009, 1(1): 1-3.<br />

3. Gregorio, F. et al., Arch Gerontol Geriat 1996, 22(1): 161-170.<br />

4. Soto, A. et al., Endocrinol Nutri 2008, 55(2): 39-52.<br />

5. Breitkreutz, J.; Boos, J., Eur J Pharm Biopharm 2007, 56, 255-260.<br />

6. Witzleb, R. et al., Powder Technol 2011, 207, 407-413.<br />

171<br />

Development of a simple, rapid and reliable assay to count enterococcus<br />

faecium with a CASY coulter counter to assess antibiotic therapy<br />

Goebgen E; Wicha SG; Kloft C<br />

Dept. of Clinical Pharmacy and Biochemistry, Institute of Pharmacy, Freie Universitaet Berlin,<br />

Kelchstraße 31, 12169 Berlin, Germany<br />

OBJECTIVES: Determination of bacterial counts plays a key role when the interaction<br />

between bacteria and antibiotic are investigated, i.e. to assess the potency of a drug,<br />

evaluate drug combinations or improve dosing schedules. Cell counts of bacteria are<br />

usually determined by densitometry or plate counting methods. As densitometry is highly<br />

imprecise and plate counting methods are time-consuming, cost intense and require long<br />

preparation times, there is a clear need for an easier method to quantify bacteria. In 1953,<br />

W. Coulter developed the technique of electronic cell counting by measuring the change in<br />

resistance for blood cell counting. About 50 years ago, Kubitschek [1] and Swanton et al.<br />

[2] demonstrated the utility of the Coulter Counter in cell counting of bacterial cells and the<br />

determination of cell volumes in growing cultures of bacteria. In comparison to the standard<br />

methods, electronic cell counting promises to save time, to reduce costs whilst providing a<br />

higher content of information (e.g. cell counts, cell volume and distribution of cell volume)<br />

[1,2]. The objective of the current analysis was to establish an assay on a CASY Coulter<br />

Counter as a fast and simple electronic cell counting method for E. faecium to assess<br />

antibacterial effects in vitro.<br />

METHODS: The Coulter Counter (CASY 1, Roche Diagnostics) included a pump, an<br />

aperture tube (60 µm) and the electronic components: one electrode inside the tube, one<br />

outside and the voltage pulse measuring unit. The number of bacteria is determined by<br />

measuring the change in resistance when a particle passed the aperture. Additionally, the<br />

amplitude of the pulse is proportional to the size and the volume of the particle. To<br />

decrease background-counts, all fluid media were filtered through a 0.2 µm filter. An overnight<br />

subculture of E. faecium (ATCC 700221) was used as stock suspension with an initial<br />

inoculum size equal to McFarland 3 in 0.9% NaCl. Serial dilutions in phosphate buffered<br />

saline with peptone (PBSP) were prepared. Dependent on the bacterial concentration of the<br />

suspension, samples of 10, 100 or 1000 µL were measured in 10 mL filtered, ready-to-use<br />

measuring liquid (Casy ton, Roche Diagnostics). The electronic counting method was<br />

compared to the standard plate count method [3]: 100 µL of each suspension was diluted<br />

and poured on Columbia agar plates. Plates containing 30 – 500 colony forming units<br />

(CFU) were read.<br />

RESULTS: The mean diameter range of E. faecium correlated well with the physical<br />

measuring range of the capillary from 1.2 µm to 45 µm. McFarland standard 3 was<br />

equivalent to 2.2x10 8 colony forming units (CFU)/mL. Analogues to a bioanalytical method,<br />

the performance of this electronic cell count assay was evaluated according to the EMA<br />

guideline [4]. Current investigations suggest agreement with the guideline with respect to<br />

linearity, accuracy and precision of the bacterial cell counts with a lower limit of<br />

quantification of 2.2x10 4 CFU/mL.<br />

CONCLUSIONS: A rapid, precise and simple assay for electronic cell counting was<br />

established and allows for bacterial cell counting of E. faecium in PBSP with a sample<br />

volume up to 1 mL. Thus further studies using the Coulter Counter with a smaller aperture<br />

tube for other bacterial species such as E. coli or P. aeruginosa are indicated. Bacterial<br />

growth experiments, in-process controls and the measurement of postantibiotic effects<br />

could be possible applications for the more efficient Coulter Counter principle so as to<br />

streamline the clinical and preclinical development of an antibacterial agent and to facilitate<br />

decision taking in the development of dosing schemes.<br />

References:<br />

1. Swanton EM, Curby WA, Lind HE: Appl. Environ. Microbiol. 1962, 10(5): 480-485.<br />

2. Kubitschek HE: Nature. 1958, 182: 234-235.<br />

3. Schwalbe R, Steele-Moore L, Goodwin AC: Antimicrobial susceptibility testing protocols (CRC Press)<br />

2007.<br />

4. EMEA/CHMP/EWP/192217/2009<br />

172<br />

Cost of drugs in the ambulatory health care sector – how much do we really<br />

spend?<br />

Aßmann, G. 1 2; Fiß, T. 1; Marschall, P. 2; Fleßa, S. 2; Hoffmann, W. 3<br />

1 German Center for Neurodegenerative Diseases (DZNE), site Rostock/<strong>Greifswald</strong>; Ellernholzstr.<br />

1/2; 17487 <strong>Greifswald</strong>, Germany<br />

2 University of <strong>Greifswald</strong>, Faculty of Law and Economics, Friedrich-Loeffler-Str, 70, 17489<br />

<strong>Greifswald</strong>, Germany<br />

3 University of <strong>Greifswald</strong>, Institute for Community Medicine; Ellernholzstr. 1/2 , 17487 <strong>Greifswald</strong>,<br />

Germany<br />

Introduction: As a result of demographic change the prevalence of age-related diseases<br />

increases. Due to the associated intensive drug therapy even in this part the German health<br />

system faces increasing costs and major challenges. Currently there is a lack of knowledge<br />

about daily drug costs in the ambulatory setting. Our aim was the calculation of daily costs<br />

for regularly taken drugs (prescribed and over-the-counter) in the AGnES-home (AGnES:<br />

GP-supporting, community-based, e-health-assisted systemic intervention) visit cohort.<br />

Additionally we focussed on associations between medication costs and sociodemographic<br />

parameters.<br />

Methods: Basis of our analysis was a subsample of the AGnES-home visit cohort who<br />

received a home medication review between 2006 and 2008 (n=778 patients with n=4985<br />

regularly taken drugs). Current drug costs were added automatically at the time of data<br />

recording based on the German drug index. Missing prices were completed by mean DDDcosts<br />

which were extracted from the German Drug Report. Sociodemographic criteria were<br />

taken from the initial proband interview.<br />

Results: Mean age of all participants was 78.8 years, women were significantly older<br />

(women n=555; 71.3%, 79.9 years, p


Herbal medicinal products containing ethanol are widely used in children [1], since ethanol<br />

it is an excellent extraction solvent for the phytochemical components of herbal drugs and<br />

contributes to the stability of fluid preparations. Toxicological and pharmacokinetic<br />

evaluations [2] have shown that the small amounts of ethanol applied with therapeutic<br />

doses of these medicines are safe even in this age group. Despite these medicines have<br />

been used safely since many decades, they occasionally are subject of discussion in the<br />

public, triggered by the increasing problem of recreational misuse of alcoholic beverages by<br />

children and young persons [3, 4]. Therefore, there is a growing need of a systematic<br />

evaluation of epidemiological data on these medicines.<br />

For an evaluation of the experience gained from the therapeutic use of these medicines, 17<br />

pro- and retrospective studies with 10 different herbal medicinal products containing<br />

ethanol at doses of 40 to 240 mg per single application, depending on the age group, have<br />

been analyzed. These studies cover 50,425 patients between 0 and 12 years of age. In<br />

these studies, altogether 15 adverse drug effects have been described, none of which was<br />

attributable to the ethanol content of the medicines.<br />

In a worldwide survey including these and further 6 herbal medicinal products it was shown<br />

that during the past few years, more than 764 millions daily doses have been sold,<br />

corresponding to more than 33 millions patients (according to data obtained from<br />

manufacturers; figures are available partly from 1993 onwards, partly from 2003/4<br />

onwards). From the packages sold in Germany in the years between 2005 and 2009, 48.1<br />

millions were attributable to self-medication, and 10.8 millions to prescriptions reimbursed<br />

by health insurance (IMS, Frankfurt). As non prescription medicines are reimbursed in<br />

Germany only in children, at least the latter part of the prescriptions can be attributed to<br />

children to an extent of at least 90 %. All of these medicines are registered or licensed by<br />

regulatory authorities, so that adverse effects are covered by the pharmacovigilance<br />

system. No adverse effects attributable to the ethanol content have been reported.<br />

This set of pharmacovigilance data supports the conclusion drawn from the experience of a<br />

safe use over decades, i.e. that the ethanol content of herbal medicinal products does not<br />

give any causes for concern regarding their safety even in children.<br />

Dedication in memoriam: This contribution is dedicated to Prof. Dr. Hilke Winterhoff, Institute for<br />

Pharmacology and Toxicology, University of Münster, Germany, who died on 9 May 2010. She has initiated<br />

and supported this work.<br />

References:<br />

1. Gundermann, K.-J. et al., Neurogastroenterol Motil 2010, 22(S1): 69.<br />

2. Kelber, O. et al., Pharm Ind 2008, 70: 1124-1128.<br />

3. Gemeinsamer Bundesausschuss (G-BA), Bundesanzeiger 21 vom 08. Februar 2011.<br />

4. HMPC, 2010. Reflection paper. EMA/HMPC/85114/2008.<br />

174<br />

Oral mesalazine treatment of ulcerative colitis – a review of current dosage<br />

forms<br />

Kersten E1, Klein S1 1<strong>Ernst</strong>-<strong>Moritz</strong>-<strong>Arndt</strong> University <strong>Greifswald</strong>, Department of Pharmacy, Institute of Biopharmacy and<br />

Pharmaceutical Technology, C_DAT, Felix-Hausdorff.Str. 3, 17489 <strong>Greifswald</strong><br />

Ulcerative colitis (UC) is a chronic and debilitating illness prevalent in the Western<br />

population characterized by chronic inflammation of the colon that is continuously<br />

distributed in the colonic mucosa. Most of the currently available agents used in UC<br />

treatment, e.g. mesalazine, are aimed to downregulate the chronic inflammation in the<br />

intestinal mucosa. The main principle of mesalazine action is a topical reduction of<br />

inflammation in the mucosa. Thus, oral mesalazine dosage forms should release the active<br />

substance selectively at the inflamed areas in the GI tract. For a long time marketed enteric<br />

coated formulations containing 250 – 500 mg of the active drug represented the<br />

formulations of choice for this purpose. However, since average daily mesalazine doses are<br />

typically > 3g/ multiple daily dosing is required. Thus, more recently, several new<br />

formulations including coated multiparticulates and an extended release tablet formulation<br />

containing much higher drug loads were released to the market to improve both<br />

effectiveness and patient compliance.<br />

To get an idea, if the newer types of formulations do represent a benefit with regard to<br />

improving UC therapy, we studied drug release of the various formulations with a pHgradient<br />

method that has been established as a predictive in vitro model in earlier studies<br />

(Klein 2005, 2008). Experiments were performed with a BioDis ® Release Rate Tester. In<br />

contrast to simulating only the recommended dosing conditions, i.e. fasted gastrointestinal<br />

(GI) pH-conditions, as has been done in previous studies, in the present study both fasted<br />

and fed state dosing conditions were addressed. Experiments were also performed with the<br />

official mesalazine test methods described in the United States Pharmacopoeia (USP) to<br />

screen, if the could also come along with a in vivo predictivity.<br />

Results obtained in our experiments clearly show that the site and extent of drug release<br />

are strongly dependent on the type of formulation whereas the drug load has a minor<br />

impact on the release pattern. Unexpectedly, drug release from all enteric dosage forms<br />

was not much affected by the different pH-conditions in the fasted and fed GI lumen.<br />

However, as expected, results from the compendial test setup were different from those<br />

obtained with the biorelevant pH-gradients and therefore will not really be predictive for in<br />

vivo drug release.<br />

Results indicate that none of the marketed dosage forms releases the active drug<br />

selectively in the colon. Moreover, as the marketed formulations show significant<br />

differences in their release profiles, the choice of the dosage form should be based on an<br />

individual basis. The enteric formulations were not prone to the pH-conditions used to<br />

simulate fed state gastric residence. However, it should be kept in mind that beside the pHconditions,<br />

other physicochemical and mechanical aspects can affect drug release,<br />

particularly when comparing fasted vs. fed state dosing conditions. As particularly, the<br />

tablet formulations are expected to be sensitive to a variety of these factors, this will be<br />

addressed future studies.<br />

References:<br />

Klein, S., J. Stein, and J. Dressman, J Pharm Pharmacol, 2005. 57(6): 709-19.<br />

Klein, S., et al. J Contr Release, 2008. 130(3): 216-219.<br />

175<br />

ATR-IR-Imaging of Irbesartan Generic Tablets - Comparison with the Originator<br />

Norwig, J.; Jansen, R.<br />

Bundesinstitut für Arzneimittel und Medizinprodukte (BfArM), Kurt-Georg-Kiesinger-Allee 3, D-<br />

53175 Bonn, Germany<br />

According to the CTD format applicants for a marketing authorisation are requested to<br />

declare the exact composition of the dosage form and of the whole product.[1] Modern<br />

ATR-IR-Imaging allows a full identification of the constituents without any sample<br />

preparation for a chromatographic method in the conventional sense.[2]<br />

Neither an extraction, nor a separation, nor any chromatographic method is required. In<br />

case of Irbesartan tablets manufacturers from Far East could be checked whether the<br />

composition complies. Moreover it is estimated which content and which particle size the<br />

active substance shows in comparison with the originator.<br />

The imaging is an impressive powerful method to check the compliance with the<br />

declaration. The time consumption is estimated to be 1 h.<br />

However impurities in the 0.1 percentage range could not be identified. Therefore a trace<br />

analytical procedure should be combined with the method for full compliance with the<br />

Pharmacopoeia.[3, 4]<br />

Further images show the whole range of excipients.<br />

References:<br />

1. Notice to Applicants, Presentation and format of the dossier CTD in EudraLex, VOLUME 2B, Medicinal<br />

products for human use, Editor: European Commission Directorate General III, Brussels, July 2003.<br />

2. ATR Imaging of Pharmaceutical Tablets, PerkinElmer Life and Analytical Sciences.<br />

3. European Pharmacopoeia 7th Edition 2010, Monograph on Irbesartan.<br />

4. Irbesartan tablets, Monograph US Pharmacopoeia 35, Rockville (<strong>2012</strong>).<br />

176<br />

Lipopeptide-modified carrier systems – the composition determines cell<br />

selectivity<br />

Sydow, K1, Nikolenko, H1, Taraschewski, A1, Dathe, M1 1 Leibniz Institut für Molekulare Pharmakologie (FMP), Robert-Roessle-Strasse 10, 13125 Berlin,<br />

Germany<br />

Liposomal und micellar delivery systems have become one of the most promising options<br />

for the improvement of transport efficiency of poorly soluble drugs and for drug targeting.<br />

To increase selective uptake, target recognizing and transmembrane transport-mediating<br />

motifs are used. To achieve these requirements we developed lipopeptides, among them<br />

P2A2[1]. Because of their amphiphilic character, the peptides self-assemble to micelles and<br />

integrate into the lipid bilayer of liposomes. Interestingly, P2A2-micelles and -liposomes<br />

differ in cell selectivity and activate different modes of cellular uptake. To investigate the<br />

role of particle size and surface charge/charge distribution as potential determinants of<br />

particular uptake mechanisms, we synthesized lipopeptides of varying amino acid<br />

composition. Zeta potential measurement and dynamic light scattering were used to<br />

characterize the different micellar and liposomal systems. Cell selectivity was investigated<br />

by uptake studies with endothelial cells of the human aorta (HAoEC) and human brain<br />

microvessels (HBMEC).<br />

All lipopeptides (P2A2, LP1, LP2) differ in amino acid composition but are identical in the<br />

number charged residues. P2A2 and LP1 have both 12 cationic residues, LP2 bears 12<br />

anionic amino acids. All lipopeptides form micelles with an hydrodynamic diameter (d[Vol])<br />

of about 10 nm. The zeta potential of the micelles correlates with the charge properties of<br />

the lipopeptide: P2A2/ LP 1 – positive; LP 2 - negative surface potential. Additionally, three<br />

other peptide-bearing carrier-systems, different in particle size and their molar lipid to<br />

peptide ratio (c l/p) were prepared. Large unilamellar vesicles (LUV, d[Vol] about 90 nm, c<br />

l/p = 1000) and small unilamellar vesicles (SUV d[Vol] about 25 nm, c l/p = 1000 or 20)<br />

have a distinct size distribution. At cl/p = 1000, the zeta potential does poorly distinguish<br />

from the potential of control liposomes. With higher lipopeptide concentration, c l/p = 20, the<br />

zeta potential changes in accordance with the amino acid motif. Uptake studies were done<br />

by using Laser Scanning Microscopy (LSM) and Fluorescence Activated Cell Sorting<br />

(FACS).<br />

All positively charged lipopeptide carriers were internalized into HAoEC and HBMEC.<br />

Negatively charged LP 2 particles were preferentially adsorbed on the cell surface. The<br />

overall interaction efficacy followed the order: micelles > SUV (c l/p = 20) > LUV (c l/p =<br />

1000) > SUV (c l/p = 1000). The uptake of P2A2-micelles into HBMEC was two times<br />

enhanced compared to HAoEC. In comparison to micelles (no cell specificity), LP 1<br />

incorporation into SUV (c l/p = 20) seems to improve cell specificity to HAoEC. A lower lipid<br />

to peptide ratio could enhance uptake of P2A2- and LP 1 - SUV.<br />

Self-aggregation of lipopeptides and binding to liposomes results in efficient cellrecognizing<br />

and uptake mediating systems. Cell-selectivity depends on both the molecular<br />

138 Poster


structure of peptides, the particle size and on the lipid to peptide ratio in liposomal<br />

formulations. The investigated lipopeptides provide a keytool for the development of drug<br />

delivery systems.<br />

References:<br />

1. Leupold, E., Nikolenko, H. & Dathe, M., Biochim. Biophys. 2009, 1788(2): 442-449.<br />

177<br />

Standard doses of intravenous and enteral moxifloxacin provide insufficient<br />

drug exposure for nosocomial pneumonia in intensive care unit patients<br />

Schaeftlein, A. 1 2; Kees, M. 3; Heininger,A. 4; Kloft, C. 1<br />

1Dept. of Clinical Pharmacy and Biochemistry, Institute of Pharmacy, FreieUniversitaet Berlin,<br />

Kelchstraße 31, 12169 Berlin,Germany<br />

2and Graduate Research Training program PharMetrX, Germany,<br />

3 Dept. of Anesthesiology and Intensive Care, Charité University Hospital – Campus Benjamin<br />

Franklin, Hindenburgdamm 30, 12200 Berlin, Germany<br />

4 Dept. of Anesthesiology and Intensive Care, University Hospital of Tuebingen, Hoppe-Seyler-<br />

Str.3, 72076 Tübingen, Germany<br />

Background: Moxifloxacin (MOX), a fourth-generation fluoroquinolone,is approved for<br />

treatment of community acquired pneumonia. Due to a broad spectrum and high enteral<br />

bioavailability (>91%) of MOX in healthy volunteers (HV) [1], intravenous/enteral sequential<br />

therapy may also be a suitable treatment option for hospital acquired pneumonia (HAP),<br />

possibly resulting in reduced costs and complications [2].In the current population<br />

pharmacokinetic analysisi.v. and enteral administration of MOX in intensive care unit<br />

patients (ICUP) were compared to determine the clinical feasibility of sequential therapy in<br />

ICUP on a pharmacokinetic basis.Additionally,patient specific characteristics (covariates)<br />

explaining interindividual variability (IIV) on pharmacokinetic (PK) parameters were<br />

investigated, and the simulated exposure of MOX was linked to the efficacybreakpoint of<br />

fluoroquinolonesfor HAP.<br />

Methods: Based on MOX concentration measurements after multiple i.v. and enteral<br />

administrations of 400 mg MOX in 25 ICUP,population PK analysis was performed using<br />

the nonlinear mixed-effect modelling approach (NOMEMTM).Covariates[demographics, coadministered<br />

drugs, state of disease markers] were tested for relevanteffectson the IIV of<br />

absorption rate constant (ka), enteral bioavailabilty, volumes of central and peripheral<br />

distribution and clearance (CL).Based on the final population PK model, 1000<br />

concentration-timecourses were simulated and the AUCtau were usedforprobability of target<br />

attainment functions(fAUC/MICHAP>75[3]), which is indicative for the efficacy and treatment<br />

response of MOX for ICUP with HAP.<br />

Results: Measured MOX concentrations in ICUP were best described and predicted by a<br />

first-order absorption and eliminationtwo compartment PK model. Estimates of CL (11.3<br />

L/h) and volume of distribution(114 L) were in accordance with the literature [4] butF (76%,<br />

95% confidence interval [59%; 94%]) was reduced in ICUP in comparison to HV [1]. IIV on<br />

ka (1.09 1/h) was high (135%) due to a lagging increase of MOX concentrations after<br />

enteral dosing observed in some ICUP which could not be integrated into the final model.<br />

Body weight, age, height, sex, administration of opioids or catecholamines, plasma<br />

proteins, presence of shock, application via gastric tube and sequential organ failure<br />

assessment score [5] had no significant effect on the IIV of MOX PK in ICUP. Only<br />

creatinine clearance (CLCR) effects the elimination of MOX and explained 1/4 of the IIVin<br />

CL, reducing CL by 0.43% for every 1 mL/min decline of CLCR. An acceptable probability<br />

(>0.9) to attain the PKPDtarget for HAP was achieved for only lowMICvalues of0.125<br />

µg/mL or less.<br />

Conclusions: Based on our PK/PD analysis, enteral bioavailability of MOX was little reliable,<br />

limiting the suitability of a switch to enteral therapy in ICUP. In addition, a favourable<br />

outcome of MOX treatment for HAP cannot generally be expected in the investigated<br />

setting, since MIC90values for typical pathogens of HAP (e.g E. coli) exceed 0.125 mg/L.<br />

Hence, unless a highly susceptible HAP pathogen is confirmed, an antibiotic regimen in<br />

ICUP with additional or alternative agents or a dose escalation might be considered.<br />

References:<br />

1. Burkhardt O, Stass H, Thuss U et al.:ClinPharmacokinet 2005, 44(9):969-76.<br />

2. Ramirez JA, Vargas S, Ritter GW et al.:Arch Intern Med1999; 159(20):2449-2454.<br />

3. Ambrose PG,Bhavnani SM, Rubino C et al.:Clin Infect Dis2007, 44(1):79-86.<br />

4. Jeffry AF, Tornøe CW, Brundage R et al.:J ClinPharmacol2011, 51(8):1152-1162.<br />

5. Vincent JL, Moreno R, Takala J et al.: Intensive Care Med1996; 22(7):707-710.<br />

178<br />

Antistatic effect of different silica types on free flowing direct compression<br />

excipients<br />

Knop K.; Martens A.;Sölter V.<br />

Institute of Pharmaceutics and Biopharmaceutics, Heinrich-Heine-University, <strong>Universität</strong>sstr. 1,<br />

40225 Düsseldorf, Germany<br />

When powders are handled, e.g. during sieving, mixing, or powder flow through a<br />

hopper,electrostatic charging may occur. Problems in the process of capsule filling or<br />

tableting can be a result of this electrostatic charge [1]. Antistatic agents such as<br />

magnesium stearate, talc, polyethylene glycolsor silicon dioxide are added to prevent these<br />

problems. The aim of the present study was to evaluate the influence of different types of<br />

silicon dioxide on the electrostatic charging of free flowing powders which are used for<br />

direct compression.<br />

Three free flowing direct compression excipientsFlowlac ® 100 (lactose monohydrate),<br />

Parteck ® M 100 (mannitol), and Ludiflash ® (co-processed mannitol with crospovidone,<br />

polyvinylacetate andpovidone) were investigated using the flowability test apparatus no. 1<br />

equipped with nozzleno. 3 (25 mm opening) described in Ph.Eur. 2.9.16 “Flowability”.The<br />

excipients were equilibrated to environmental conditions (21°C, 45% rel. humidity) for at<br />

least 48 h. Six different types of silicon dioxide were added in concentration of 0.5 and 1%:<br />

Hydrophilic (Aerosil ® 200 and 300), hydrophobic (Aerosil ® R972), and granulated fumed<br />

silica (Aeroperl ® 300) as well as porous silica (Syloid ® 244FP and Sylysia ® 350FCP).All<br />

types were tested in undried and dried status (105°C) to take the different water content<br />

into account. The powders were mixed in a Turbula ® mixer for 5 min. The flow rate was<br />

registered by the weight gain per time on a balance and the electrostatic potential was<br />

measured by an electrostatic sensor, focussed on the powder-stream in a well defined<br />

distance. All measurements were carried out in fivefold in an air-conditioned room at 21°C<br />

and 45% relative humidity.<br />

Pure Flowlac ® showed a high flowability and a negative electrostatic charge of about -0.5<br />

kV. The flowability was only slightly improved by the addition of silica. The electrostatic<br />

charge could not be reduced by the addition of the powdered fumed silica but by the<br />

granulated fumed silica and the porous silica types.<br />

Parteck ® M without additives was negatively charged to -1.5 kV during flow. This high<br />

voltage could be lowered to about -0.3 kV by the fumed silica types. The addition of the<br />

porous silica types led to a change in polarity and a charge of about +0.3 kV. The flowability<br />

of Parteck ® was worse than that of Flowlac ® but could be improved by the addition of each<br />

type of silica.<br />

Ludiflash ® was positively charged during flow up to +0.5 kV. All silica types with the<br />

exception of the granulated type reduced the charging to values around zero. Ludiflash ®had<br />

a good flowability like Flowlac ®. The addition of fumed silica led to a decline in flow whereas<br />

the porous silica had no influence.<br />

Neither the concentration of the silica in the powder mixture (0.5 or 1%) nor the water<br />

content of the silicon dioxides (dried or undried) showed a consistent influence onthe<br />

electrostatic charge and the flowability of the powder mixtures. In the investigated rage,<br />

both factors were only of minor influence.<br />

The choice of the best antistatic agent from the group of silicon dioxides has to be made<br />

individually for every powder or powder mixture. The porous silica types showed a charge<br />

reduction for all three investigated systems whereas the powdered fumed silica failed in the<br />

case of Flowlac ®.<br />

References:<br />

1. Szendeleit S.M., Elektrostatische Aufladung im Prozess der Tablettierung, Diplomathesis,<br />

Halle/Düsseldorf 2010.<br />

179<br />

Taste sensing system for the selection of drug candidates in early<br />

pharmaceutical development stage and rational development of taste masked<br />

orodispersible dosage forms<br />

Guhmann M1 2, Gerber F2, Poellinger N2, Breitkreutz J3, Weitschies W1 1 Department of Biopharmaceutics and Pharmaceutical Technology, University of <strong>Greifswald</strong>,<br />

17489 <strong>Greifswald</strong>, Germany<br />

2 Glatt Pharmaceutical Services, Technology Centre, Glatt GmbH, 79589 Binzen, Germany<br />

3 Institute of Pharmaceutics and Biopharmaceutics, Heinrich-Heine-University of Düsseldorf, 40225<br />

Düsseldorf, Germany<br />

The ability of an electronic taste sensing system to guide the selection of drug candidates<br />

and rationalize the development of taste masked orodispersible formulations was<br />

evaluated.<br />

Diclofenac was used as drug model investigating the free acid, the sodium and the<br />

potassium salt forms. Calibrations curves were established with different concentration<br />

ranges. Measurements were performed using the electronic taste sensing system TS-<br />

5000Z (Insent Inc., Japan) qualified based on ICH guideline Q2 [1]. The system was<br />

equipped with 7 lipid membrane sensors representing bitterness (1,2 and 3), sourness,<br />

saltiness, umami, and astringency and corresponding aftertaste qualities. Sensor<br />

responses (mV) of three consecutive measurements were analyzed by univariate data<br />

evaluation. Taste masked Orally Disintegrating Tablets (ODTs) were developed using the<br />

most suitable diclofenac drug form considering taste modalities, and evaluated by the<br />

electronic tongue versus placebos using Principal Component Analysis (PCA).<br />

Clear differences between the free acid form of diclofenac and its salt counterparts were<br />

detected by the electronic tongue. Diclofenac salts induced bitter (taste and aftertaste) and<br />

astringent (taste) responses with different detection intensities; saltiness stimuli was<br />

additionally recorded for the potassium salt. In contrast, only a bitter aftertaste from sensor<br />

3 was detected for the acid form. Based on these results, diclofenac acid was selected for<br />

manufacturing taste masked ODTs. Taste masking strategies used granules produced by<br />

coating of single drug crystals or wet granulation, further compressed into ODTs. The PCA<br />

map showed that intermediated products (granules) and finished products (ODTs) could be<br />

distinguished by the electronic tongue. All formulations differed from the pure diclofenac<br />

acid. Using PC-1 (77% of the information), the taste reduction was 33% and 54% for drug<br />

coated granules and matrix granules respectively; 67% and 71% for their corresponding<br />

ODTs.<br />

Investigating diclofenac as a drug model, the electronic tongue proved to be a valuable tool<br />

for the selection of different drug candidates in the early pharmaceutical development<br />

phase and the rational development of taste masked orally disintegrating dosage forms<br />

Acknowledgments: The authors gratefully acknowledge the Institute of Pharmaceutics and<br />

Biopharmaceutics of the Heinrich-Heine University Düsseldorf, especially Maren Preis, for providing support<br />

and advice.<br />

Reference:<br />

1. Woertz, K et al.: J. Pharm. Biomed. Anal. 2010, 51: 497-506.<br />

180<br />

Complexation of Itraconazole with various cyclodextrin derivatives: Solubility<br />

and 1H-NMR studies<br />

Taupitz, T. 1; Bodtke; A. 2; Böheim, K. 2; Link, A. 2; Klein, S. 2<br />

1 Goethe University Frankfurt am Main, Institute of Pharmaceutical Technology, Max-von-Laue-Str.<br />

9, 60438 Frankfurt (Main)<br />

2 <strong>Ernst</strong>-<strong>Moritz</strong>-<strong>Arndt</strong> University <strong>Greifswald</strong>, Department of Pharmacy, Institute of Biopharmacy and<br />

Pharmaceutical Technology, C_DAT, Felix-Hausdorff.Str. 3, 17489 <strong>Greifswald</strong><br />

In the present study we wanted to investigate the complexation behaviour of different<br />

cyclodextrin derivatives with itraconazole and moreover elucidate the impact of these<br />

derivatives on the solubility of this poorly soluble compound. For this purpose binary<br />

cyclodextrin complexes of itraconazole, a weakly basic BCS class II drug, were prepared<br />

using the following cyclodextrin derivatives: I) 2-hydroxypropyl-β-cyclodextrin (HP-β-CD),<br />

II) sulfobutylether-β-cyclodextrin (SBE-β-CD) and the recently developed hydroxybutenylβ-cyclodextrin<br />

(HBen-β-CD). The main objective was to obtain formulations that show an<br />

improved solubility superior to that of the pure drug and further to investigate the<br />

interactions in the different itraconazole cyclodextrin complexes in detail.<br />

A freeze drying procedure was used to prepare all inclusion complexes of itraconazole. The<br />

pure drug and all formulations were then subject to pX-RD analysis and solubility tests. To<br />

elucidate the complex formation of each cyclodextrin derivative with the compound, 1H-<br />

NMR studies were performed. Finally, solubility tests were performed in compendial and<br />

biorelevant media simulating conditions in the stomach and the upper small intestine.<br />

Poster 139


PX-RD diagrams of the complex formulations indicated that amorphous inclusion<br />

complexes were obtained. Spectra obtained from the 1H-NMR studies revealed chemicals<br />

shifts of the triazol- and the aromatic protons indicating interactions between each<br />

cyclodextrin derivative and these areas of the drug molecule. Results of the solubility<br />

experiments showed a significant increase of itraconazole aqueous solubility. Particularly,<br />

the recently developed HBen-β-CD derivative could tremendously improve the solubility<br />

under gastric and small intestinal conditions.<br />

Based on the results of the present study, we assume that the interactions between each of<br />

the cyclodextrin derivatives and itraconazole lead to an improvement of the compounds<br />

solubility. However, the extent of this effect varied as a result of the derivative used for<br />

complex formation. Overall, it can be said, that the investigated cyclodextrin derivates were<br />

very useful to increase the solubility of itraconazole in compendial and biorelevant test<br />

media and therefore represent a promising group of excipients for future use in solubility<br />

(and bioavailability) enhancement of poorly soluble compounds.<br />

140 Poster


Autorenverzeichnis<br />

Abdel-Aziz H 98, 103, 137<br />

Abd-Ellatif A 132<br />

Abrahmsén-Alami S 32<br />

Abrahmsson B 32<br />

Abromeit H 102<br />

Abstiens K 80<br />

AbulAzm S 104<br />

Achenbach J 56<br />

Adams R H 114<br />

Adler M 137<br />

Ahmad K 113<br />

Ahmed M 82<br />

Alban S 58<br />

Albrecht M 122<br />

Al-Halhouli A T 130<br />

Alhazmi H 33<br />

Alhussein A 122<br />

Ali H 69<br />

Alresly Z 106<br />

Altenhofen W 117<br />

Altmann K S 118<br />

Ammon H P T 122<br />

Andermark V 111<br />

Anschütz M 32<br />

Arnold N 106<br />

Aßmann G 137<br />

Astner I 91<br />

Aurich K 134<br />

Baaske P 80<br />

Bäcker C 106<br />

Bakowsky U 70<br />

Banoglu E 117<br />

Bantscheff M 25<br />

Baranski J 115<br />

Bartusch A 136<br />

Barzen S 105<br />

Bauer J 102<br />

Bauer K 103<br />

Bauer S 103, 117<br />

Baumann K 79, 104, 125<br />

Bausch A 115<br />

Becker D 116<br />

Bednarski P J 65, 106, 118, 119<br />

Beese K 120<br />

Behrends S 130<br />

Behrendt C T 99<br />

Beißner N 101<br />

Beitz E 118<br />

Below A 110<br />

Bendas G 120, 121<br />

Berg S 43<br />

Bernat V 23, 105<br />

Bernhardt G 28, 98<br />

Bertram L 123<br />

Bertram N 35<br />

Bertsche T 87<br />

Besheer A 80<br />

Bettio S 122<br />

Bibb J A 114<br />

Bickmann D 110<br />

Biller A 137<br />

Bischoff F 100<br />

Bitterlich A 78, 124<br />

Bittner F 117<br />

Blackert S 134<br />

Block S 60<br />

Bloßfeld M 112<br />

Blume H 32, 134<br />

Bock R 126<br />

Bodem J 95, 96, 97<br />

Bodtke A 106, 110, 139<br />

Boeckler F M 55<br />

Bogan R 135<br />

Böheim K 139<br />

Borchert P 120<br />

Böttger S 107<br />

Bracher F 114<br />

Braig S 100<br />

Brand T 113<br />

Brandl F 86, 130<br />

Brandl F P 124<br />

Brandt S 60<br />

Brandt W 109<br />

Breitkreutz J 83, 110, 129, 137, 139<br />

Breunig M 133<br />

Brock D 131<br />

Brückner A 111<br />

Brückner C 102<br />

Brüßler J 70<br />

Bublitz K 120<br />

Bui T H 105<br />

Bundscherer L 118<br />

Bunjes H 75, 78, 124, 128, 130<br />

Buschauer A 28, 98<br />

Busker M 130<br />

Busse M 134<br />

Butterweck V 111<br />

Büttgenbach S 127, 130<br />

Chahar M 101<br />

Chamseddin C 105, 106<br />

Chbeib M 107<br />

Chen R 129<br />

Cheng X 63<br />

Cianciulli C 33, 110<br />

Claes D 108<br />

Clement B 117, 118<br />

Closs E I 115<br />

Collnot E M 69<br />

Crüsemann M 106<br />

Daily A 38<br />

Damm G 42<br />

Dannhardt G 115<br />

Dathe M 138<br />

Dayyoub E 70<br />

de Jong J C 100, 114<br />

Deally A 114<br />

Decker M 36<br />

Dehm F 101<br />

Delves M 98<br />

Dengler M 78<br />

Deuter A 119<br />

Dhein S 94<br />

Diel P 112<br />

Dietzel A 130<br />

Dolberg A M 116<br />

Donath F 32<br />

Dörje F 134<br />

Dorn A 101, 104<br />

Dos Santos Capelo R 121<br />

Dötsch V 119<br />

Duhr S 80<br />

Dzikowski R 61<br />

Eckert C 136<br />

Eeckman L 126<br />

Efferth T 37, 45<br />

Eiden K 119<br />

El Gaghlab K 120<br />

Elz S 102<br />

Emmrich T 65, 106, 118<br />

Engel A 43<br />

Engels B 95, 96<br />

Enzensperger C 103, 104<br />

Erdmann D 28<br />

Externbrink A 132<br />

Fahrmayr C 40<br />

Falck E 34, 122<br />

Ferandin Y 61<br />

Fersht A R 55<br />

Fey P 95<br />

Finke J H 127, 130<br />

Fischer D 73<br />

Fischer M 99<br />

Fischer S 104<br />

Fiß T 89, 137<br />

Flemming S 121<br />

Fleßa S 137<br />

Foerster S 122<br />

Förster F 100<br />

Fossen T 105<br />

Franzmann E 27<br />

Fréchet J M J 72<br />

Freichel M 48<br />

Freitag A 107<br />

Frentsch M 71<br />

Freyse E-J 43<br />

Friedrich C 135<br />

Frieß W 20, 81<br />

Fromm M F 40<br />

Fuchs S 97<br />

Fugel W 61<br />

Funke A 90<br />

Fürst R 97, 114, 115<br />

Gantzsch S 127<br />

Garbacz G 30, 109<br />

Gehringer M 95<br />

Gerber F 139<br />

Gerber U 120<br />

Germer K 131<br />

Gerstmeier J 117<br />

Geyer H 125<br />

Gieré R 74<br />

Gieseler H 122<br />

Glöckl G 113, 125, 134<br />

Gminski R 74<br />

Gobleder S 102<br />

Goebgen E 137<br />

Goettert M 103, 117<br />

Gohlke H 22<br />

Goos K-H 105<br />

Autorenverzeichnis 141


Göpferich A 86, 124, 130, 132, 133<br />

Görke K 109<br />

Gothsch T 127<br />

Grabow N 113, 126<br />

Graeser R 117<br />

Greinacher A 50, 59, 60<br />

Grez M 22<br />

Groll M 99<br />

Grotefend S 94<br />

Grün J 96<br />

Grüning B A 107<br />

Grützkau A 121<br />

Guhmann M 139<br />

Gündisch D 103<br />

Günther S 54, 107, 121<br />

Gust R 52<br />

Guthoff R 94<br />

Ha K 101<br />

Hackbarth C 60<br />

Hackenberg F 68<br />

Haertel B 119, 134<br />

Häfner A-K 119<br />

Hager B 114<br />

Hähn S 99<br />

Hahne T 33, 110<br />

Hamacher A 66<br />

Hamburger M 97<br />

Hammer E 122<br />

Hammer N 86, 124, 130<br />

Hammerschmidt S 60<br />

Hanke T 101<br />

Hanke U 116, 134<br />

Hansen F K 101<br />

Hansen S 31<br />

Harden D 131<br />

Harder C 126<br />

Hare E 69<br />

Härter A 111<br />

Hartmann R W 65, 100, 114<br />

Hartung E 97<br />

Hasenpusch D 107<br />

Haug K G 111<br />

Häupl T 121<br />

Haupt O 112<br />

Hausmann M 115<br />

Häussler S 100<br />

Haustein M 120<br />

Havemeyer A 117, 118<br />

Heilmann J 98<br />

Heinemann K 120<br />

Heininger A 139<br />

Held J 98, 99<br />

Heller E 25<br />

Hellmuth C 121<br />

Henn C 100<br />

Hennig L 111<br />

Hennig R 132, 133<br />

Hentschel-Humeida U 97<br />

Hessler N 73<br />

Hiddemann L 109<br />

Hinz B 111, 120<br />

Hipler U-C 73<br />

Hirt C 65<br />

Hochhaus G 111<br />

Hoffmann B 132<br />

Hoffmann C 25<br />

Hoffmann W 89, 137<br />

Hofmann B 105, 119<br />

Holl K 99<br />

Holloway S 95<br />

Holzgrabe U 25, 96, 103, 123<br />

Hornburger M C 115<br />

Hörold M 136<br />

Horvat M 129<br />

Hoser S 103, 137<br />

Humar M 54<br />

Iber S 134<br />

Ihling C 106<br />

Ihling C H 47<br />

Ilko D 123<br />

Illarionov B 99<br />

Jaax M 60<br />

Jaehde U 87<br />

Jäger C 54<br />

Jain A K 32<br />

Jakobs H 117<br />

Janas C 127<br />

Jansen R 138<br />

Jantscheff P 121<br />

Jauch J 122<br />

Jedlitschky G 50<br />

Jenett-Siems K 106<br />

Jenner D 96<br />

Jensen A A 103<br />

Jira T 105<br />

Joerger A C 55<br />

Juchum M 96<br />

Juli C 96<br />

Jung M 53, 112, 116, 120<br />

Junmahasathien T 135<br />

Kabisch J 109<br />

Kacprowski T 122<br />

Kage H 95<br />

Kahnt A S 121<br />

Kaminski D 125<br />

Kann B 127<br />

Karcher S 104<br />

Kaske M 28<br />

Kassack M U 66<br />

Katritzky A R 101<br />

Kaufeld A M 98<br />

Keck C M 108, 129, 131, 135<br />

Keck P R W E F 100<br />

Kees M 139<br />

Keiser M 42, 43<br />

Kelber O 98, 103, 137<br />

Keller M 28<br />

Kersten E 138<br />

Kesselring J 95<br />

Kessner S 136<br />

Khalil F 27, 123<br />

Kiefer T 65<br />

Kiefer W 96<br />

Kiene F 126<br />

Kipping T 136<br />

Kirchhof S 86, 124, 130<br />

Kirsch B 100<br />

Klages C-P 130<br />

Klein C 95<br />

Klein S 30, 132, 138, 139<br />

Kleinebudde P 128, 129, 132<br />

Kloft C 109, 136, 137, 139<br />

Klos S 103<br />

Knop K 139<br />

Knopke C 32<br />

Kobilka B K 23<br />

Koeberle A 100, 102<br />

Koeck J 129<br />

Kolb P 23, 105<br />

Kölbel K 47, 106<br />

Koletzko B 121<br />

Kölln C 112<br />

Kolodziej H 98, 99<br />

Komoß C 78, 124<br />

Könczöl M 74<br />

König J 40<br />

Konzuch S 99<br />

Kordaß B 125<br />

Korpis K 118<br />

Kortmann C 129<br />

Köster R 114<br />

Kovacevic A 108<br />

Koziolek M 30, 109<br />

Kraft K 137<br />

Kralisch D 73<br />

Krämer I 123<br />

Krauel K 60<br />

Kraus B 36<br />

Krauß A 115<br />

Krauth F 136<br />

Krebs S 134<br />

Kressirer C 100<br />

Kreutzer M F 95<br />

Kriemen E 120<br />

Kroemer H K 50<br />

Kruggel S 61, 109<br />

Kuehnl S 102<br />

Kühn J-P 134<br />

Kummer J 94<br />

Kumpfmüller J 109<br />

Kunfermann A 99<br />

Kunick C 61, 109<br />

Kupetz E 130<br />

Kurz T 98, 99<br />

Kwade A 78, 124, 127<br />

Laabs F 128<br />

Lalk M 106, 109<br />

Lally G 68<br />

Lamers C 56<br />

Lämmerhofer M 101<br />

Lamprecht A 126<br />

Lang C 26<br />

Lange C 106<br />

Laufer S 95, 103, 104, 117<br />

Laufer S A 100, 101, 107<br />

Lehmann J 103, 104<br />

Lehr C M 69, 127<br />

Lehr T 29<br />

Lemcke T 61, 107, 108, 109, 110<br />

Lemmerhirt H 65, 106, 118<br />

Lendlein A 71<br />

Leonard F 69<br />

Lestari M L A D 104<br />

Leuner K 134<br />

Leven M 98<br />

Liebeke M 46<br />

Liebl J 114<br />

Liedl K 21<br />

Liedtke A J 100<br />

Lienau C 99<br />

Liening S 101<br />

Lill A 105<br />

Lindequist U 106, 119, 134<br />

Link A 65, 106, 118, 120, 139<br />

Linnebacher M 120<br />

Lipke U 126<br />

Lipperheide C 126<br />

142 Autorenverzeichnis


Liu T 125<br />

Loch C 94<br />

Löhr J H 110<br />

Lohse M J 25<br />

Lu C 100<br />

Lucas X 107<br />

Luderer S 117<br />

Lukowski R 49, 115<br />

Lütnant I 98<br />

Maedler K 19<br />

Mai A 63<br />

Maison W 27, 108, 120, 123<br />

Majer M 115<br />

Manna C M 114<br />

Marschalek R 113<br />

Marschall P 137<br />

Martens A 139<br />

Martiné U 115<br />

Martz K E 101<br />

Marxer E 70<br />

Masch A 64<br />

Massing U 121<br />

Masur K 118<br />

Mathew S 71<br />

Matz F 97<br />

Matz M 79<br />

Maurer C K 100, 114<br />

Mayer B A 115<br />

Meijer L 61, 100, 109<br />

Mell N 69<br />

Melzig M F 94, 107, 129<br />

Mendel R 117<br />

Menrath C 97<br />

Menzen T 81<br />

Merfort I 54, 74<br />

Merkenschlager A 112<br />

Merkord J 120<br />

Mersch-Sundermann V 74<br />

Messmann V 86<br />

Meßmann V 124, 130<br />

Metz A 22<br />

Meyer A 111<br />

Meyerdierks N 115<br />

Michael S 98<br />

Michel A 119<br />

Minichmayr I 109<br />

Moll H 95<br />

Monbaliu J-C M 101<br />

Mordmüller B 98, 99<br />

Morgenstern O 118<br />

<strong>Moritz</strong> S 73<br />

Möschwitzer J P 104, 124, 125, 129<br />

Moscow J A 38<br />

Moser M 114<br />

Mosig J 132<br />

Müller A 73<br />

Müller C 114<br />

Müller F A 73<br />

Müller R 18, 100, 109<br />

Müller R H 104, 108, 124, 125, 129,<br />

131, 135<br />

Müller-Goymann C C 127, 130<br />

Mundt S 105, 110<br />

Münsterberg M 108<br />

Munteanu M 73<br />

Müsken M 100<br />

Muth F M 103<br />

Nagaraj M 82<br />

Nagel S 94, 109, 113, 126<br />

Nagel-Steger L 111<br />

Nauert C 137<br />

Neidhardt I 110<br />

Nett M 95<br />

Neumann M 84<br />

Nieber K 98, 103, 105, 112, 137<br />

Niedermeyer T H J 38, 110<br />

Niemann S 127<br />

Nikolenko H 138<br />

Nimtz M 105<br />

Northoff H 102<br />

Norville I H 96<br />

Norwig J 138<br />

Nowotny B 97<br />

Nüssler A 42<br />

Oak P 37<br />

Oberholzer A E 61<br />

Oehmigen K 134<br />

Oetjen E 107<br />

Okpanyi S N 137<br />

Olausson B E S 106<br />

Oli S 97<br />

Oliferenko A A 101<br />

Oswald S 41, 43<br />

Ott I 67, 111, 114<br />

Parr M K 112, 125<br />

Patel H 54, 107<br />

Patil S 68<br />

Paul M 125<br />

Pauly A 134<br />

Pein M 129, 136<br />

Peissner W 121<br />

Perfahl S 119<br />

Pergola C 117<br />

Pertz H H 98<br />

Peters K 111<br />

Petrovick G F 137<br />

Pettelkau J 106<br />

Pfeifer B 109<br />

Pham T L H 110<br />

Plouffe D 98<br />

Pluym N 28<br />

Poellinger N 139<br />

Pohl J 42<br />

Poll B 109<br />

Pollinger K 132, 133<br />

Popella S-D 101<br />

Porzel A 106<br />

Potterat O 97<br />

Pradel G 96<br />

Preisitsch M 110<br />

Pressburger N 61<br />

Pretor S 130<br />

Preu L 130<br />

Priese F 133<br />

Proschak E 56<br />

Puerstinger J 110<br />

Quintanilla-Fend L 122<br />

Quodbach J 128<br />

Quosdorf S J 99<br />

Radebold J 42<br />

Rakow T 119<br />

Ramer R 111, 120<br />

Raszek M 105<br />

Ratin M 61<br />

Rau O 56<br />

Rauch B 50<br />

Rauh D 62, 64<br />

Rauwald H J 111<br />

Rauwald H W 94<br />

Raynor A 121<br />

Redweik S 33, 94<br />

Reichl S 51, 101, 112, 116, 130<br />

Reichmann D 117<br />

Rein H 77, 136<br />

Reitz E 131<br />

Ren X 133<br />

Rethwilm A 97<br />

Retzar A 135<br />

Richter C 126, 127<br />

Rieger M 96<br />

Rischer M 85<br />

Ritter C 82<br />

Ritter C A 122<br />

Robaa D 103, 104<br />

Roch T 71<br />

Rockmann T 131<br />

Rödl C B 105<br />

Roessler C 64<br />

Rollinger J M 102<br />

Rösch P 96<br />

Rosenbaum D M 23<br />

Ross T 121<br />

Rothenhöfer M 98<br />

Rotili D 63<br />

Rotmann A 115<br />

Rumpf T 120<br />

Runge D 42<br />

Rustenbeck I 79<br />

Ruth P 115<br />

Salamon A 120<br />

Salazar J 124<br />

Sarkar-Tyson M 96<br />

Savtschenko A 94<br />

Schad C 95<br />

Schaefer U 31<br />

Schaefer U F 127<br />

Schaeftlein A 139<br />

Schäfer J 70<br />

Schaible A M 102<br />

Schanda J 22<br />

Schänzer W 125<br />

Scherer O 102<br />

Scherübl R 98<br />

Scheuch E 43<br />

Schiedel M 116<br />

Schirmeister T 95, 96, 97, 107<br />

Schlesinger M 121<br />

Schmidt D 23, 105<br />

Schmidt H 136<br />

Schmidt I 96<br />

Schmitz P 120<br />

Schmolke H 130<br />

Schmuhl E 111<br />

Schneider F 84<br />

Schneider G 56<br />

Schneider T 95, 96<br />

Schnittker C 126<br />

Schollmeyer D 100<br />

Schönherr D 116<br />

Schramek N 92<br />

Schröder G 105<br />

Schröder M 119<br />

Schröder T 106<br />

Schubeis T 82<br />

Schubert-Zsilavecz M 56, 101<br />

Schuchmann H P 129<br />

Schug B 32<br />

Autorenverzeichnis 143


Schulz K 134<br />

Schulz R 65, 106, 118<br />

Schulz S 37<br />

Schulz U 118<br />

Schulze M 103<br />

Schumacher K 79<br />

Schümmelfeder J 70<br />

Schur J 114<br />

Schurigt U 95, 96<br />

Schutkowski M 64<br />

Schütt R 113<br />

Schütze N 120<br />

Schwarz R 47<br />

Schweder T 109<br />

Schweimer K 96<br />

Scriba G K E 102<br />

Seeling A 110<br />

Seibel J 96<br />

Seidlitz A 94, 113, 126<br />

Semmling B 113, 126<br />

Seufert F 96<br />

Shaalan N 117<br />

Shehata A M 122<br />

Shoichet B K 23<br />

Siegmund W 42, 43, 116<br />

Siems K 106<br />

Sinha B 129<br />

Sinz A 34, 47, 106<br />

Smolka K 125<br />

Snitko M 95<br />

Söderlind E 32<br />

Sölter V 139<br />

Sotriffer C 95, 96, 97<br />

Sotriffer C A 107<br />

Spitzer G 21<br />

Stark H 105<br />

Staudacher V 114<br />

Steinbach A 100, 114<br />

Steinhilber D 56, 57, 105, 113, 119, 121<br />

Steinhoff B 137<br />

Steri R 56<br />

Sternberg K 113, 126<br />

Stintzing F C 39<br />

Storz M P 114<br />

Straßer A 24, 102<br />

Straubinger J 115<br />

Strauss A-C 134<br />

Streciwilk W 68<br />

Strunz A K 99<br />

Stuppner H 102<br />

Sutter A 90<br />

Swiatecka-Hagenbruch M 38<br />

Sydow K 138<br />

Tabares P 97<br />

Tacke M 68, 114<br />

Tackenberg M W 129<br />

Tajarobi F 32<br />

Talmann L 134<br />

Tänzler D 47<br />

Taraschewski A 138<br />

Taupitz T 139<br />

Tchoukouegno Ngueu S 112<br />

Tessmar J 132, 133<br />

Teßmar J 130<br />

Teßmar J K V 124<br />

Thiessen A 35<br />

Thom K 134<br />

Thommes M 76, 126, 129, 131<br />

Thürmann P A 88<br />

Tränkle C 103<br />

Trauner D 37<br />

Treuer E 117<br />

Trip E 126<br />

Tschammer N 23, 105<br />

Tschan S 98<br />

Tshuva E Y 114<br />

Ullrich A 42<br />

Venema K 113<br />

Viridé A 32<br />

Vissiennon C 105<br />

Vogel S 107<br />

Völker U 44, 122<br />

Vollmar A 100<br />

Vollmar A M 37, 97, 100, 114, 115<br />

von Schwarzenberg K 37, 100<br />

von Woedtke T 118, 119, 134<br />

Vongehr F 135<br />

Voß U 98<br />

Wacker M 127<br />

Wagner S 107<br />

Wagner T 112<br />

Waltenberger C 107<br />

Wanner G 37<br />

Wätzig H 33, 94, 110, 119<br />

Weber B 111<br />

Weber C 60<br />

Weber F 118<br />

Weber M 121<br />

Wedemeyer R-S 134<br />

Wegener J 115<br />

Weidel E 114<br />

Weiser D 98, 103<br />

Weitschies W 30, 32, 43, 84, 94,<br />

109, 113, 116, 125, 126, 134, 139<br />

Weiwad M 96<br />

Welinder A 32<br />

Welker A 95, 96, 97<br />

Wende K 105, 119<br />

Wendling F 97<br />

Wentsch H 104<br />

Wentzlaff M 126<br />

Wenzel R 133<br />

Wenzel S 109<br />

Werz O 100, 101, 102, 117<br />

Wesar F 73<br />

Wessjohann L A 106<br />

Wich P R 72<br />

Wicha S G 136, 137<br />

Wichmann C 22<br />

Wiedmann R M 37<br />

Wiegand C 73<br />

Wiest J 123<br />

Wilcken R 55<br />

Wilde F 106, 118<br />

Wilde L 125<br />

Willbold D 111<br />

Willer E A 97<br />

Wilson C G 113<br />

Windbergs M 127<br />

Winkelmann V 103<br />

Winter G 80<br />

Wischke C 71<br />

Witt S 136<br />

Wittmann H-J 24, 102<br />

Wolber G 21<br />

Wolf B 131, 133<br />

Wolf C 134<br />

Wolff H 36<br />

Wolff M 111<br />

Wollatz U 125<br />

Wollenhaupt S 104<br />

Wray V 105<br />

Wszelaki N 129<br />

Wu H 95<br />

Wünsch B 34, 97, 98, 99, 118, 122<br />

Wurster M 106<br />

Xu Y 33<br />

Zahler S 100, 114<br />

Zapf T 126<br />

Zhang S 114<br />

Zimmer A 82<br />

Zimmer C 100, 114<br />

Zimmermann T 135<br />

Zischka H 37<br />

Zlotos D P 103<br />

Zwernemann G 136<br />

144 Autorenverzeichnis


Notizen<br />

145


Notizen<br />

146


Notizen<br />

147


Notizen<br />

148


Notizen<br />

149


Notizen<br />

150


151


152<br />

<strong>Greifswald</strong> Innenstadt<br />

1 Tierpark<br />

2 Botanischer Garten<br />

3 Credneranlagen<br />

4 Amtsgericht/Finanzgericht<br />

5 Bahnhof<br />

6 Einrichtungen der <strong>Universität</strong><br />

7 Stadtbefestigung<br />

8 Kirche St. Jacobi<br />

9 Historische Sternwarte<br />

10 Rubenowplatz und –denkmal<br />

11 Einkaufszentrum Dompassage<br />

12 Hauptgebäude der <strong>Universität</strong><br />

13 Sozio-kulturelles Zentrum St. Spiritus<br />

14 Audimax<br />

15 Caspar-David-Friedrich-Zentrum<br />

16 Musikschule<br />

17 Alte <strong>Universität</strong>sbibliothek<br />

18 Dom St. Nikolai<br />

19 Museumshafen<br />

20 Kirche St. Joseph<br />

21 Fußgängerbrücke<br />

Geschäftführer und Leiter der Geschäftsstelle Apotheker Dr. Michael Stein<br />

<strong>DPhG</strong> Geschäftsstelle<br />

Hamburger Allee 26 - 28<br />

60486 Frankfurt/Main<br />

Tel.: 069-7191596-0<br />

Fax: 069-7191596-29<br />

Email: info@dphg.de<br />

http://www.dphg.de<br />

25.09.<strong>2012</strong><br />

22 Caspar-David-Friedrich-Denkmal<br />

23 Alfried Krupp Wissenschaftskolleg <strong>Greifswald</strong><br />

24 Volkshochschule<br />

25 Fischmarkt<br />

26 Rathaus<br />

27 Kunstwerkstätten<br />

28 Literaturzentrum „Wolfgang Koeppen“<br />

29 Stadtbibliothek<br />

30 Stadthaus (im Bau)<br />

31 Gotische Giebelhäuser<br />

32 Fangenturm<br />

33 Internationales Begegnungszentrum<br />

34 Kirche St. Marien<br />

35 Museumswerft<br />

36 Pommersches Landesmuseum<br />

37 Stadtarchiv<br />

38 Mensa<br />

39 Fallada-Geburtshaus<br />

40 Stadthalle/Theatercafé<br />

41 Landesarchiv Vorpommern<br />

42 Theater Vorpommern


Kartenmaterial zur Verfügung gestellt vom Amt für Wirtschaftsförderung


Kartenmaterial zur Verfügung gestellt vom Tourismusverband Vorpommern e.V., Layout von: Grafikagentur Progress 4<br />

1 Biotechnikum (Targets 11./12.10.)<br />

2 Center of Drug Absorption and Transport C_DAT<br />

3 Mathematisch-Naturwissenschaftliche Fakultät mit<br />

Institut für Pharmazie<br />

4 Leibniz-Institut für Plasmaforschung und Technologie<br />

(INP <strong>Greifswald</strong> e.V.)<br />

5 Zentrum für Zahn-, Mund- und Kieferheilkunde<br />

6 Institut für Biochemie (Moleküle 11./12.10.)<br />

7 Klinik und Poliklinik für Hals-, Nasen-, Ohrenkrankheiten,<br />

Kopf- und Halschirurgie<br />

8 Institut für Physik (Tabletten 11./12.10.)<br />

9 Arboretum des Botanischen Gartens<br />

10 <strong>Universität</strong>sbibliothek<br />

11 Klinik und Poliklinik für Hautkrankheiten<br />

12 Johannes-Kirche<br />

13 BDH-Klinik <strong>Greifswald</strong> GmbH<br />

14 Volksstadion<br />

15 Kleine Mensa<br />

16 <strong>Universität</strong>sklinikum <strong>Greifswald</strong><br />

mit Hörsaal Nord (13.10.)<br />

A EUROPA HOTEL <strong>Greifswald</strong>

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!