27.12.2012 Views

Munich - 17th Annual Meeting German Society for Gene Therapy ...

Munich - 17th Annual Meeting German Society for Gene Therapy ...

Munich - 17th Annual Meeting German Society for Gene Therapy ...

SHOW MORE
SHOW LESS

Create successful ePaper yourself

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

In cooperation with<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the<br />

<strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong><br />

(DG-GT e.V.)<br />

The <strong>German</strong> <strong>Society</strong> of Virology, Study-Group<br />

«Viral Vectors and <strong>Gene</strong> <strong>Therapy</strong>»<br />

The DFG Research Priority Program 1230<br />

«Mechanisms of <strong>Gene</strong> Vector Entry and Persistence»<br />

The British <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong><br />

Nanosystems Initiative <strong>Munich</strong><br />

Helmholtz Zentrum München<br />

October 7 – 9, 2010<br />

LMU Campus Großhadern<br />

<strong>Munich</strong>


1 |<br />

Imprint<br />

Editors:<br />

Manfred Ogris<br />

Verena Brand<br />

Pirmin Burth<br />

Cover Layout:<br />

Pirmin Burth<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong>


Table of Content<br />

Welcome 3<br />

<strong>Gene</strong>ral In<strong>for</strong>mation 4<br />

Organizing Committee 6<br />

Approach and Maps 6<br />

<strong>Meeting</strong> Program 9<br />

Sponsors 15<br />

Abstracts 16<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong><br />

| 2


3 |<br />

Welcome<br />

Dear members and friends of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong>,<br />

on behalf of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> we cordially welcome you to our 17 th <strong>Annual</strong><br />

<strong>Meeting</strong> held at the Chemistry and Pharmacy Campus of the University of <strong>Munich</strong>. The meeting is<br />

organized in conjunction with the British <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong>, the Study Group Viral Vectors of<br />

the <strong>German</strong> <strong>Society</strong> <strong>for</strong> Virology, the Research Priority Program SPP1230, the Nanosystems Initiative<br />

<strong>Munich</strong> (NIM) and the <strong>Munich</strong> Helmholtz <strong>Society</strong>.<br />

We have sought to put together an exciting and diversified program presenting state-of-the-art<br />

advances in the field of gene therapy, a field that has considerably evolved within recent years. The<br />

first day (Thursday, Oct., 7 th ) is dedicated to the Educational Sessions. Out of the portfolio of areas<br />

contributing to the progress of gene therapeutical approaches, we have chosen this year viral and<br />

non-viral vector design, tumor imaging and clinical aspects of gene therapy. The meeting is opened<br />

with an inaugural talk by our key note lecturer Mark Kay (Stan<strong>for</strong>d University) on Thursday evening<br />

and is continued on Friday and Saturday with internationally highly recognized experts in areas like<br />

vector development, cancer gene therapy, treatment of metabolic diseases and others. In addition,<br />

21 oral presentations were selected out of >100 submitted abstracts. During coffee- and lunch<br />

breaks, >60 posters are presented. Also the socializing part will be addressed accordingly with a<br />

Bavarian evening on Thursday offering ‘a saubere Brotzeit’ with classics from the Bavarian cuisine<br />

and brewery and a social event on Friday evening themed ‘Sports Night’. We hope that you will all<br />

enjoy the meeting and take the chance to discuss science and to relish <strong>Munich</strong>.<br />

Thank you all <strong>for</strong> coming to <strong>Munich</strong>!<br />

Manfred Ogris Hildegard Büning Anja Ehrhardt<br />

Andrew Baker Wolfgang Hammerschmidt Ernst Wagner<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong>


<strong>Gene</strong>ral In<strong>for</strong>mation<br />

Organization and contact<br />

PD Dr. Manfred Ogris<br />

Phone +49 (0)89 2180 77842<br />

Fax +49 (0)89 2180 77791<br />

Email: dg-gt2010@cup.uni-muenchen.de<br />

Butenandtstr. 5-13, 81377 München, <strong>German</strong>y<br />

Venue<br />

Ludwig-Maximilians-Universität München<br />

Dekanat der Fakultät für Chemie und Pharmazie<br />

Butenandtstr. 5-13<br />

81377 München<br />

Registration<br />

The registration desk in the foyer of house F is open on:<br />

Thursday October 7 th from 12:00 pm to 06:30 pm<br />

Friday October 8 th from 08:00 am to 06:30 pm<br />

Saturday October 9 th from 08:00 am to 02:00 pm<br />

The registration fee includes:<br />

- access to scientific sessions<br />

- program and abstract book<br />

- coffee/tea during the morning and afternoon breaks<br />

- lunch during the poster sessions at noon<br />

- Post Oktoberfest event<br />

Oral Presentations<br />

All presentations will take place in the Buchner lecture hall (see map). The lecture hall is equipped<br />

with computers (Windows, NOT Mac) and a projector <strong>for</strong> PowerPoint presentations. For those<br />

people who use Mac please bring your own notebook. For those who use Windows please bring<br />

your presentation on a memory stick.<br />

There is a possibility to prepare the presentations in the Liebig lecture hall (next to Buchner lecture<br />

hall). It’s equipped with the same computers and projectors <strong>for</strong> PowerPoint presentations<br />

The Liebig lecture hall is open:<br />

Thursday October 7 th from 01:00 pm to 06:30 pm<br />

Friday October 8 th from 08:00 am to 10 am, from 1 pm to 06:30 pm<br />

Saturday October 9 th from 08:00 am to 02:00 pm<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong><br />

| 4


5 |<br />

Posters<br />

The posters will be displayed in the foyer of house F (see map) from Friday 11:00 am to Saturday<br />

03:00 pm.<br />

Poster authors are present at their posters during poster sessions (see program).<br />

The posters can be handed over at the registration desk on Thursday or should be mounted on<br />

Friday until 10:30 am and can be removed on Saturday at 03:00 pm.<br />

Post Oktoberfest Event<br />

The Post Oktoberfest Event will take place in front of the Buchner lecture hall on Thursday, October<br />

7 th .<br />

Speakers Dinner<br />

The speakers dinner will take place at the Restaurant 181 on the top of the Olympiaturm. There will<br />

be a shuttle bus from the campus to the restaurant at 07:30 pm (return at 10:30 pm).<br />

Social Event “Sports Night”<br />

The social event is NOT included in the registration fee. Please wear your name badge!<br />

The social event will take place at Primafila in Laim (near Sportpark Nymphenburg) on Friday,<br />

October 8 th , 08:30 pm.<br />

Approach:<br />

There will be a shuttle bus from the campus to the restaurant at 08:00 pm.<br />

By public transport:<br />

From the campus take the subway U6 in the direction of Garching Forschungszentrum and get off at<br />

Holzapfelkreuth. Then take bus 51 in the direction of Olympia Einkaufszentrum and get off at Laim.<br />

Pass the underground passage and follow Wotanstraße, then turn left and walk along Margarethe-<br />

Danzi-Straße until you reach the Sportpark Nymphenburg and Primafila.<br />

You can also go there by S-Bahn and get off at Laim (see map).<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong>


Organizing Committee<br />

PD Dr Manfred Ogris<br />

PD Dr Hildegard Büning<br />

PD Dr Anja Ehrhardt<br />

Prof Dr Andrew Baker<br />

Prof Dr Wolfgang Hammerschmidt<br />

Prof Dr Ernst Wagner<br />

Approach and Maps<br />

How to reach us by car<br />

• From the Nürnberg highway: get onto the Mittlerer Ring (direction Autobahn Lindau), then<br />

onto the Lindau highway to the exit Blumenau, keep going to Gräfelfing from where<br />

Würmtalstrasse will lead you to our campus.<br />

• From the Stuttgart highway: from the end of the highway in Obermenzing turn off to Pasing,<br />

from Pasing drive in the direction of Gräfelfing, then turn left to Großhadern.<br />

• From the Salzburg or Garmisch highways: drive onto the "Mittlerer Ring" in the direction of<br />

Großhadern and Stuttgart, then, in Großhadern turn to Gräfelfing and follow Würmtalstrasse<br />

which passes by our campus.<br />

Destination address <strong>for</strong> navigation system and parking<br />

Marchioninistraße, 81377 München<br />

Follow the parking signs at “Klinikum Großhadern”<br />

By train and public transport<br />

By IC, EC, ICE to <strong>Munich</strong> Central Station (München Hauptbahnhof). From there, take the subway U1<br />

or U2 to Sendlinger Tor. Change to U6 in the direction of Klinikum Großhadern and get off at<br />

Großhadern. Take the stairway to your left and keep left. You are now on Würmtalstraße. Now you<br />

can walk along Würmtalstraße until the campus comes into view (large modern buildings) on the left<br />

(about 10 to 15 minutes) or take the bus 266 or 268 in the direction of Planegg to Waldhüterstraße<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong><br />

| 6


7 |<br />

By plane and public transport<br />

From the <strong>Munich</strong> airport take the S-Bahn S8 (or S1) to Marienplatz. Change to the subway U6 in the<br />

direction of Klinikum Großhadern and get off at Großhadern. Take the stairway to your left and keep<br />

left. You are now on Würmtalstraße. Now you can walk along Würmtalstraße until the campus<br />

comes into view (large modern buildings) on the left (about 10 to 15 minutes) or take the bus 266 or<br />

268 in the direction of Planegg to Waldhüterstraße.<br />

Ticketing <strong>for</strong> public transport<br />

You can use:<br />

- MVV-3-days ticket <strong>for</strong> the inner district (=white zone) from the moment of validation until 6<br />

am on the fourth day (3-Tages-Ticket Innenraum, 12,80€).<br />

- MVV-1-day ticket <strong>for</strong> the inner district (=white zone) from the moment of validation until 6<br />

am the following day (1-Tages-Ticket Innenraum, 5,20€).<br />

All tickets must be validated once in a blue ticket-cancelling machine be<strong>for</strong>e starting with your trip,<br />

except <strong>for</strong> tickets from the bus and tram ticket machines. These are already validated.<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong>


<strong>Meeting</strong> Program<br />

Educational Session<br />

THURSDAY OCTOBER 7<br />

13:00 – 14:30 Educational Session Part 1<br />

Session Chair: Christian Kupatt, Klinikum Großhadern, LMU <strong>Munich</strong><br />

• Inv 1 Christina Rauschhuber (Department of Virology, Max von Pettenkofer-Institute,<br />

<strong>Munich</strong>, <strong>German</strong>y)<br />

Design of Recombinant Adenoviral Vectors <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong>: Improvements and<br />

Challenges<br />

• Inv 2 Martina Anton (Klinikum rechts der Isar, TUM, <strong>Munich</strong>, <strong>German</strong>y)<br />

Lentiviral vectors in gene and cell therapy approaches<br />

• Inv 3 Christian Kupatt (Klinikum Großhadern, LMU, <strong>Munich</strong>, <strong>German</strong>y)<br />

AAV based vectors <strong>for</strong> cardiovascular diseases<br />

14:30 – 15:00 Coffee break<br />

15:00 – 17:00 Educational Session Part 2<br />

Session Chair: Carsten Rudolph, Von Haunersches Kinderspital, LMU, <strong>Munich</strong><br />

• Inv 4 Carsten Rudolph (Von Haunersches Kinderspital, LMU, <strong>Munich</strong>)<br />

Introduction into nonviral gene delivery – physical and chemical delivery methods<br />

• Inv 5 Christine Spitzweg (Klinikum Großhadern, LMU, <strong>Munich</strong>)<br />

Imaging in cancer gene therapy<br />

• Inv 6 Len Seymour (University of Ox<strong>for</strong>d, UK)<br />

<strong>Gene</strong> therapy around the globe<br />

• Inv 7 Josef Rosenecker (Von Haunersches Kinderspital, LMU, <strong>Munich</strong>)<br />

Clinical experience with gene therapy in pediatric patients<br />

17:00 – 18:00 Coffee break<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong><br />

| 8


9 |<br />

<strong>Gene</strong>ral <strong>Meeting</strong><br />

18:00 – 18:15 Opening Remarks and Opening ceremony<br />

18:15 – 19:05 Keynote Lecture<br />

Session Chair: Hildegard Büning, University of Cologne<br />

• Inv 8 Mark Kay (School of Medicine, Stan<strong>for</strong>d University, USA)<br />

<strong>Gene</strong> transfer approaches <strong>for</strong> gene addition, knockdown and cellular reprogramming<br />

in vivo<br />

19:05 – 20:30 Post Oktoberfest Event<br />

20:30 – 22:30 Speakers dinner<br />

FRIDAY OCTOBER 8<br />

9:00 – 11:00 Vector Development<br />

Session Chairs: Florian Kreppel, University of Ulm<br />

Ernst Wagner, Department of Pharmacy, LMU, <strong>Munich</strong><br />

• Inv 9 Len Seymour (University of Ox<strong>for</strong>d, UK)<br />

Delivery issues <strong>for</strong> oncolytic viruses<br />

• Inv 10 Andrew Baker (University of Glasgow, Scotland, UK)<br />

Modification of the adenovirus capsid: integrating virus biology and vector<br />

engineering<br />

• Selected Talk from abstracts:<br />

Or 1: Sigrid Espenlaub (Department of <strong>Gene</strong> <strong>Therapy</strong>, University of Ulm, <strong>German</strong>y)<br />

Analysis of intracellular particle trafficking and bioresponsive bonds <strong>for</strong> Ad vector<br />

shielding by capsomer specific fluorescent labeling<br />

Or 2: Jessica Sallach (Clinic I of Internal Medicine and Center <strong>for</strong> Molecular Medicine<br />

Cologne, University of Cologne, <strong>German</strong>y)<br />

Primary human keratinocyte-selective AAV2-based targeting vectors<br />

Or 3: Nadja Noske (Department of Virology, Max von Pettenkofer-Institute, LMU, <strong>Munich</strong>,<br />

<strong>German</strong>y)<br />

Development of novel PhiC31 integrase fusion proteins <strong>for</strong> improving efficacy and<br />

safety of transgene insertion in therapeutic applications<br />

Or 4: Keiji Itaka (Division of Clinical Biotechnology, Graduate School of Medicine, The<br />

University of Tokyo, Japan)<br />

Biocompatible polyplex nanomicelle <strong>for</strong> safe and effective gene transfer<br />

11:00 – 11:15 Coffee Break and Poster Session<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong>


11:15 – 12:00 Vector Development<br />

• Inv 11 Michael Gait (MRC Laboratory of Molecular Biology, Cambridge, UK)<br />

Peptide-enhanced delivery of oligonucleotide analogues targeting Duchenne<br />

muscular dystrophy<br />

• Selected Talk from abstracts:<br />

Or 5: Frauke M. Koenig (Department of Chemistry, LMU, <strong>Munich</strong>, <strong>German</strong>y)<br />

EGF receptor targeting of polyplexes with the short artificial peptide GE11 studied by<br />

live cell imaging<br />

12:00 – 13:30 On the Route to Clinical Application<br />

Session Chair: Christoph von Kalle, National Center <strong>for</strong> Tumor Diseases , Heidelberg<br />

• Inv 12 Marinee Chuah (Vesalius Research Center, Leuven, Belgium)<br />

Emerging transposon technology <strong>for</strong> gene transfer and IPS applications<br />

• Inv 13 Simon Waddington (Imperial College London, UK)<br />

Perinatal gene therapy <strong>for</strong> lethal genetic diseases<br />

• Selected Talk from abstracts:<br />

Or 6: Stylianos Michalakis (Department of Pharmacy – Center <strong>for</strong> Drug Research, LMU,<br />

<strong>Munich</strong>, <strong>German</strong>y)<br />

Restoration of cone vision in the CNGA3–/– mouse model of congenital complete lack<br />

of cone photoreceptor function<br />

Or 7: Anna Paruzynski (Department of Translational Oncology, National Center <strong>for</strong> Tumor<br />

Diseases (NCT) and <strong>German</strong> Cancer Research Center (DKFZ), Hannover, <strong>German</strong>y)<br />

High throughput integration site analysis reveals a polyclonal lineage-specific<br />

integration site distribution in a successful WAS gene therapy trial<br />

13:30 – 14:30 Lunch Break and Poster Session<br />

14:30 – 16:00 <strong>Gene</strong> <strong>Therapy</strong><br />

Session Chairs: Boris Fehse, University Medical Centre Hamburg-Eppendorf<br />

Manuel Grez, Georg-Speyer-Haus, Frankfurt<br />

• Inv 14 Robin Ali (Institute of Ophthalmology, London, UK)<br />

<strong>Gene</strong> therapy <strong>for</strong> inherited retinal dystrophies<br />

• Inv 15 Harald Petry (Amsterdam Molecular Therapeutics (AMT), Netherlands)<br />

Alipogene Tiparvovec: the first gene therapy <strong>for</strong> a general metabolic disorder<br />

• Selected Talk from abstracts:<br />

Or 8: Teresa Trenkwalder (Klinikum Großhadern, LMU, <strong>Munich</strong>, <strong>German</strong>y)<br />

Enhanced therapeutic neovascularization via AAV2.9/Thymosin β4: Evidence <strong>for</strong> a<br />

Myovascular Crosstalk<br />

Or 9: Abdullah Cim (King’s College London, School of Medicine, London, UK)<br />

Nonviral Delivery of the rat PDX1 gene to rat liver <strong>for</strong> the in vivo transdifferentiation<br />

of liver cells to pancreatic β-cells<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong><br />

| 10


11 |<br />

16:00 – 16:30 Coffee Break and Poster Session<br />

16:30 – 17:15 <strong>Gene</strong> <strong>Therapy</strong><br />

• Inv 16 Manuel Grez (Georg-Speyer-Haus, Frankfurt, <strong>German</strong>y)<br />

<strong>Gene</strong> therapy of chronic granulomatous disease: the past and the future<br />

• Selected Talk from abstracts:<br />

Or 10: Ute Modlich (Hannover Medical School, Hannover, <strong>German</strong>y)<br />

Correction of Mpl deficiency by lentiviral vectors with lineage-specific expression<br />

17:15 – 18:45 Cancer <strong>Gene</strong> <strong>Therapy</strong><br />

Session Chairs: Dorothee von Laer, Innsbruck Medical University<br />

Dirk Nettelbeck, DKFZ Heidelberg<br />

• Inv 17 Roberto Cattaneo (Mayo Clinic, Minnesota, USA)<br />

Viruses as cancer therapeutics: three points of attack<br />

• Inv 18 Caroline Breitbach (Jennerex, Inc., San Francisco, USA)<br />

Targeted and armed oncolytic poxviruses: a novel multi-mechanistic<br />

therapeutic class <strong>for</strong> cancer<br />

• Selected Talk from abstracts:<br />

Or 11: Alexander Muik (Georg-Speyer-Haus, Frankfurt am Main, <strong>German</strong>y)<br />

LCMV-Pseudotyped VSV-based systems <strong>for</strong> treatment of malignant glioma<br />

Or 12: Jennifer Altomonte (Klinikum rechts der Isar, TU München, <strong>Munich</strong>, <strong>German</strong>y)<br />

18:45 – 19:00 Award Ceremony DG-GT Forscherpreis<br />

19:00 – 20:00 DGGT <strong>Gene</strong>ral Assembly<br />

20:30 – open end Social Event themed “Sports Night”<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong>


SATURDAY OCTOBER 9<br />

9:00 – 9:45 Cancer <strong>Gene</strong> <strong>Therapy</strong><br />

• Inv 19 John Bell (Ottawa Hospital Research Institute, Canada)<br />

Oncolytic vaccinia virus <strong>for</strong> the treatment of cancer<br />

• Selected Talk from abstracts:<br />

Or 13: Geoffrey K. Grünwald (Klinikum Grosshadern, LMU, <strong>Munich</strong>, <strong>German</strong>y)<br />

An α-fetoprotein promoter driven, conditionally replicating adenovirus that expresses<br />

the sodium iodide symporter (NIS) <strong>for</strong> radiovirotherapy of HCC<br />

9:45 – 10:45 Tumor Biology and (Cancer) Stem Cells<br />

Session Chair: Axel Schambach, Hannover Medical School<br />

• Inv 20 Norman Maitland (YCR Cancer Research Unit, University of York, UK)<br />

Prostate cancer stem cells: a new target <strong>for</strong> therapy<br />

• Selected Talk from abstracts:<br />

Or 14: Kerstin Knoop (Klinikum Grosshadern, LMU, <strong>Munich</strong>, <strong>German</strong>y)<br />

Tumor stroma-specific NIS gene delivery using mesenchymal stem cells<br />

Or 15: Axel Schambach (Department of Experimental Hematology, Hannover Medical<br />

School, Hannover, <strong>German</strong>y)<br />

Monitoring and excising reprogramming factors: a novel lentiviral expression system<br />

<strong>for</strong> reprogramming strategies<br />

10:45 – 11:15 Coffee Break and Poster Session<br />

11:15 – 13:15 Cancer Immune <strong>Therapy</strong> and T-cell <strong>Therapy</strong><br />

Session Chair: Wolfgang Uckert, Max Delbrück Center <strong>for</strong> Molecular Medicine, Berlin<br />

• Inv 21 Renata Stripecke (Hannover Medical School, <strong>German</strong>y)<br />

Lentiviral vector-induced dendritic cells <strong>for</strong> melanoma immunotherapy<br />

• Inv 22 Hinrich Abken (Uniklinikum Köln, <strong>German</strong>y)<br />

Arming immune cells to fight cancer<br />

• Inv 23 Farzin Farzaneh (Kings College, London, UK)<br />

Immune gene therapy <strong>for</strong> acute myeloid leukaemia<br />

• Selected Talk from abstracts:<br />

Or 16: Matthias Leisegang (Max-Delbrück-Center <strong>for</strong> Molecular Medicine, Berlin, <strong>German</strong>y)<br />

MHC-restricted fratricide of recipient lymphocytes expressing transgenic T-cell<br />

receptors specific <strong>for</strong> the apoptosis-inhibitor protein survivin<br />

Or 17: Eliana Ruggiero (<strong>German</strong> Cancer Research Center and National Center <strong>for</strong> Tumor<br />

Diseases, Heidelberg, <strong>German</strong>y)<br />

Integration site analysis of reprogrammed T-cells in a mouse model of T-cell receptor<br />

gene therapy developing graft versus host disease<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong><br />

| 12


13 |<br />

13:15 – 14:00 Lunch Break and Poster Session<br />

14:00 – 16:00 Pharmacology and Toxicology<br />

Session Chair: Hildegard Büning, University of Cologne<br />

• Inv 24 Anne Galy (<strong>Gene</strong>thon, Evry, France)<br />

Preclinical safety and efficacy data leading to a clinical trial <strong>for</strong> the gene therapy of<br />

Wiskott Aldrich Syndrome<br />

• Inv 25 Klaus Cichutek (Paul-Ehrlich-Institut, Langen, <strong>German</strong>y)<br />

Lentivector transduction of novel cell targets<br />

• Selected Talk from abstracts:<br />

Or 18: Katarina Farkasova (Pharmaceutical Biotechnology, LMU, <strong>Munich</strong>, <strong>German</strong>y)<br />

Luciferase-based dual bioluminescence imaging of tumor metastases after systemic<br />

transgene delivery with a synthetic gene carrier<br />

Or 19: Niels Heinz (Experimental Hematology, Hannover Medical School, Hannover,<br />

<strong>German</strong>y)<br />

Retroviral and transposon-based Tet-regulated all-in-one vectors with reduced<br />

background expression and improved dynamic range<br />

Or 20: Simone J. Scholz (<strong>German</strong> Cancer Research Center and National Center <strong>for</strong> Tumor<br />

Diseases, Heidelberg, <strong>German</strong>y)<br />

High-throughput integration site analysis <strong>for</strong> vector biosafety assessment in CGD<br />

gene therapy<br />

Or 21: Margaret R. Duffy (Glasgow Cardiovascular Research Centre, University of Glasgow,<br />

Glasgow, UK)<br />

Modification of the FX serine protease domain ablates HSPG engagement by Ad5-FX<br />

complexes<br />

16:00 – 16:15 Poster Award and Concluding Remarks<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong>


Sponsors<br />

Gold Sponsors Silver Sponsors<br />

Thanks <strong>for</strong> the support to our sponsors!<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong><br />

| 14


15 |<br />

Abstracts<br />

Invited presentations 16 - 27<br />

Oral presentations 27 - 40<br />

Poster presentations 41 - 78<br />

Abstract author index 79 - 82<br />

All abstract are published in the September 2010 issue of Human <strong>Gene</strong> <strong>Therapy</strong> and can be<br />

downloaded via this link: http://www.liebertonline.com/doi/abs/10.1089/hum.2010.804<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong>


DG-GT 2010 Invited Presentations<br />

Inv 1<br />

Design of Recombinant Adenoviral Vectors<br />

<strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong>: Improvements and<br />

Challenges<br />

Christina Rauschhuber<br />

Department of Virology, Max von Pettenkofer-<br />

Institute, <strong>Munich</strong>, <strong>German</strong>y<br />

Over the past decade recombinant adenoviral<br />

vectors (rAdVs) became one of the most<br />

prominent gene therapeutic vector systems used<br />

in preclinical and clinical approaches. Adenoviral<br />

vectors exhibit several targets <strong>for</strong> modifications,<br />

thus they are useful <strong>for</strong> a variety of applications.<br />

Different generations of rAdV based on human<br />

adenovirus serotype 5 were generated starting<br />

with first and second generation adenoviruses,<br />

which lack one or two viral genes to more<br />

sophisticated technologies such as high capacity<br />

adenoviral vectors (HD-Ad) deleted <strong>for</strong> all viral<br />

coding sequences. Besides these types of<br />

vectors which will be discussed in detail,<br />

oncolytic adenoviruses exhibiting restricted<br />

replication in tumor tissues have to be mentioned<br />

but are not the major focus of this lecture.<br />

Common <strong>for</strong> all rAdV is their ability to efficiently<br />

transduce a broad range of dividing and nondividing<br />

cells and that they can be produced at<br />

high titers. However, in contrast to first and<br />

second generation adenoviruses, HD-AdVs have<br />

a significantly larger packaging capacity <strong>for</strong> large<br />

trangene expression cassettes and they display<br />

long-term transgene expression in preclinical<br />

studies. Nevertheless, the biggest challenge in<br />

adenoviral gene therapy is to circumvent the<br />

immune response against the vector. As HD-AdV<br />

lacks all viral coding sequences, there is no<br />

response to de novo synthesized viral proteins<br />

but immune response directed against the<br />

incoming viral capsid components may impair the<br />

outcome of a gene therapeutic approach. Thus,<br />

improvements with respect to the administration<br />

route and methods to modify the surface of the<br />

virion are the major focus of ongoing research<br />

and will be discussed within this lecture.<br />

Session: Educational Session<br />

Inv 2<br />

Lentiviral Vectors in <strong>Gene</strong> and Cell <strong>Therapy</strong><br />

Approaches<br />

Martina Anton<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong><br />

| 16<br />

Institute of Experimental Oncology and <strong>Therapy</strong><br />

Research, Technische Universität München,<br />

<strong>Munich</strong>, <strong>German</strong>y<br />

Retroviruses are evolutionally optimized <strong>for</strong><br />

transfer of their genetic material into cells and<br />

integrating their genome into the host cell<br />

genome. They are thus well suited <strong>for</strong> long-term<br />

stable gene expression. Recombinant vectors<br />

based on gammaretroviruses (MLV) have been<br />

widely used in the past in preclinical research as<br />

well as clinical trials. However, during clinical<br />

trials of SCID, severe side effects occurred<br />

leading to induction of leukaemia in five cases.<br />

More recently researchers have tried to harness<br />

lentiviruses, another genus of the retroviridae and<br />

developed recombinant vectors based on Human<br />

Immunodeficiency Virus (HIV-1), Simian<br />

Immunodeficiency Virus (SIV), Feline (FIV)<br />

Immunodeficiency Virus, equine infectious<br />

anaemia virus (EIAV) and others. Whereas<br />

commonly used gammaretroviruses like MLV<br />

depend on cell division and breakdown of the<br />

nuclear membrane, lentiviruses and their vectors<br />

can also infect and transduce non-proliferating<br />

cells. The general concepts on how these<br />

pathogens can be converted into efficient and<br />

safe gene delivery tools <strong>for</strong> cell modification, the<br />

correction of inherited or acquired diseases will<br />

be introduced. Packaging concepts and targeting<br />

of lentiviral vectors (LV) will be discussed, as well<br />

as self inactivating (SIN) LV that are thought to<br />

be less likely to activate or disrupt neighbouring<br />

genes upon integration. Additionally important<br />

areas of research focus on how lentiviral vectors<br />

can be modified to avoid integration and thus<br />

reduce risk of insertional mutagenesis or to target<br />

integration to specific sites in the genome. An<br />

overview on construction and use of lentiviral<br />

vectors as tools in molecular biology,<br />

transcriptional targeting and de-targeting of LV,<br />

regulation of LV-mediated gene expression, preclinical<br />

as well as recent and future clinical<br />

applications will be presented.<br />

Session: Educational Session


17 |<br />

Inv 3<br />

AAV-Based Vectors <strong>for</strong> Cardiovascular<br />

Diseases: Therapeutic Potential in Chronic<br />

Ischemia<br />

C. Kupatt, R. Hinkel<br />

Klinikum Großhadern, Medizinische Klinik I und<br />

Poliklinik, Ludwig-Maximilians-University <strong>Munich</strong>,<br />

<strong>Munich</strong>, <strong>German</strong>y<br />

Therapeutic neovascularization of chronic<br />

ischemic muscle tissue is a treatment option <strong>for</strong><br />

otherwise no-option patients, which are not<br />

suitable <strong>for</strong> interventional or surgical<br />

interventions. Protein application of proangiogenic<br />

factors, such as VEGF or bFGF<br />

provided vessel growth and perfusion gains in<br />

small and large animal studies. However, in<br />

patient studies no clearcut success was achieved<br />

by pro-angiogenic protein application. Similar<br />

discrepancies were idetified <strong>for</strong> plasmid therapy,<br />

e.g., by liposomal transfection: significant<br />

improvement in preclinical experiments was<br />

followed by mixed results in human. Even<br />

overexpression of pro-angiogenic genes via<br />

adenoviral vector, leading to a transgene<br />

overexpression <strong>for</strong> 5-7 days, failed to significantly<br />

improve perfusion of an ischemic myocardium<br />

(Yla-Herttuala, 2007, Lavu JMCC 2010) Since<br />

prolonged transgene expression currently<br />

appears as a critical variable <strong>for</strong> pro-angiogenic<br />

gene therapy, long-acting adeno associated viral<br />

vectors might no-option patients. Adenoassociated<br />

virus (AAV), a member of the<br />

parvovirus family, is a non-pathogenic DNA virus,<br />

which transduces dividing and non-dividing cells<br />

and leading to a long-term overexpression,<br />

displaying a favorable immunogenic profile<br />

compared to adenoviral vectors (Gruchala,<br />

2004). Recently, we identified the pseudotyped<br />

virus strains AAV2/6 (carrying a partial genome<br />

of AAV2 and the envelope of AAV6) and AAV2/9<br />

as highly efficient vectors in large animals (pigs,<br />

Raake et al., JACC 2008, Kupatt et al., JACC<br />

2010), the latter sufficing to resolve hibernating<br />

myocardium after chronic coronary artery<br />

occlusion. Moreover, the long-lasting transgene<br />

expression after AAV transduction may require<br />

inducible transgene activity. We established the<br />

combination of AAV vectors and a Tet-off<br />

transgene system, where the pro-angiogenic<br />

factor Thymosin β4 was used to provide<br />

neovascularization being active only 2d a week.<br />

In a model of chronic hindlimb ischemia, pulsed<br />

pro-angiogenic activity appeared non-inferior to<br />

constitutive transgene expression up to 4 weeks<br />

after vector application. Taken together, the<br />

combination of of AAV vector and an efficient<br />

pro-angiogenic transgene may offer therapeutic<br />

potential in patients with ischemic<br />

cardiomyopathy or peripheral artery disease and<br />

exhausted conventional options.<br />

Session: Educational Session<br />

Inv 4<br />

Introduction into Nonviral <strong>Gene</strong> Delivery –<br />

Overview of Physical and Chemical Delivery<br />

Methods<br />

Carsten Rudolph<br />

Ludwig Maximilians University, <strong>Munich</strong>, <strong>German</strong>y<br />

Physicochemical methods <strong>for</strong> gene delivery have<br />

been intensively investigated during the last<br />

decades and first products have been approved<br />

<strong>for</strong> veterinary use. Although transfection rates<br />

achieved with nonviral delivery systems are<br />

frequently lower and gene expression is of only<br />

short duration when compared with viral vectors,<br />

they offer the advantage of being less<br />

immunogenic, less restricted to the size of the<br />

delivered transgene and being less expensive<br />

with respect to production. The currently used<br />

repertoire of physicochemical gene delivery<br />

methods have evolved from various basic<br />

concept using either naked DNA or complexed<br />

with gene transfer agents. Moreover, a variety of<br />

methods have been established which make use<br />

of different physical phenomena to introduce<br />

DNA into cells. In this lecture, basic concepts of<br />

various physicochemical gene delivery methods<br />

making use of either nanoparticle <strong>for</strong>mation or<br />

physical <strong>for</strong>ces <strong>for</strong> transfection will be discussed.<br />

In addition, advantages and restrictions of the<br />

physicochemical gene delivery methods will be<br />

discussed.<br />

Session: Educational Session<br />

Inv 5<br />

Imaging in Cancer <strong>Gene</strong> <strong>Therapy</strong><br />

Christine Spitzweg<br />

Department of Internal Medicine II, Klinikum<br />

Großhadern, Ludwig Maximilians University,<br />

<strong>Munich</strong>, <strong>German</strong>y<br />

The field of gene therapy has made considerable<br />

strides in the last decade by the development of<br />

new vectors and an increasing repertoire of<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong>


therapeutic genes. Non-invasive monitoring of<br />

the in vivo distribution of viral and non-viral<br />

vectors as well as biodistribution, level and<br />

duration of transgene expression have been<br />

recognized as critical elements in the design of<br />

clinical gene therapy trials. Besides<br />

bioluminescence imaging using luciferase,<br />

reporter genes that have been evaluated <strong>for</strong><br />

possible human studies are the herpes simplex<br />

thymidine kinase and the dopamine D2 receptor.<br />

Cloning of the sodium iodide symporter (NIS),<br />

that mediates the active transport of iodide in the<br />

thyroid gland and represents the molecular basis<br />

<strong>for</strong> radioiodine scintigraphy, has provided us with<br />

one of the most promising reporter genes<br />

available today. NIS represents a nonimmunogenic<br />

protein with a well-defined body<br />

distribution that mediates the transport of readily<br />

available radionuclides such as 131 I, 123 I, 124 I,<br />

99m 188 211<br />

Tc, Re or At, which can be used <strong>for</strong><br />

gamma camera scintigraphic imaging, SPECT<br />

and PET imaging. Several research groups<br />

including our own have studied the potential of<br />

NIS as reporter gene in various applications,<br />

demonstrating that in vivo imaging of radioiodine<br />

accumulation correlates well with the results of ex<br />

vivo gamma counter measurements as well as<br />

NIS mRNA and protein analysis. NIS was<br />

successfully used to monitor in vivo<br />

biodistribution of synthetic vectors and<br />

mesenchymal stem cells as gene delivery<br />

vehicles after systemic application as well as<br />

replication-competent viral vectors using<br />

123 99m<br />

conventional I- or Tc-gamma camera<br />

imaging or 99m Tc-SPECT/CT fusion imaging. In<br />

addition, PET imaging using<br />

124 I provides<br />

significant advantages <strong>for</strong> exact localization and<br />

quantitative analysis of NIS-mediated radioiodine<br />

accumulation due to enhanced resolution and<br />

sensitivity. Taken together, currently available<br />

data clearly demonstrate the enormous potential<br />

of NIS as a novel reporter gene, that in its dual<br />

function as reporter and therapy gene also allows<br />

therapeutic radionuclide application.<br />

Session: Educational Session<br />

Inv 6<br />

<strong>Gene</strong> <strong>Therapy</strong> Around the Globe<br />

Len Seymour<br />

Department of Clinical Pharmacology, University<br />

of Ox<strong>for</strong>d, Ox<strong>for</strong>d, UK<br />

<strong>Gene</strong> therapy is a whole new approach to<br />

medicine. Instead of designing drugs to treat the<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong><br />

| 18<br />

symptoms of a disease, gene therapy seeks to<br />

identify the root genetic cause of a disorder and<br />

to treat it at that level. For diseases caused by<br />

mutations in ‘single genes’, this provides the<br />

possibility of treating the disease very effectively<br />

by providing healthy copies of the mutant gene,<br />

allowing production of normal proteins and<br />

restoring the healthy phenotype. Accordingly this<br />

approach has exciting potential <strong>for</strong> treatment of<br />

many gene-based diseases. Despite its promise,<br />

however, gene therapy is often limited by the<br />

difficulty of delivering therapeutic genes<br />

effectively into the diseased cells. Normally<br />

scientists try to do this using viruses as genevectors;<br />

however, even viruses find it challenging<br />

to access all diseased cells within a body. This is<br />

the central challenge of gene therapy. Where<br />

delivery has been successfully addressed, the<br />

results have been very encouraging. For<br />

example, children with bone marrow-based<br />

immune deficiencies can now be treated very<br />

effectively by introducing normal genes into their<br />

bone marrow, a procedure that is normally<br />

per<strong>for</strong>med in the test tube be<strong>for</strong>e reintroducing<br />

the bone marrow to the patient. This approach<br />

has a success rate comparable to or higher than<br />

bone marrow transplantation, and can be applied<br />

to all patients (with no requirement to identify a<br />

matched bone marrow donor). Similarly a recent<br />

study has shown excellent treatment of<br />

adrenoleukodystrophy (the disease featured in<br />

the film ‘Lorenzo's Oil’) using this approach.<br />

However, the choice of viral vectors is important,<br />

since treatment of these disorders requires lifelong<br />

expression of the therapeutic genes, and<br />

early generations of viruses used have been<br />

found to damage the DNA upon inserting into the<br />

genome, leading to leukaemia. Accordingly<br />

scientists are now seeking to identify safer<br />

viruses, to enable this approach to be used<br />

widely. Another approach to efficient delivery is in<br />

the field of retinal blindness, where viruses<br />

expressing therapeutic genes can be identified<br />

directly into the diseased area. Again, very<br />

encouraging results have been seen, with clear<br />

improvements in sight resulting. <strong>Gene</strong> therapy<br />

can also apply to the use of genetic vaccines,<br />

siRNA, and tumour-killing ‘oncolytic’ viruses, all<br />

areas where we are seeing rapid progress. As<br />

knowledge increases rapidly, so the day when<br />

genetic medicines are a routine part of life comes<br />

steadily closer.<br />

Session: Educational Session


19 |<br />

Inv 7<br />

Clinical Experience with <strong>Gene</strong> <strong>Therapy</strong> in<br />

Pediatric Patients<br />

Josef Rosenecker<br />

Department of Pediatrics, Ludwig Maximilians<br />

University, <strong>Munich</strong>, <strong>German</strong>y<br />

The division of infectious diseases at the<br />

Department of Pediatrics, University of <strong>Munich</strong>,<br />

takes care <strong>for</strong> three patients who were treated<br />

with gene therapy protocols. This presentation<br />

will highlight their clinical follow up after gene<br />

therapy. The first patient was diagnosed with<br />

SCID-X1 in early infancy. No HLA-identical<br />

sibling was avialable and no matched bone<br />

marrow donor was available. There<strong>for</strong>e, the<br />

patient was transferred to Hospital Necker-<br />

Enfants Malades, Dr. Fischer, where an ex vivo<br />

retroviral mediated gene therapy was per<strong>for</strong>med.<br />

The lymphocyte development after gene therapy<br />

showed a successful reconstitution of the<br />

immune system. After gene therapy the patient<br />

developed a clonal T-cell proliferation, but is now<br />

in good clinical condition. The second patient was<br />

diagnosed with ADA-SCID and was transferred to<br />

Hospital San Raffaele, Dr. Aiuti, Milano, where an<br />

ex vivo gene therapy protocol was per<strong>for</strong>med.<br />

After gene therapy the patient showed<br />

reconstitution of the immune system and is now<br />

also in good clinical condition. A third patient was<br />

diagnosed of chronic granulomatous disease and<br />

at the age of 5 years was having severe infection<br />

with Aspergillus nidulans of the lung and of the<br />

thoracic vertebral column. He showed<br />

progressive tetraparesis. Antimycotic therapy<br />

showed only limited success. No HLA-identical<br />

sibling was avialable. No matched unrelated<br />

donor found. In this situatuion the patient was<br />

transferred to Zurich, where an ex vivo gene<br />

therapy protocol has been per<strong>for</strong>med. After gene<br />

therapy the patient showed 20% gene corrected<br />

cells in the PBL <strong>for</strong> about 4 weeks, then less than<br />

1%. He showed progressive recovery and plays<br />

no football and goes to school.<br />

Session: Educational Session<br />

Inv 8<br />

<strong>Gene</strong> Transfer Approaches <strong>for</strong> <strong>Gene</strong> Addition,<br />

Knockdown and Cellular Reprogramming In<br />

Vivo<br />

Mark A. Kay<br />

Departments of Pediatrics and <strong>Gene</strong>tics,<br />

Stan<strong>for</strong>d University, Stan<strong>for</strong>d, CA, USA<br />

Vectors that allow <strong>for</strong> DNA directed-RNA<br />

transcription can be used to treat a broad number<br />

of diseases. Our laboratory has been developing<br />

both minicircle plasmid based vectors and<br />

recombinant adenoassociated viral (rAAV)<br />

vectors <strong>for</strong> the purpose of developing plat<strong>for</strong>ms<br />

<strong>for</strong> gene addition, knockdown, and cellular<br />

reprogramming in vivo. These plat<strong>for</strong>m<br />

technologies are being tested in mice, dogs, and<br />

humans with hemophilia B (Factor IX deficiency),<br />

mouse models of Hepatitis C Virus Infection, and<br />

mouse models of juvenile onset insulin<br />

dependent diabetes mellitus, as examples of<br />

gene addition, knockdown, and cellular<br />

reprogramming strategies, respectively. Minicircle<br />

DNA plasmid vectors are devoid of all bacterial<br />

plasmid backbone DNA providing 20 to 1000<br />

times more persistent transgene expression<br />

compared to routine plasmids when transfected<br />

into quiescent cells in vivo. The mechanistic<br />

differences compared to routine plasmid DNA<br />

vectors are beginning to be elucidated. From a<br />

practical standpoint, we have developed a<br />

simplified method <strong>for</strong> minicircle vector<br />

preparation that is nearly equivalent to a routine<br />

plasmid preparation making it feasible <strong>for</strong> these<br />

DNAs to replace routine plasmids <strong>for</strong> all<br />

mammalian expression studies. Novel<br />

recombinant AAV vectors with altered<br />

transduction properties and/or site-specific<br />

integration into the ribosomal DNA locus are<br />

being derived. These expanded vector properties<br />

increase their utility <strong>for</strong> treating serious diseases<br />

in people. We will present our current studies<br />

using these improved vectors <strong>for</strong> treating the<br />

three plat<strong>for</strong>m diseases stated above.<br />

Session: Keynote Lecture<br />

Inv 9<br />

Delivery Issues <strong>for</strong> Oncolytic Viruses<br />

Len Seymour<br />

Department of Clinical Pharmacology, University<br />

of Ox<strong>for</strong>d, Ox<strong>for</strong>d, UK<br />

While there are several examples where gene<br />

therapy approaches are looking promising, they<br />

all reflect situations where the gene vectors can<br />

be introduced efficiently into target cells – either<br />

by direct injection or by transduction ex vivo.<br />

Indeed, cancer gene therapy can be very<br />

effective if tumour-killing viruses are injected<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong>


directly into tumours. Un<strong>for</strong>tunately three quarters<br />

of people who develop cancer in the West go on<br />

to die from metastatic disease. In this situation it<br />

is not possible to inject gene therapy vectors into<br />

all the tumour nodules, and intravenous<br />

‘systemic’ therapy is required. However, the<br />

human bloodstream represents a very aggressive<br />

environment <strong>for</strong> most gene therapy vectors.<br />

Therapeutic microbes delivered via the<br />

bloodstream encounter many host defences and<br />

anatomical barriers that must be surmounted to<br />

enable their access to disseminated cancers.<br />

This is particularly true <strong>for</strong> adenovirus type 5 in<br />

humans, where most recipients have powerful<br />

pre-existing adenovirus-neutralising activity. We<br />

have recently shown that human (but not murine)<br />

erythrocytes provide an additional barrier by<br />

sequestering adenovirus onto the Coxsackie and<br />

Adenovirus Receptor and (via antibodies)<br />

complement receptor 1. Coating adenovirus with<br />

a layer of hydrophilic polymer can prevent this<br />

interaction and allow virus to circulate free in the<br />

plasma, showing passive targeting to<br />

disseminated tumours and mediating good<br />

anticancer efficacy. Entry of polymer-coated virus<br />

particles into the tumour mass is a product of<br />

fluid transfer, and is directly proportional to the<br />

area under the plasma concentration-time curve.<br />

Increasing extravasation of fluid through tumourassociated<br />

endothelium using permeabilityenhancers<br />

such as Tumour Necrosis Factor<br />

alpha can improve virus particle entry into<br />

tumours over 100-fold, reaching as high as 10%<br />

injected dose (virus particles) per tumour. This<br />

provides the possibility <strong>for</strong> highly efficient<br />

targeting to tumours and good anticancer<br />

efficacy. An alternative approach is to target<br />

agents to infect tumour-associated vasculature.<br />

However while this provides a vulnerable target<br />

to traditional gene therapy approaches,<br />

endothelial cells do not normally support<br />

‘oncolytic’ viruses. One approach to overcoming<br />

this problem is to encode syncytium-<strong>for</strong>ming<br />

proteins within the endothelial, to enable transcomplementation<br />

of virus replication by tumourassociated<br />

factors.<br />

Session: Vector Development<br />

Inv 10<br />

Adenovirus Vector Engineering <strong>for</strong> <strong>Gene</strong><br />

<strong>Therapy</strong><br />

Andrew H Baker<br />

University of Glasgow, Glasgow, UK<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong><br />

| 20<br />

Adenoviral vectors are used frequently <strong>for</strong> gene<br />

therapy but much of their potential is limited by<br />

their tropism <strong>for</strong> liver and spleen, leading to<br />

effects on virion, sequestration, limited gene<br />

transfer to alternate tissue and toxicity. Recent<br />

studies have elucidated the mechanism<br />

underlying much of this tropism and the<br />

consequence of this infectivity profile. 1–4 We have<br />

shown the precise role of coagulation factor X<br />

(FX) in mediating liver uptake of adenovirus in<br />

rodent models in recent years. This occurs<br />

through a nM interaction between the FX Gla<br />

domain and the hypervariable regions of the Ad5<br />

hexon trimer. We modeled our cryoelectron<br />

microscopy data from the Ad5:FX interaction. We<br />

observed specific contact points within the hexon<br />

in hypervariable regions 5 and 7. Creation of<br />

novel vectors with deletions and amino acid<br />

mutations in these regions had dramatic effects<br />

on FX-mediated gene delivery in vitro and in vivo.<br />

We have also assessed the impact of HSPG<br />

structure on adenovirus uptake. These findings<br />

highlight the fundamental importance of the<br />

hexon:FX interaction dictating in vivo tropism as<br />

well as novel avenues <strong>for</strong> vector retargeting to<br />

alternate sites in vivo. 1 Parker AL et al., Blood<br />

(2006); 2 Waddington SN et al., Cell, (2008);<br />

3 4<br />

Kalyuzhniy, O et al., PNAS (2008); Di Paola, N<br />

et al., Immunity (2009).<br />

Session: Vector Development<br />

Inv 11<br />

Peptide-Enhanced Delivery of Oligonucleotide<br />

Analogues Targeting Duchenne Muscular<br />

Dystrophy<br />

Michael J Gait 1 , Amer F Saleh 1 , Andrey A<br />

Arzumanov 1 , Haifang Yin 2 , Matthew Wood 2<br />

1Laboratory of Molecular Biology, Medical<br />

Research Council, Cambridge, UK; 2 University of<br />

Ox<strong>for</strong>d, Ox<strong>for</strong>d, UK<br />

Duchenne muscular dystrophy (DMD) is an Xlinked<br />

genetic disease that affects 1 in 3500 male<br />

births. Mutations in the dystrophin gene result in<br />

aberrant splicing of the pre-mRNA and hence a<br />

truncated and inactive protein. Dystrophin<br />

connects actin to the dystrophin-associated<br />

complex at the sarcolemma membrane and the<br />

lack of dystrophin leads to progressive muscle<br />

degeneration and a significantly shorter life span.<br />

There is no effective treatment currently<br />

available. A number of potential gene therapy<br />

approaches are in development. Meanwhile a<br />

promising treatment has reached clinical trials,


21 |<br />

which involves use of antisense oligonucleotides<br />

(ON). ONs are targeted to bind to dystrophin premRNA<br />

and redirect splicing, resulting in “exon<br />

skipping” that restores the correct protein reading<br />

frame. The resultant expressed protein is shorter<br />

than natural dystrophin but can substitute very<br />

effectively. This phenotype is similar to Becker<br />

muscular dystrophy that shows generally only<br />

mild symptoms in patients. Two chemistry types<br />

<strong>for</strong> an exon 51 targeting ON are currently in<br />

clinical trials, ′ 2 -O-methyl phosphorothioates in<br />

Holland and Belgium and phosphoramidate<br />

morpholino oligonucleotides (PMO) in the UK.<br />

Early results in both cases have shown some<br />

dystrophin production in patients following<br />

systemic delivery. However, levels of dystrophin<br />

restored are variable and it is unclear yet whether<br />

a therapeutically useful effect will be reached in<br />

all muscle types at moderate dosage levels. To<br />

enhance activity, Arginine-rich peptides have<br />

been developed as conjugates of PMO targeting<br />

exon 23 that showed in an mdx mouse model of<br />

DMD substantially improved dystrophin<br />

production compared to naked PMO. We have<br />

been developing novel Arginine-rich Pip peptides<br />

that when attached to exon 23-targeted PMO<br />

have shown very high dystrophin generation in<br />

mdx mice, including in hard-to-reach heart<br />

muscle. These and other Arginine-rich peptides<br />

involving muscle-specific targeting domains are<br />

currently being evaluated as candidates <strong>for</strong><br />

possible use as a peptide-PMO conjugate in a<br />

future clinical trial <strong>for</strong> DMD.<br />

Session: Vector Development<br />

Inv 12<br />

Hyperactive Transposons <strong>for</strong> <strong>Gene</strong>tic<br />

Modification of Induced Pluripotent and Adult<br />

Stem Cells: A Novel Non-Viral Paradigm <strong>for</strong><br />

Coaxed Differentiation<br />

Marinee K. L. Chuah 1 , Eyayu Belay 1 , Janka<br />

Mátrai 1 , Abel Acosta-Sanchez 1 , Ling Ma 1 , Mattia<br />

Quattrocelli 2 , Lajos Mátés 3 , Pau Sancho-Bru 2 ,<br />

Martine Geraerts 2 , Bing Yan 1 , Joris Vermeesch 4 ,<br />

Ermira Samara-Kuko 1 , Zoltán Ivics 3,5 , Catherine<br />

Verfaillie 2 , Maurillio Sampaolesi 2 , Zsuzsanna<br />

Izsvák 3,5 , Thierry VandenDriessche 1<br />

1 Vesalius Research Centrum KUL-VIB, Leuven,<br />

Belgium; 2 Stem Cell Institute, University of<br />

Leuven, Leuven, Belgium; 3 Max Delbrück Center<br />

<strong>for</strong> Molecular Medicine, Berlin, <strong>German</strong>y; 4 Center<br />

<strong>for</strong> Human <strong>Gene</strong>tics, University Hospital<br />

Gasthuisberg, Leuven, Belgium; 5 University of<br />

Debrecen, Debrecen, Hungary<br />

Adult stem cells and induced pluripotent stem<br />

cells (iPS) hold great promise <strong>for</strong> regenerative<br />

medicine. The development of robust non-viral<br />

approaches <strong>for</strong> stem cell gene transfer would<br />

facilitate functional studies and potential clinical<br />

applications. We there<strong>for</strong>e generated hyperactive<br />

transposases derived from Sleeping Beauty,<br />

using an in vitro molecular evolution and<br />

selection paradigm. These hyperactive<br />

transposases resulted in superior gene transfer<br />

efficiencies and expression in mesenchymal and<br />

muscle stem/progenitor cells, consistent with<br />

higher expression levels of therapeutically<br />

relevant proteins including coagulation factor IX.<br />

Their differentiation potential and karyotype was<br />

not affected. Most importantly, relatively efficient<br />

stable gene transfer could be obtained in bona<br />

fide hematopoietic stem cells that are capable of<br />

hematopoietic reconstitution and multi-lineage<br />

gene marking (Mates, Chuah et al., Nature<br />

<strong>Gene</strong>tics, 41(6):753–761, 2009). Moreover,<br />

stable transposition could also be achieved in iPS<br />

which retained their ability to differentiate along<br />

neuronal, cardiac and hepatic lineages without<br />

causing cytogenetic abnormalities. Most<br />

importantly, transposon-mediated delivery of the<br />

myogenic PAX3 transcription factor into iPS<br />

coaxed their differentiation into MYOD +<br />

myogenic progenitors and multinucleated<br />

myofibers, suggesting that PAX3 may serve as a<br />

myogenic “molecular switch” in iPS. Hence, this<br />

hyperactive transposon system represents an<br />

attractive non-viral gene transfer plat<strong>for</strong>m with<br />

broad implications <strong>for</strong> regenerative medicine, cell<br />

and gene therapy.<br />

Session: On the Route to Clinical Application<br />

Inv 13<br />

Perinatal <strong>Gene</strong> <strong>Therapy</strong> <strong>for</strong> Lethal <strong>Gene</strong>tic<br />

Diseases<br />

Simon N Waddington 1 , Ahad A. Rahim 2 , Citra<br />

Mattar 3 , Andrew M.S. Wong 4 , Klemens Hoeffer 4 ,<br />

Suzanne M.K. Buckley 5 , Jonathan D. Cooper 4 ,<br />

Jerry Chan 3<br />

1 University College London, London, UK;<br />

2 Institute <strong>for</strong> Women's Health, University College<br />

London, London, UK; 3 National University of<br />

Singapore, Singapore; 4 Paediatric Storage<br />

Disease Laboratory, Kings College London,<br />

London, UK; 5 Department of Haematology,<br />

University College London, London, UK;<br />

A number of inherited neurological diseases are<br />

characterised by neurodegenerative changes at<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong>


or around birth. Prognosis remains dismal and <strong>for</strong><br />

several of these diseases, including Acute<br />

Neuronopathic Gaucher Disease there is no<br />

treatment and palliative care remains the only<br />

option. Owing to the aggressive nature of this<br />

and related diseases fetal or neonatal gene<br />

therapy may be the only potential means of<br />

treatment. Recently, AAV9 has been shown to<br />

cross the blood brain barrier following<br />

intravenous injection into neonatal mice, cats and<br />

macaques. We have investigated this further by<br />

studying vector tropism after intravenous injection<br />

into fetal mice. In utero intravenous injection of<br />

single-stranded (ss) and self-complimentary (sc)<br />

AAV9 expressing green fluorescent protein<br />

(GFP) into mice (embryonic day 16) resulted in<br />

efficient global transduction of neurons in the<br />

CNS. Furthermore, there was a stark contrast in<br />

cell type transduction when compared to<br />

neonatal administration, confirmed by scanning<br />

confocal microscopy. The efficiency varied<br />

depending upon ss or sc configuration of the<br />

vector. Examination of injected mice by<br />

fluorescent microscopy, immunohistology, and<br />

GFP ELISA revealed extensive and efficient<br />

transduction in the visceral organs in addition to<br />

muscle, bone, eye, and skin. <strong>Gene</strong> expression<br />

was also seen in the peripheral nervous system.<br />

Similarly, extensive transduction was also<br />

observed following intravenous administration of<br />

AAV9 to the fetal macaque. The combination of<br />

CNS and visceral organ transduction is well<br />

suited to the study, and potential treatment <strong>for</strong><br />

diseases such as acute neuronopathic Gaucher<br />

disease where both CNS and visceral pathology<br />

require targeting and a suitable mouse model is<br />

available.<br />

Session: On the Route to Clinical Application<br />

Inv 14<br />

Clinical Trial of <strong>Gene</strong> <strong>Therapy</strong> <strong>for</strong> Early Onset<br />

Severe Retinal Dystrophy Resulting from<br />

Defects in RPE65<br />

Robin R Ali 1 , JWB Bainbridge 1 , AJ Smith 1 , FW<br />

Fitzke 1 , GE Holder 1 , A Stockman 1 , SS<br />

Bhattacharya 1 , GS Rubin 1 , S Yzer 2 , I van den<br />

Born 2 , AT Moore 1<br />

1 Division of Molecular <strong>Therapy</strong>, UCL Institute of<br />

Ophthalmology and UCL/Moorfields Eye Hospital<br />

Biomedical Research Centre <strong>for</strong> Ophthalmology,<br />

London, UK; 2 Rotterdam Eye Hospital,<br />

Rotterdam, The Netherlands<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong><br />

| 22<br />

Early-onset severe retinal dystrophy caused by<br />

defects in the gene encoding the retinal<br />

isomerase RPE65 is associated with poor vision<br />

at birth and complete loss of vision in early<br />

adulthood. In a phase I/II dose-escalation trial,<br />

we have delivered subretinally recombinant<br />

adeno-associated virus (rAAV) vector expressing<br />

RPE65 under the control of an RPE65 promoter<br />

in 9 human subjects with early onset severe<br />

retinal dystrophy associated with mutations in<br />

RPE65. We have examined systemic vector<br />

dissemination and immune responses following<br />

vector delivery, assessed visual function pre- and<br />

post-vector delivery using a range of<br />

psychophysical techniques, and per<strong>for</strong>med<br />

detailed electrophysiology and retinal imaging<br />

studies. There have been no serious adverse<br />

effects of surgical delivery of vector in the<br />

subjects enrolled to date. We have detected no<br />

systemic dissemination of vector genome.<br />

Although we have detected an increase in<br />

systemic neutralising antibodies to AAV capsid in<br />

two subjects, there have been no evidence of<br />

immune responses to RPE65 protein. We have<br />

measured significant improvements in retinal<br />

sensitivity by microperimetry and dark-adapted<br />

perimetry, and improved per<strong>for</strong>mance in a test of<br />

visually-guided mobility. The outcomes in the first<br />

9 subjects to date suggest that subretinal delivery<br />

of rAAV vector can be safe in humans in the<br />

short term and can improve retinal sensitivity.<br />

These findings support further clinical studies in<br />

subjects with RPE65 deficiency and the<br />

development of gene therapy <strong>for</strong> other inherited<br />

retinal disorders.<br />

Session: <strong>Gene</strong> <strong>Therapy</strong><br />

Inv 15<br />

Alipogene Tiparvovec: the First <strong>Gene</strong> <strong>Therapy</strong><br />

<strong>for</strong> a <strong>Gene</strong>ral Metabolic Disorder<br />

Harald Petry 1 , D. Gaudet 2 , A. Carpentier 3 , E.<br />

Stroes 4 , J. Twisk 1 , S. Greentree 1<br />

1Amsterdam<br />

Molecular Therapeutics (AMT),<br />

Amsterdam, The Netherlands; 2 Department of<br />

medicine, Université de Montreal, Quebec,<br />

Canada; 3 Department of Endocrinology,<br />

University of Sherbrooke, Quebec, Canada;<br />

4<br />

Academisch Medical Center (AMC), Amsterdam,<br />

The Netherlands<br />

<strong>Gene</strong> therapy is coming of age. Correction or<br />

supplementation of genetic defects through gene<br />

therapy is amenable to both monogenic disorders<br />

as well as to diseases with a key protein playing


23 |<br />

a key role in the pathology. Alipogene tiparvovec<br />

(Glybera ® , AMT-011) is the first gene therapy<br />

product <strong>for</strong> a metabolic disease in late stage<br />

clinical development. The product contains a<br />

gain-of-function variant of the human lipoprotein<br />

lipase gene in an AAV1 based vector (AAV1-<br />

LPLS447X). Alipogene tiparvovec has been<br />

developed <strong>for</strong> the long term correction of<br />

lipoprotein lipase deficiency, to control or abolish<br />

symptoms and prevent complications in adult<br />

patients clinically diagnosed with lipoprotein<br />

lipase deficiency (LPLD). LPLD is a rare,<br />

seriously debilitating autosomal inherited<br />

monogenic disorder of lipid metabolism.<br />

Deficiency of LPL function results in<br />

chylomicronaemia. Recurrent pancreatitis is<br />

known as the most frequent, potentially lethal,<br />

complication; other severe sequelae include<br />

diabetes and increased tendency <strong>for</strong><br />

atherosclerosis. During the presentation the<br />

various steps to develop a gene therapy<br />

medicinal product will be discussed. Alipogene<br />

tiparvovec in LPLD will be used to demonstrate<br />

that persistent gene transduction after one-time<br />

intramuscular administration is feasible, and<br />

results in long-term clinical improvements in the<br />

‘chylomicronaemia syndrome’ caused by LPLD.<br />

Session: <strong>Gene</strong> <strong>Therapy</strong><br />

Inv 16<br />

<strong>Gene</strong> <strong>Therapy</strong> <strong>for</strong> Chronic Granulomatous<br />

Disease: the Past and the Future<br />

Manuel Grez 1 , CGD Consortium 2<br />

1Georg-Speyer-Haus, Frankfurt am Main,<br />

<strong>German</strong>y; 2 Frankfurt, Heidelberg, Idar Oberstein,<br />

London, Zürich<br />

In our recent gene therapy trial <strong>for</strong> X-CGD we<br />

demonstrated reconstitution of superoxide activity<br />

in phagocytic cells and elimination of preexisting<br />

infections in two treated patients. However, an<br />

unexpected expansion of myeloid progenitors<br />

occurred five months after transplantation. Both<br />

patients developed a myelodysplastic syndrome<br />

(MDS) caused by insertional activation of<br />

MDS1/EVI1 followed by clonal progression and<br />

the gradual loss of chromosome 7. P1 died 27<br />

months after gene therapy of MDS in<br />

combination with severe septicemia, the latter<br />

resulting from loss of bacterial killing activity in<br />

transduced cells. P2 underwent allogeneic stem<br />

cell transplantation. Forced overexpression of<br />

MDS1/EVI1 or EVI1 in human cells disrupted<br />

normal centrosome duplication, linking<br />

MDS1/EVI1 activation to the development of<br />

genomic instability. Optimized SIN<br />

gammaretroviral vectors have been shown to<br />

have an enhanced safety profile. From several<br />

SIN-gammaretroviral vectors expressing<br />

gp91phox, we selected a construct containing the<br />

promoter of the human c-fes gene <strong>for</strong> detailed<br />

preclinical studies. We used X-CGD mice <strong>for</strong><br />

monitoring safety and functional reconstitution of<br />

NADPH oxidase activity in primary and<br />

secondary animals. The SINfes vector was safe,<br />

was resistant to CpG methylation and<br />

reconstituted superoxide activity to clinical<br />

relevant levels. We plan to use this vector <strong>for</strong> the<br />

next Phase I gene therapy trial.<br />

Session: <strong>Gene</strong> <strong>Therapy</strong><br />

Inv 17<br />

Reprogrammed Measles Viruses as Cancer<br />

Therapeutics Three Points of Attack<br />

Roberto Cattaneo<br />

Department of Molecular Medicine, Mayo Clinic,<br />

and Virology and <strong>Gene</strong> <strong>Therapy</strong> track, Mayo<br />

Graduate School, Rochester, USA<br />

Soon after viruses were recognized more than<br />

100 years ago, tumor regressions occasionally<br />

documented after accidental infections suggested<br />

the idea of using them to fight cancer. Early<br />

virotherapy clinical trials based on wild type<br />

viruses were unsuccessful, but recent ones<br />

based on genetically modified viruses are built on<br />

much stronger foundations. They <strong>for</strong>esee<br />

extensive monitoring of viral replication, gene<br />

expression, and host immunity. Therapeutic<br />

efficacy is being assessed by well-defined<br />

biological end points, and can be improved. For<br />

future clinical trials more specific and potent<br />

oncolytic viruses are developed based on three<br />

types of modification: targeting, arming, and<br />

shielding. Targeting introduces multiple layers of<br />

cancer specificity; arming amplifies locally the<br />

effects of approved cancer therapeutics; and<br />

shielding provides temporary relief from the<br />

immune response. We have shown that the<br />

envelope of measles (MV) and related<br />

paramyxoviruses can be targeted to many<br />

designated receptors. Retargeted MV envelopes<br />

have become preferred tools to pseudotype<br />

lentiviral and other gene transfer vectors. We<br />

have developed protease activation targeting,<br />

and innate immunity control targeting. We have<br />

armed MV with a prodrug convertase, and shown<br />

that this virus synergizes with the<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong>


chemotherapeutic fludarabine to eliminate mantle<br />

cell lymphoma xenografts. MV-based<br />

therapeutics developed by the Russell and Peng<br />

groups at Mayo are being administered to<br />

patients who, having exhausted other therapeutic<br />

options, have enrolled in clinical trials of ovarian<br />

carcinoma, glioma, and myeloma. Survival and<br />

quality of life data are being collected and will be<br />

critical <strong>for</strong> the approval of new therapeutic<br />

products. Cattaneo, R. (2010) Paramyxovirus<br />

entry and targeted vectors <strong>for</strong> cancer therapy.<br />

PLoS Pathogens 6: e1000973.<br />

Session: Cancer <strong>Gene</strong> <strong>Therapy</strong><br />

Inv 18<br />

Targeted and Armed Oncolytic Poxviruses: A<br />

Novel Multimechanistic Therapeutic Class <strong>for</strong><br />

Cancer<br />

Caroline Breitbach 1 , John C Bell 1,2 , David H Kirn 1<br />

1 Jennerex Inc, San Francisco, USA; 2 Centre <strong>for</strong><br />

Cancer Therapeutics, Ottawa Hospital Research<br />

Institute, Ottawa, Canada<br />

Engineered viruses have been developed <strong>for</strong><br />

cancer therapy both as non-replicating gene<br />

therapy agents and as cancer vaccines.<br />

Oncolytic viruses, in contrast, were developed to<br />

replicate within and subsequently lyse, cancer<br />

cells. Clinical efficacy to date with each of these<br />

approaches has been limited by multiple factors<br />

including the inability to infect enough tumor cells<br />

in vivo locally within a tumor or systemically, and<br />

resistance of complex advanced tumors to a<br />

single mechanism-of-action (MOA). Over the last<br />

several years, however, a novel therapeutic class<br />

has emerged that combines the best features of<br />

all three approaches: targeted and armed<br />

oncolytic poxviruses. Recent preclinical and<br />

clinical results demonstrate convincingly that<br />

products from this therapeutic class can achieve<br />

highly selective and potent cancer destruction<br />

systemically through a multi-pronged MOA.<br />

Given recent clinical validation, we expect this<br />

therapeutic class to expand rapidly.<br />

Session: Cancer <strong>Gene</strong> <strong>Therapy</strong><br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong><br />

| 24<br />

Inv 19<br />

Oncolytic Vaccinia Virus <strong>for</strong> the Treatment of<br />

Cancer<br />

John C. Bell<br />

Centre <strong>for</strong> Cancer Therapeutics, Ottawa Hospital<br />

Research Institute, Ottawa, Canada<br />

The creation of novel “cancer lysing” or<br />

“oncolytic” viruses over the last 10-15 years has<br />

led to the development of a number of innovative<br />

anti-cancer therapeutic products many of which<br />

have entered into clinical testing. We are<br />

examining a number of poxvirus backbones <strong>for</strong><br />

their ability to selectively target and kill tumour<br />

cells. A derivative of the Wyeth Strain called JX-<br />

594 has engineered deletions, natural mutations<br />

and expresses a GM-CSF transgene, all of which<br />

contribute to its ability to infect and kill tumour<br />

tissues while leaving normal tissues unscathed.<br />

The results of our preclinical models and<br />

molecular studies have shown that JX-594<br />

attacks cancers using a multi-pronged approach<br />

that includes tumour vascular targeting, immune<br />

stimulation and direct cancer cell lysis. JX-594<br />

has now been tested in over 70 patients in a<br />

variety of different trial designs and consistently<br />

shows tumour targeting activity and a very strong<br />

safety profile. The results of our ongoing<br />

translational research ef<strong>for</strong>ts and clinical data<br />

from our phase I and II trials will be presented.<br />

Session: Cancer <strong>Gene</strong> <strong>Therapy</strong><br />

Inv 20<br />

Prostate Cancer Stem Cells: A New Target <strong>for</strong><br />

<strong>Therapy</strong><br />

Norman J Maitland<br />

YCR Cancer Research Unit, Dept of Biology,<br />

University of York, Heslington, UK<br />

Human prostate cancer is characterised by<br />

cellular heterogeneity; perhaps one reason <strong>for</strong><br />

the resistance of prostate cancer to many<br />

medical therapies. Once a prostate cancer has<br />

failed on hormone-based therapies, the<br />

prognosis <strong>for</strong> the patients is poor. Our approach<br />

to tissue heterogeneity in the prostate has been<br />

to identify and purify individual cell populations<br />

from human prostate cancers and matched<br />

normal tissues. We have fractionated cancer<br />

epithelium into a majority luminal/secretory<br />

population (which comprises more than 99% of<br />

the cancer mass) and a minor population


25 |<br />

consisting of committed basal cells, transamplifying<br />

cells and a stem cell population. The<br />

latter cells are responsible <strong>for</strong> tumour initiation<br />

and in vitro colony <strong>for</strong>mation. Affymetrix<br />

microarray analysis of these populations confirms<br />

a distinct phenotype <strong>for</strong> each cell type and<br />

functional expression of fate influencing genes.<br />

Further analysis has revealed populations of<br />

genes whose expression is tightly linked to the<br />

differentiation process, while others are<br />

associated with the cancerous properties of the<br />

cells. The tumour origin of the identified cells has<br />

been confirmed by xenografting into a highly<br />

immuno-compromised mouse strain at orthotopic,<br />

kidney capsule and subcutaneous sites with<br />

demonstrable metastases. <strong>Gene</strong>s associated<br />

with prostate cancer are expressed in the<br />

cultures and their functional significance<br />

assessed by SiRNA knock-down in stem cells<br />

and in their daughter cells. Epigenetic control, in<br />

contrast to permanent genetic alterations or<br />

gross alterations involving a reprogramming of<br />

cells to express specific transcription factor sets,<br />

seems to be a predominant mechanism <strong>for</strong> an<br />

adaptable stem cell in prostate. A better<br />

knowledge of the phenotype of the prostate<br />

CSC's should permit therapeutic targeting.<br />

However, whilst stem cell directed therapies (to<br />

treat less than 0.1% of the tumour mass) are<br />

unlikely to have an immediate effect on tumour<br />

volume, one consequence of targeting the wrong<br />

cell types within a heterogeneous tumour is to<br />

perturb the natural history of the tumour and<br />

results in an amplification of CSC's, in agreement<br />

with cancer relapse patterns in man.<br />

Session: Tumor Biology and (Cancer) Stem Cells<br />

Inv 21<br />

A Consortium <strong>for</strong> Clinical Development of<br />

Lentiviral Vector-Induced DCs <strong>for</strong> Melanoma<br />

Immunotherapy<br />

Renata Stripecke 1 , Sandra Kuhs 1 , Ralf Gutzmer 2 ,<br />

Henk Garritsen 3 , Farzin Farzaneh 4<br />

1 Department of Hematology, Hemostasis,<br />

Oncology and Stem Cell Transplantation,<br />

Hannover Medical School, Hannover, <strong>German</strong>y;<br />

2 Department of Dermatology, Hannover Medical<br />

School, Hannover, <strong>German</strong>y; 3 Division of<br />

Transfusion Medicine, Braunschweig City<br />

Hospital, Braunschweig, <strong>German</strong>y; 4 Department<br />

of Haematological Medicine, King's College<br />

London, Rayne Institute, London, UK<br />

Dendritic Cells (DCs) are key players in the<br />

development of adaptive immunity. Their use to<br />

boost anti-tumor responses have reached phase<br />

III clinical trials but ex vivo production is costly<br />

and difficult to standardize. A novel concept <strong>for</strong><br />

DC production consists of overnight lentiviral<br />

vector (LV) transduction of growth factors and<br />

full-length antigens into monocytes that results<br />

into induction of “SMART-DCs” (Selfdifferentiated<br />

Myeloid-derived Antigenpresenting-cells<br />

Reactive against Tumors). This<br />

concept has been validated in the preclinical B16<br />

melanoma mouse model <strong>for</strong> potency and safety<br />

with MART-1 and TRP2 as melanoma-associated<br />

antigens, overexpressed in 90% of the metastatic<br />

melanoma (Koya et al., 2007; Pincha et al.,<br />

submitted). Third generation self-inactivating<br />

tricistronic lentiviral vectors containing<br />

interspacing 2A elements co-expressing human<br />

GM-CSF, IL-4 and MART-1 or TRP-2 were<br />

constructed. Cytokine preconditioning and 16h<br />

transduction of CD14 + monocytes with high titer<br />

LVs resulted in persistent (3 weeks) autocrine<br />

production of GM-CSF (average 10 ng/ml), IL-4<br />

(average 12 ng/ml) and expression of MART-1 or<br />

TRP2 (detectable by intracellular staining).<br />

Transduced monocytes readily self-differentiated<br />

into SMART-DCs (CD209 + , MHCII + , CD80 +<br />

CD86+) and were stable in culture <strong>for</strong> 3 weeks.<br />

SMART-DCs/MART-1 were recognized by T cell<br />

clones specific <strong>for</strong> MART-1 in an HLA-restricted<br />

manner, assessed by IFN-γ-ELISPOT-Assay.<br />

PBMCs obtained from healthy donors that were<br />

primed/boosted in vitro with autologous SMART-<br />

DCs/MART-1 demonstrated the induction of<br />

MART-1-specific T cell responses assayed by<br />

IFN-γ-ELISPOT-Assay. SMART-DCs/MART-1<br />

and SMART-DCs/TRP2 are currently being<br />

tested <strong>for</strong> stimulation of T cell responses in<br />

melanoma patients. A consortium <strong>for</strong> clinical<br />

development of SMART-DCs has been <strong>for</strong>med:<br />

GMP-grade batch of the tricistronic vectors is<br />

planned <strong>for</strong> additional preclinical testing (optimal<br />

vector dose, number of integrated copies,<br />

toxicity), monocytes will be transduced with LVs<br />

in a closed GMP-grade bag system, development<br />

of Standard Operating Procedures and<br />

establishment of identity and potency markers<br />

are underway.<br />

Session: Cancer Immune <strong>Therapy</strong> and T-Cell<br />

<strong>Therapy</strong><br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong>


Inv 22<br />

Arming Immune Cells to Fight Cancer<br />

Hinrich Abken<br />

Zentrum für Molekulare Medizin Köln und Klinik I<br />

für Innere Medizin, Uniklinik Köln, Cologne,<br />

<strong>German</strong>y<br />

Cancer cells frequently escape T cell recognition<br />

by repression of the antigen processing<br />

machinery making them invisible <strong>for</strong> a cytolytic T<br />

cell attack. Strategies in adoptive immunotherapy<br />

during the last decade aimed to overcome the<br />

situation by engineering T cells with a<br />

recombinant chimeric antigen receptor (CAR)<br />

which uses a single chain fragment of variable<br />

region (scFv) antibody <strong>for</strong> target binding and the<br />

intracellular CD3zeta domain <strong>for</strong> T cell activation.<br />

To provide full T cell activation, the CD3zeta<br />

chain was fused with the CD28 costimulatory<br />

domain in one polypeptide chain; other<br />

costimulatory moieties are likewise used. We<br />

here discuss the impact of CD28 costimulation on<br />

the threshold in T cell activation and on the<br />

redirected anti-tumor response in the presence of<br />

TGF-beta and regulatory T cells. We moreover<br />

introduce a novel generation of CARs with<br />

combined CD28-OX40 costimulation in order to<br />

harness central memory T cells <strong>for</strong> a redirected<br />

anti-tumor attack. Finally, we demonstrate the<br />

power of a redirected T cell attack in eradicating<br />

established tumor lesions by eliminating minor<br />

tumor cell subsets.<br />

Session: Cancer Immune <strong>Therapy</strong> and T-Cell<br />

<strong>Therapy</strong><br />

Inv 23<br />

Immune <strong>Gene</strong> <strong>Therapy</strong> <strong>for</strong> Acute Myeloid<br />

Leukaemia<br />

Farzin Farzaneh 1 , Lucas Chan 1 , Nicola<br />

Hardwick 1 , Wendy Ingram 1 , Ghulam Mufti 1 , Mark<br />

Aloysius 2 , Adrian Robins 3 , Nagy Habib 4 , Joti<br />

Bhalla 1 , Oleg Eremin 2 , Tye Gee Jun 1 , David<br />

Oppenheim 1 , James Wells 5 , Christopher<br />

Cowled 5 , Alistair Noble 5<br />

1 Department of Haematological Medicine, King's<br />

College London, Rayne Institute, London, UK;<br />

2 Section of Surgery, Biomedical Research Unit,<br />

Nottingham Digestive Diseases Centre,<br />

University of Nottingham, Nottingham, UK;<br />

3 Institute of Infection and Immunity, School of<br />

Molecular Medical Sciences, Nottingham<br />

University Hospitals, University of Nottingham,<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong><br />

| 26<br />

Nottingham, UK; 4 Section of Surgery,<br />

Department of Surgical Oncology and<br />

Technology, Imperial College London, London,<br />

UK; 5 MRC and Asthma UK Centre in Allergic<br />

Mechanisms of Asthma, King's College London,<br />

Guy's Hospital, London, UK<br />

Immune mediated rejection of established<br />

tumours needs adequate stimulation of cell<br />

mediated immunity, as well as the suppression of<br />

inhibitory responses that are induced by chronic<br />

exposure to tumour associated antigens.<br />

Preclinical studies have demonstrated that<br />

tumour cells expressing immune costimulatory<br />

molecules and appropriate Th1 cytokines can<br />

induce immune mediated rejection of previously<br />

established tumours. This strategy is now being<br />

assessed in a Phase-I clinical trial <strong>for</strong> the<br />

vaccination of relapsed poor prognosis acute<br />

myeloid leukaemia (AML) with autologous AML<br />

cells that are genetically modified to express B7.1<br />

(CD80) and IL-2. We have also shown that<br />

vaccination of advanced solid tumour patients,<br />

with dendritic cells that are pulsed with<br />

telomerase (hTERT) peptides, can result in the<br />

expansion of antigen specific T cells with some<br />

evidence of a modest and short-lived therapeutic<br />

benefit. In a further recently started Phase-I study<br />

we are now evaluating the safety, and potential<br />

efficacy of, hTERT peptide loaded dendritic cell<br />

vaccination in combination with<br />

cyclophosphamide mediated inhibition of immune<br />

suppression by regulatory T cells (Treg). A much<br />

less demanding alternative to the use of dendritic<br />

or tumour cell vaccines is the direct vaccination<br />

with tumour associated antigens, provided that<br />

the vaccine could indeed induce cell mediated<br />

immunity. With this goal in mind we have recently<br />

developed a new vaccination strategy based on<br />

the combined adjuvants <strong>for</strong> synergistic activation<br />

of cellular immunity (CASAC). CASAC contains<br />

different combinations of defined molecules that<br />

act synergistically to induce dendritic cell<br />

activation. Subcutaneous vaccination with two<br />

doses of a single peptide (OVA or Trp2) plus<br />

CASAC, induces IL-12 secretion, stimulation of<br />

Th1-biased CD4 T-cells, and high levels<br />

(routinely 50% by tetramer staining) of antigen<br />

specific cytolytic CD8 T cells. The magnitude of<br />

CASAC mediated immune stimulation is<br />

substantially greater than can be achieved by<br />

other adjuvants (e.g., about 100-fold greater than<br />

complete Freund's adjuvant). The antigen<br />

specific CTL activity induced by CASAC<br />

mediated peptide vaccination allows the in vivo<br />

lysis of greater than 90% of antigen positive<br />

tumour cells in mouse tumour models, resulting<br />

in long-lasting immunity with a robust recall<br />

response. This strategy will shortly entre clinical


27 |<br />

studies <strong>for</strong> WT1 peptide vaccination of myeloid<br />

leukaemia patients.<br />

Session: Cancer Immune <strong>Therapy</strong> and T-Cell<br />

<strong>Therapy</strong><br />

Inv 24<br />

Vector Safety Data Leading to a Clinical Trial<br />

<strong>for</strong> the <strong>Gene</strong> <strong>Therapy</strong> of Wiskott Aldrich<br />

Syndrome: A Perspective on Future<br />

Developments<br />

Anne Galy, Sabine Charrier, Otto-Wilhelm<br />

Merten, Didier Caizergues<br />

<strong>Gene</strong>thon, Evry, France<br />

Hematopoietic gene therapy currently relies on<br />

the ex vivo gene correction of patient autologous<br />

hematopoietic stem cells. This approach has now<br />

been tested <strong>for</strong> over 10 years in pilot phase I/II<br />

trials in several inherited diseases. The domain is<br />

evolving and the use of lentiviral vectors derived<br />

from HIV-1 provide significant advantages in<br />

terms of genomic stability, biological properties<br />

but also in the area of pharmaceutical product<br />

characterization. The rare primary<br />

immunodeficiency Wiskott Aldrich syndrome<br />

(WAS) is a combined platelet and immune defect.<br />

In the perspective of developing a gene therapy<br />

<strong>for</strong> WAS, we have characterized an advanced<br />

generation rHIV lentiviral vector pseudotyped<br />

with VSVg and implemented a large-scale<br />

industrial process to produce and to control this<br />

vector under good manufacturing practices<br />

(GMP). Recently several European centers were<br />

approved to start gene therapy trials <strong>for</strong> WAS<br />

with this lentiviral vector. With a growing number<br />

of potential disease applications <strong>for</strong> lentiviral<br />

vector-mediated hematopoietic gene therapy, it<br />

seems that there is ground to integrate<br />

in<strong>for</strong>mation on preclinical vector safety features<br />

particularly at the level of design, manufacture<br />

process or controls. We will discuss these<br />

various points through the different aspects<br />

leading to the development of WAS gene<br />

therapy.<br />

Session: Pharmacology and Toxicology<br />

Inv 25<br />

Lentivector Transduction of Novel Target<br />

Cells<br />

Klaus Cichutek<br />

Paul-Ehrlich-Institut, Langen, <strong>German</strong>y<br />

Session: Pharmacology and Toxicology<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong>


DG-GT 2010 Oral Presentations<br />

Or 1<br />

Analysis of Intracellular Particle Trafficking<br />

and Bioresponsive Bonds <strong>for</strong> Ad Vector<br />

Shielding by Capsomer-Specific Fluorescent<br />

Labeling<br />

Sigrid Espenlaub 1 , Stephanie Corjon 1 , Tatjana<br />

Engler 1 , Carolin Fella 2 , Manfred Ogris 2 , Ernst<br />

Wagner 2 , Stefan Kochanek 1 , Florian Kreppel 1<br />

1Department of <strong>Gene</strong> <strong>Therapy</strong>, University of Ulm,<br />

Ulm, <strong>German</strong>y; 2 Pharmaceutical Biotechnology,<br />

Department of Pharmacy, Ludwig-Maximilians-<br />

University, <strong>Munich</strong>, <strong>German</strong>y<br />

Adenovirus (Ad) vectors are widely used <strong>for</strong> gene<br />

therapy approaches. Due to the high abundance<br />

of the natural adenovirus receptors CAR/integrins<br />

on a wide variety of cells numerous methods<br />

have been developed to redirect the virions to<br />

specific receptors on target cell surfaces. An<br />

increasing number of recent publications<br />

evidenced that the success of targeting strategies<br />

does not only depend on receptor binding and<br />

cellular uptake, but also on intracellular trafficking<br />

processes. There<strong>for</strong>e, improved knowledge on<br />

the intracellular fate of targeted Ad vector<br />

particles is mandatory <strong>for</strong> a rational design of<br />

targeted Ad vectors. However, while in principle<br />

potent tools are available <strong>for</strong> fluorescent labeling<br />

of Ad vectors in order to analyze post-receptor<br />

trafficking, these tools suffer from significant<br />

limitations: (i) capsids can so far only be labeled<br />

upon amino groups present in all proteins all over<br />

the particle surface and this imposes the risk of<br />

interference with particle infectivity, (ii)<br />

capsomere-specific labeling requires extensive<br />

genetic modifications and has only been<br />

demonstrated at protein IX, (iii) two-color labeling<br />

approaches are not available. Here we present a<br />

robust and straight <strong>for</strong>ward labeling procedure<br />

based on a genetic-chemical approach that<br />

overcomes these limitations. It allows <strong>for</strong> specific<br />

labeling of the capsomeres fiber, protein IX, or<br />

hexon and permits two-color labeling. Using this<br />

technology we analyzed by means of live cell<br />

imaging two different bioresponsive bonds that<br />

potentially can be used <strong>for</strong> the reversible<br />

attachment of shielding/targeting moieties to the<br />

capsids: disulfide and hydrazone bonds. In our<br />

experiments we could not observe significant<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong><br />

| 28<br />

reduction of disulfides within the observation time<br />

of 80 minutes post cell entry. Acid-responsive<br />

hydrazone bonds in contrast became quickly<br />

hydrolyzed after uptake of the virions (30 min)<br />

and may thus be favorable <strong>for</strong> the generation of<br />

bioresponsive vectors and potent tools to<br />

overcome the limitations in intracellular trafficking<br />

processes of targeted Ad vectors.<br />

Session: Vector Development<br />

Or 2<br />

Primary Human Keratinocyte-Selective AAV2-<br />

Based Targeting Vectors<br />

Jessica Sallach 1 , Fernando Laguzzi Larcher 2 ,<br />

David Gomez Almarza 2 , Luca Perabo 1 , Michael<br />

Hallek 1 , Hildegard Büning 1<br />

1 Clinic I of Internal Medicine and Center <strong>for</strong><br />

Molecular Medicine Cologne (CMMC), University<br />

Hospital Cologne, University of Cologne,<br />

Cologne, <strong>German</strong>y; 2 Ciemat-Centro de<br />

Investigacion Energetica Medioambiental y<br />

Tecnologica, Moncloa-Aravaca, Madrid, Spain<br />

Vectors based on the adeno-associated virus<br />

(AAV), a non-enveloped DNA virus, have<br />

emerged as one of the leading gene transfer<br />

systems in gene therapy. However, transduction<br />

efficiency of clinical relevant cell types such as<br />

keratinocytes is insufficient and seems to depend<br />

on donor-specific factors. We there<strong>for</strong>e<br />

conducted AAV peptide display selection on<br />

human primary <strong>for</strong>eskin keratinocytes in order to<br />

identify AAV2-based targeting mutants with<br />

improved efficacy and specificity of gene transfer<br />

into keratinocytes. Aiming to <strong>for</strong>ce a ligandreceptor<br />

interaction that is independent of<br />

heparan sulphate proteoglycan (HSPG) binding,<br />

a cell surface molecule that is expressed on a<br />

wide range of cell types, we depleted our AAV<br />

peptide library of mutants capable of HSPG<br />

binding. Following five selection rounds on<br />

human primary keratinocytes obtained from<br />

different donors, viral genomes were sequenced<br />

to identify peptide sequence(s) that mediated<br />

viral infection. Interestingly, all selected mutants


29 |<br />

displayed an integrin binding motif as peptide<br />

insertion at amino acid position 587 of the AAV2<br />

capsid. Selected peptides were then introduced<br />

into the capsid of recombinant AAV vectors<br />

(rAAV) which were subsequently assayed <strong>for</strong><br />

their transduction efficiency and cell type<br />

specificity. Compared to rAAV2 the targeting<br />

vectors showed an increased transduction<br />

efficiency of up to 15-fold in primary human<br />

keratinocytes and a dramatic decrease (up to 30fold)<br />

in off-target transduction. Inhibitor studies<br />

confirmed HSPG-independent, ligand-mediated<br />

cell entry. Further characterization of the entry<br />

mode and its consequences of host-cell<br />

interaction is ongoing and results will be<br />

discussed.<br />

Session: Vector Development<br />

Or 3<br />

Development of Novel PhiC31 Integrase<br />

Fusion Proteins <strong>for</strong> Improving Efficacy and<br />

Safety of Transgene Insertion in Therapeutic<br />

Applications<br />

Nadja Noske 1 , Nina Harms 1 , Toni Cathomen 2 ,<br />

Anja Ehrhardt 1<br />

1 Department of Virology, Max von Pettenkofer-<br />

Institute, LMU, <strong>Munich</strong>, <strong>German</strong>y; 2 Department of<br />

Experimental Hematology, Hannover Medical<br />

School, Hannover, <strong>German</strong>y<br />

Integration systems are promising and attractive<br />

tools <strong>for</strong> the development of new gene<br />

therapeutic strategies. Based on site-directed<br />

recombination the bacteriophage derived PhiC31<br />

system provides outstanding opportunities <strong>for</strong><br />

transgene integration into the host genome.<br />

However, due to unpredicted insertion events in<br />

the genome mediated by PhiC31, the<br />

optimization of activity and directing integration<br />

into specific target sites are important goals <strong>for</strong><br />

the usage in therapeutic applications. There<strong>for</strong>e,<br />

we constructed several fusion proteins of the<br />

integrase and different DNA binding domains<br />

(DBD) like the synthetic polydactyl zinc finger<br />

E2C and the AAV Rep protein. The motifs were<br />

either fused N- or C-terminal to the integrase<br />

separated by a short TS or a long GGS5 linker.<br />

Immunoblot and flow cytometry analysis revealed<br />

that the fusion proteins were expressed in<br />

comparable amounts after transfection into 293<br />

cells. Excision and integration activity retained up<br />

to 80% of the wildtype integrase (WT) when the<br />

binding motifs were attached to the C-terminus of<br />

the integrase separated by the 45-bp long GGS5<br />

interdomain. As a further step we generated<br />

fusion mutants lacking the native DBD of the<br />

integrase (ΔDBD). As expected, these were<br />

catalytically inactive when measuring<br />

recombination efficiencies between wild type<br />

PhiC31 integrase recognition sites. However,<br />

after per<strong>for</strong>ming initial colony <strong>for</strong>ming assays to<br />

measure integration events mediated by the<br />

ΔDBD fusion constructs, we observed 31%<br />

activity of the WT. To further improve the PhiC31<br />

integrase system we plan to build up on our<br />

previous study in which we identified hyperactive<br />

variants of the integrase showing up to 5.5-fold<br />

increased integration efficiency (Liesner et al.,<br />

2010). We also characterized critical amino acids<br />

involved either in attaching DNA or in the<br />

insertion process. There<strong>for</strong>e, mutants which lost<br />

their native DNA binding properties and their<br />

restoration by fusing known binding motifs would<br />

be an interesting approach to develop novel tools<br />

<strong>for</strong> site-specific integration. In combination with<br />

hyperactive PhiC31 variants this approach could<br />

lead to a more specific and efficient PhiC31<br />

integrase system.<br />

Session: Vector Development<br />

Or 4<br />

Biocompatible Polyplex Nanomicelle <strong>for</strong> Safe<br />

and Effective <strong>Gene</strong> Transfer<br />

Keiji Itaka 1 , Kazunori Kataoka 2<br />

1 Division of Clinical Biotechnology, CDBIM,<br />

Graduate School of Medicine, The University of<br />

Tokyo, Tokyo, Japan; 2 Department of Materials<br />

Science and Engineering, Graduate School of<br />

Engineering, The University of Tokyo, Tokyo,<br />

Japan<br />

<strong>Gene</strong> delivery using cationic polymers has<br />

attracted much attention due to their potential<br />

advantages, such as large DNA loading capacity,<br />

ease of large-scale production, and reduced<br />

immunogenicity. We are developing a polyplex<br />

system <strong>for</strong>ming micelle from polyethyleneglycol<br />

(PEG) - poly[N-[N-(2-aminoethyl)-2aminoethyl]aspartamide]<br />

(PEG-PAsp(DET)) and<br />

plasmid DNA. By highly-regulated structure of a<br />

diameter of ≈ 100 nm with a PEG palisade, the<br />

nanomicelle shows increased tolerance under<br />

physiologic conditions with the remarkably low<br />

cytotoxicity. A key feature <strong>for</strong> safe and effective<br />

gene transfer is the biodegradability of<br />

PAsp(DET). PAsp(DET) was revealed to undergo<br />

rapid degradation by Gel permeation<br />

chromatography (GPC) and electrospray<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong>


ionization mass spectrometry (ESI-MS)<br />

measurements. In contrast, a derivative<br />

polycation, N-substituted polyglutamide<br />

(PGlu(DET)), showed no degradability, indicating<br />

that the degradation of PAsp(DET) was induced<br />

by a specific self-catalytic reaction between the<br />

PAsp backbone and the sidechain amide<br />

nitrogen. Degradation products of PAsp(DET)<br />

caused no cytotoxicity, even at high<br />

concentrations in the culture medium. Repeated<br />

transfection by administering the polyplexes <strong>for</strong><br />

every 24 h showed that biodegradable<br />

PAsp(DET) provided a continuous increase in<br />

transgene expression, while non-degradable<br />

PGlu(DET) showed a decrease in transgene<br />

expression after 48 h, coupled with fluctuations in<br />

expression profiles of endogenous genes. The<br />

biocompatibility of nanomicelles plays a<br />

significantly role in in vivo applications. By<br />

obtaining sustained transgene expressions with<br />

minimized toxicity and inflammatory responses<br />

such as cytokine induction, we achieved<br />

successful therapeutic outcomes by gene<br />

introduction using nanomicelles, including in vivo<br />

bone regeneration by introducing osteogenic<br />

factors to bone defect area and intratracheal<br />

gene transfer of adrenomedullin to treat<br />

pulmonary hypertension. Hydrodynamic gene<br />

introduction to skeletal muscle was also well<br />

achieved compared with naked pDNA. We are<br />

now trying various applications <strong>for</strong> disease<br />

treatment and tissue regeneration using model<br />

animals.<br />

Session: Vector Development<br />

Or 5<br />

EGF Receptor Targeting of Polyplexes with<br />

the Short Artificial Peptide GE11 Studied by<br />

Live Cell Imaging<br />

Frauke M Koenig 1 , Nadia Ruthardt 1 , Ernst<br />

Wagner 2,3 , Manfred Ogris 2,3 , Christoph<br />

Braeuchle 1,3<br />

1Physical Chemistry, Department of Chemistry,<br />

Ludwig-Maximilians-University, <strong>Munich</strong>,<br />

<strong>German</strong>y; 2 Pharmaceutical Biotechnology,<br />

Department of Pharmacy, Ludwig-Maximilians-<br />

University, <strong>Munich</strong>, <strong>German</strong>y; 3 Center <strong>for</strong><br />

Nanoscience (CeNS), Ludwig-Maximilians-<br />

University, <strong>Munich</strong>, <strong>German</strong>y<br />

For the selective gene therapy of cancer cells in<br />

the human body, a tumor specific targeting is<br />

required. The EGF receptor provides a promising<br />

target as it is overexpressed in many human<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong><br />

| 30<br />

cancers. A number of anticancer therapeutics<br />

directed against the EGF receptor have been<br />

successfully developed so far. Polyplexes<br />

consisting of DNA complexed with a cationic<br />

polymer can be equipped with EGF as a ligand<br />

resulting in their fast, receptor-dependent uptake<br />

into cancer cells. However, full-length EGF has a<br />

high tendency to aggregate and is expensive in<br />

production. Furthermore, it activates the<br />

mitogenic signaling cascade of target cells,<br />

leading to undesirable cell proliferation. In this<br />

study, we there<strong>for</strong>e examined the alternative<br />

usage of GE11, a short artificial peptide with high<br />

affinity to the EGF receptor, <strong>for</strong> the specific<br />

targeting of polyplexes HUH7 cells. By live-cell<br />

imaging with highly sensitive fluorescence<br />

microscopy, we directly compared the<br />

internalization kinetics of GE11 polyplexes to the<br />

uptake of EGF coupled and untargeted<br />

polyplexes. To determine the extent of mitogenic<br />

signaling after polyplex binding to the receptor,<br />

phosphorylated signaling molecules were<br />

detected by western blotting. The correlation<br />

between receptor signaling and polyplex uptake<br />

was then examined in more detail by inhibitor<br />

studies with Erlotinib, a specific inhibitor of EGF<br />

receptor phosphorylation. Our results reveal that<br />

the uptake of GE11 polyplexes is slow compared<br />

to EGF polyplexes but much more efficient<br />

compared to untargeted polyplexes. After two<br />

hours of incubation, GE11- and EGF polyplexes<br />

reach similar internalization levels resulting in<br />

comparable transfection efficiencies on a long<br />

time scale. The activation of mitogenic signaling<br />

is significantly reduced <strong>for</strong> GE11- compared to<br />

EGF polyplexes. Further studies reveal that the<br />

different uptake kinetics of GE11 and EGF can<br />

be correlated to the activation of receptor<br />

signaling. Taken together, GE11 polyplexes are a<br />

promising candidate <strong>for</strong> clinical therapy as they<br />

show a high transfection efficiency, low<br />

aggregation properties and a reduced mitogenic<br />

activity.<br />

Session: Vector Development


31 |<br />

Or 6<br />

Restoration of Cone Vision in the CNGA3 −/−<br />

Mouse Model of Congenital Complete Lack of<br />

Cone Photoreceptor Function<br />

Stylianos Michalakis 1,2 , Regine Mühlfriedel 3 ,<br />

Naoyuki Tanimoto 3 , Vidhyasankar<br />

Krishnamoorthy 4 , Susanne Koch 1,2 , M. Dominik<br />

Fischer 3 , Elvir Becirovic 1,2 , Lin Bai 1,2 , Gesine<br />

Huber 3 , Susanne C Beck 3 , Edda Fahl 3 , Hildegard<br />

Büning 4 , François Paquet-Durand 3 , Xiangang<br />

Zong 1,2 , Tim Gollisch 5 , Martin Biel 1,2 , Mathias W.<br />

Seeliger 3<br />

1 Department of Pharmacy – Center <strong>for</strong> Drug<br />

Research, Ludwig-Maximilians-Universität<br />

München, <strong>Munich</strong>, <strong>German</strong>y; 2 Center <strong>for</strong><br />

Integrated Protein Science <strong>Munich</strong> (CIPSM),<br />

Department of Pharmacy – Center <strong>for</strong> Drug<br />

Research, Ludwig-Maximilians-Universität<br />

München, <strong>Munich</strong>, <strong>German</strong>y; 3 Division of Ocular<br />

Neurodegeneration and Division of Experimental<br />

Ophthalmology, Institute <strong>for</strong> Ophthalmic<br />

Research, Centre <strong>for</strong> Ophthalmology, Eberhard<br />

Karls-Universität, Tübingen, <strong>German</strong>y; 4 Clinic I of<br />

Internal Medicine and Center <strong>for</strong> Molecular<br />

Medicine Cologne (CMMC), University Hospital<br />

Cologne, University of Cologne, Cologne,<br />

<strong>German</strong>y; 5 Visual Coding Group, Max Planck<br />

Institute of Neurobiology, Martinsried, <strong>German</strong>y<br />

Congenital absence of cone photoreceptor<br />

function is associated with strongly impaired<br />

daylight vision and loss of color discrimination in<br />

human achromatopsia. Here, we introduce rAAVmediated<br />

gene replacement therapy as a<br />

potential treatment <strong>for</strong> this disease in the<br />

CNGA3 −/− mouse model. We show that such<br />

therapy can restore cone-specific visual<br />

processing in the CNS even if cone<br />

photoreceptors had been nonfunctional from<br />

birth. The restoration of cone vision was<br />

assessed at different stages along the visual<br />

pathway. Treated CNGA3 −/− mice became able to<br />

generate cone photoreceptor responses and to<br />

transfer these signals to bipolar cells. In support,<br />

we found morphologically that treated cones<br />

expressed regular CNG channel complexes and<br />

opsins in outer segments, which previously they<br />

did not. Moreover, expression of CNGA3<br />

normalized cGMP levels in cones, reduced the<br />

inflammatory response of Müller glia cells that is<br />

typical of retinal degenerations and delayed cone<br />

cell loss. Furthermore, ganglion cells from<br />

treated, but not from untreated CNGA3 −/− mice<br />

displayed cone-driven light-evoked spiking<br />

activity, indicating that signals generated in the<br />

outer retina are transmitted to the brain. Finally,<br />

we demonstrate that this newly acquired sensory<br />

in<strong>for</strong>mation was translated into cone-mediated<br />

vision-guided behavior.<br />

Session: On the Route to Clinical Application<br />

Or 7<br />

High Throughput Integration Site Analysis<br />

Reveals a Polyclonal Lineage-Specific<br />

Integration Site Distribution in a Successful<br />

WAS <strong>Gene</strong> <strong>Therapy</strong> Trial<br />

Anna Paruzynski 1 , Kaan Boztug 2 , Ali Nowrouzi 1 ,<br />

Claudia R. Ball 1 , Anne Arens 1 , Christina Lulay 1 ,<br />

Marie Böhm 2 , Sonja Naundorf 3 , Klaus Kühlcke 3 ,<br />

Rainer Blasczyk 4 , Irina Kondratenko 5 , Laszló<br />

Maródi 6 , Hanno Glimm 1 , Christoph Klein 2 ,<br />

Christof von Kalle 1 , Manfred Schmidt 1<br />

1Department of Translational Oncology, National<br />

Center <strong>for</strong> Tumor Diseases (NCT) and <strong>German</strong><br />

Cancer Research Center (DKFZ), Hannover,<br />

<strong>German</strong>y; 2 Department of Pediatric<br />

Hematology/Oncology, Hannover Medical<br />

School, Hannover, <strong>German</strong>y; 3 EUFETS AG, Idar-<br />

Oberstein, <strong>German</strong>y; 4 Institute <strong>for</strong> Transfusion<br />

Medicine, Hannover Medical School, Hannover,<br />

<strong>German</strong>y; 5 Department of Clinical Immunology,<br />

Russian Clinical Children's Hospital, Moscow,<br />

Russia; 6 Department of Infectious and Pediatric<br />

Immunology, Medical and Health Science Center,<br />

University of Debrecen, Debrecen, Hungary<br />

The Wiskott Aldrich Syndrome (WAS) is a<br />

complex primary immunodeficiency disease<br />

affecting both the lymphoid and myeloid<br />

compartment and leading to the development of<br />

thrombocytopenia, eczema, autoimmunity and an<br />

increased risk of malignancies. In a <strong>German</strong><br />

WAS clinical gene therapy trial, two patients were<br />

successfully treated by receiving autologous<br />

CD34 + cells transduced with a MLV-based vector<br />

encoding the WAS gene. By per<strong>for</strong>ming linear<br />

amplification-mediated PCR (LAM-PCR)<br />

combined with high-throughput sequencing, we<br />

were able to retrieve > 5700 unique integration<br />

sites (IS) <strong>for</strong> patient 1 (P1) and > 9500 unique IS<br />

<strong>for</strong> patient 2 (P2). In both patients, the genecorrected<br />

hematopoiesis showed a polyclonal IS<br />

pattern up to 3 years after gene therapy. The<br />

analysis of the clonal composition revealed a<br />

high clustering of IS in specific genomic regions<br />

with more than 70 common integration sites (CIS)<br />

of ≥ 10th order. Strikingly, this high clustering<br />

could be found amongst others within or close to<br />

the proto-oncogenes MDS1-EVI1 (P1: 81 IS; P2:<br />

94 IS), PRDM16 (P1: 10 IS; P2: 28 IS), LMO2<br />

(P1: 13 IS; P2: 29 IS) and CCND2 (P1: 11 IS; P2:<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong>


18 IS). A quantitative analysis further revealed<br />

that the most active cell clones showed a vector<br />

integration upstream of CCND2 in P1 and within<br />

the MDS1 locus in P2 constantly contributing to<br />

the gene-corrected hematopoiesis. The analysis<br />

of sorted cell fractions showed a distinct IS<br />

distribution with the appearance of IS within or<br />

close to the MDS1-EVI1 and PRDM16 gene loci<br />

almost exclusively in the myeloid fraction,<br />

whereas IS within or close to the LMO2 and<br />

CCND2 gene loci were preferentially detected in<br />

the lymphoid compartment. Even though we<br />

detected several IS within or close to already<br />

known proto-oncogenes, our IS analysis shows a<br />

highly polyclonal reconstitution of the<br />

hematopoietic system until the latest time point<br />

analyzed (1108 and 1071 days after gene<br />

therapy, respectively) without any apparent signs<br />

of clonal outgrowth. The monitoring of the clonal<br />

inventory and a large scale tracking of individual<br />

clones is ongoing to further study the efficacy and<br />

safety of the vector used <strong>for</strong> this clinical WAS<br />

gene therapy trial.<br />

Session: On the Route to Clinical Application<br />

Or 8<br />

Enhanced Therapeutic Neovascularization via<br />

AAV2.9/Thymosin β4: Evidence <strong>for</strong> a<br />

Myovascular Crosstalk<br />

Teresa Trenkwalder 1 , Rabea Hinkel 1 , Shahana<br />

Sultana 1 , Georg Stachel 1 , Achim Pfosser 1 ,<br />

Corinna Lebherz 1 , Ildiko Bock-Marquette 2 ,<br />

Elisabeth Deindl 3 , Christian Kupatt 1<br />

1Internal Medicine I, Klinikum Großhadern, LMU<br />

München, <strong>Munich</strong>, <strong>German</strong>y; 2 Southwestern<br />

Medical Center, Dallas, USA; 3 Walter-Brendel-<br />

Zentrum für Experimentelle Medizin, LMU<br />

München, <strong>Munich</strong>, <strong>German</strong>y<br />

Thymosin β4 (Tβ4) is known to be involved in<br />

wound healing, coronary vessel <strong>for</strong>mation and<br />

cardioprotection, alltogether implying an activated<br />

Protein Kinase B. We investigated the role of<br />

Thymosin β4 and AKT activation in angiogenesis<br />

and arteriogenesis using recombinant AAV2/9<br />

(rAAV) gene transfer in a model of chronic<br />

hindlimb ischemia. We hypothesise a<br />

myovascular crosstalk in which the Tβ4 induced<br />

stimulation of myocytes results in an enhanced<br />

neovascularisation. Methods: Mice (n = 8) were<br />

subjected to 3 × 10 12 r.AAV encoding <strong>for</strong> Tβ4, wt-<br />

AKT, AKT-DN, AKT-DN-Tet-off, GFP-Tet-off or<br />

Lac-Z 14 days be<strong>for</strong>e femoral artery ligation (d0).<br />

Recovery of blood flow was assessed by Laser<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong><br />

| 32<br />

Doppler flowmetry on day 3, 7 and 14 after<br />

ligation. To further study the reversibility of the<br />

AAV system, we generated Tet-off vectors<br />

encoding <strong>for</strong> GFP and AKTDN. Doxycycline<br />

(Dox) was applied from day 3 to day 14 after<br />

femoral ligation and thereby disabeling gene<br />

transcription. Capillary density was assessed via<br />

immunostaining <strong>for</strong> PECAM-1 (c/mf). Results:<br />

rAAV-based overexpression of Tβ4 (0.80 ± 0.13)<br />

or wt-AKT (0.79 ± 0.02) showed an increase in<br />

perfusion in flow measurements on day 7 in<br />

comparison to the control group (0.59 ± 0.1). The<br />

application of AKT-DN (0.43 ± 0.12) resulted in<br />

distinctly lower perfusion. Furthermore, capillary<br />

density was significantly increased in mice<br />

treated with Tβ4 (1.71 ± 0.05 c/mf) or wt-AKT<br />

(1.67 ± 0.06 c/mf) in comparison to the AKT-DN<br />

group (1.14 ± 0.08 c/mf) which stayed on control<br />

level (1.1 ± 0.01 c/mf). Using the inducible Tet-<br />

System GFP-positive myocytes were only<br />

detectable in the hindlimb muscles in the<br />

absence of Dox. Injection of rAAV.Tet-off-AKT<br />

reduced perfusion in the ischemic limb (0.3 ±<br />

0.01) in the absence of Dox, whereas application<br />

of Doxycyline from day 3 on normalized recovery<br />

(0.73 ± 0.04). We conclude that rAAV-Tβ4 or<br />

rAAV.AKT-transduction of skeletal myoctes<br />

activates a myovascular crosstalk resulting in<br />

enhanced angiogenesis and arteriogenesis. The<br />

Tet-off system, switching off gene transcription in<br />

the presence of Doxycycline, elucidates the<br />

relevance and reversibility of early AKT-inhibition<br />

<strong>for</strong> functional neovascularization.<br />

Session: <strong>Gene</strong> <strong>Therapy</strong>


33 |<br />

Or 9<br />

Nonviral Delivery of the Rat PDX1 <strong>Gene</strong> to Rat<br />

Liver For the In Vivo Transdifferentiation of<br />

Liver Cells to Pancreatic Beta-Cells<br />

Abdullah Cim 1 , Greta Sawyer 1 , Xiahong Zhang 1 ,<br />

Haibin Su 1 , Louise Collins 1 , Peter Jones 2 ,<br />

Michael Antoniou 3 , Hans J. Lipps 4 , John Fabre 4<br />

1 Department of Hepatology and Transplantation,<br />

King's College London, School of Medicine,<br />

London, UK; 2 Beta Cell Development & Function<br />

Group, King's College London, School of<br />

Biomedical & Health Sciences, London, UK;<br />

3 Department of Medical and Molecular <strong>Gene</strong>tics,<br />

King's College London, School of Medicine,<br />

London, UK; 4 Department of Cell Biology,<br />

University of Witten/Herdecke, Witten/Herdecke,<br />

<strong>German</strong>y<br />

Adenoviral vectors delivering single or multiple<br />

pancreatic transcription factors can induce liver<br />

cells or pancreatic exocrine cells to<br />

transdifferentiate towards pancreatic beta-cells.<br />

Nonviral approaches <strong>for</strong> gene delivery would be<br />

much more clinically applicable, but the level and<br />

longevity of gene expression necessary are<br />

unknown. We there<strong>for</strong>e evaluated five DNA<br />

expression constructs with different functional<br />

elements <strong>for</strong> the level and longevity of expression<br />

of the rat transcription factor pdx1 delivered<br />

hydrodynamically to rat liver. We also evaluated<br />

the degree of transdifferentiation towards a<br />

pancreas phenotype. Groups of three or four rats<br />

were harvested at days 1, 3, 7, 14 and 28. Pdx1<br />

from the plasmids and insulin 2 as a marker of<br />

transdifferentiation were assayed quantitatively<br />

using Taqman PCR. The pEPI plasmid, with a<br />

CMV promoter and a ≈2kb Scaffold/Matrix<br />

Attachment Region, gave strong pdx1 expression<br />

at day 1, but levels at day 3 were ≈150 fold lower.<br />

Substituting alpha1-antitrypsin promoters, with or<br />

without a ubiquitous chromatin opening element<br />

(UCOE), gave sustained pdx1 expression to day<br />

3, with a more gradual decline thereafter.<br />

However, actual levels of expression were 20 to<br />

300-fold lower than with the pEPI with CMV<br />

promoter. A CpG-depleted plasmid (InvivoGen)<br />

gave both high levels and sustained expression.<br />

Only the CpG-depleted plasmid induced a<br />

pancreatic phenotype in rat liver. Insulin 2<br />

expression 30 to 70-fold higher than day 1 liver<br />

was found in two of three rats at day 14. To<br />

conclude, nonviral delivery of pdx1 alone to liver<br />

in a plasmid giving sustained pdx1 expression<br />

gave weak insulin expression in some rats.<br />

Multiple transcription factors are currently under<br />

investigation.<br />

Session: <strong>Gene</strong> <strong>Therapy</strong><br />

Or 10<br />

Correction of Mpl Deficiency by Lentiviral<br />

Vectors with Lineage-Specific Expression<br />

Ute Modlich 1 , Dirk Heckl 1 , Daniel Wicke 1 ,<br />

Guntram Büsche 2 , Martijn Brugman 1 , Johann<br />

Meyer 1 , Axel Schambach 1 , Matthias Ballmaier 3 ,<br />

Christopher Baum 1<br />

1Department of Experimental Hematology,<br />

Hannover Medical School, Hannover, <strong>German</strong>y;<br />

2 Department of Pathology, Hannover Medical<br />

School, Hannover, <strong>German</strong>y; 3 Department of<br />

Pediatric Hematology, Hannover Medical School,<br />

Hannover, <strong>German</strong>y<br />

Congenital amegakaryocytic thrombocytopenia<br />

(CAMT) is a rare genetic disorder that is caused<br />

by inactivating mutations in the Thrombopoietin<br />

receptor MPL. Children with CAMT develop<br />

severe thrombocytopenia and aplastic anemia<br />

leading to early death. With the aim to develop a<br />

gene therapy <strong>for</strong> CAMT we retrovirally<br />

overexpressed Mpl. First experiments expressing<br />

Mpl C57Bl/6 wt mice caused severe adverse<br />

reactions (SAE), e.g., uncontrolled expansion of<br />

stem and progenitor cells (CMPD), stem cell<br />

alterations (MDS like phenotype) and increased<br />

frequency of insertional leukemias. To avoid<br />

those SAE, we developed self-inactivating<br />

lentiviral vectors that expressed Mpl by the<br />

lineage-specific human and murine Mpl or human<br />

GPIba promoter. For comparison we used the<br />

ubiquitously active PGK promoter. In vivo<br />

expression was restricted to megakaryocytes<br />

(CD41 + ) and hematopoietic stem cells (LSK<br />

CD34 − ). We established a murine model <strong>for</strong><br />

CAMT gene therapy based on the transplantation<br />

of Mpl −/− hematopoietic cells into Mpl −/− mice. Mpl<br />

expressed by the PGK promoter caused death 6<br />

days post transplantation (6/7 mice) due to<br />

spleen rupture after erythroid hyperproliferation<br />

showing the extreme sensitivity to ectopic Mpl<br />

expression. Mice transplanted with bone marrow<br />

(BM) that express Mpl from the lineage specfic<br />

promoters survived long term while mice that<br />

were transplanted with eGFP transduced Mpl −/−<br />

BM cells showed increased lethality during<br />

establishment of long term hematopoiesis (>12<br />

weeks). Both early post-transplant complications<br />

were prevented when using the lineage-specific<br />

promoters. Furthermore platelet production was<br />

restored in ≈ 50% of transplanted mice (n = 23)<br />

and multinucleated megakaryocytes were found<br />

in the BM 6-7 months after transplantation. In<br />

addition, the number LSK cells was significantly<br />

increased in Mpl-corrected animals. The<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong>


engraftment of secondary Mpl −/− recipients with<br />

Mpl corrected BM of primary mice gave further<br />

evidence <strong>for</strong> the correction of long term HSC. Our<br />

results show the potential of newly developed<br />

transcriptionally regulated lentiviral vectors to<br />

circumvent genotoxic and transgene related,<br />

potentially lethal adverse reactions.<br />

Session: <strong>Gene</strong> <strong>Therapy</strong><br />

Or 11<br />

LCMV-Pseudotyped VSV-Based Systems <strong>for</strong><br />

Treatment of Malignant Glioma<br />

Alexander Muik 1 , Inna Kneiske 1 , Tsanan<br />

Giroglou 1 , Gert Zimmer 2 , Stefan Momma 3 ,<br />

Dorothee von Laer 4<br />

1Applied Virology and <strong>Gene</strong> <strong>Therapy</strong> Unit, Georg-<br />

Speyer-Haus, Frankfurt am Main, <strong>German</strong>y;<br />

2 Institute of Virology and Immunoprophylaxis,<br />

Mittelhäusern, Switzerland; 3 Institute of<br />

Neurology, Johann Wolfgang Goethe University,<br />

Frankfurt am Main, <strong>German</strong>y; 4 Section of<br />

Virology, Innsbruck Medical University,<br />

Innsbruck, Austria<br />

Malignant glioma is the most frequent primary<br />

brain tumor and still has a very poor prognosis<br />

despite advances in neurosurgical resection and<br />

adjuvant radio- and chemotherapy. A promising<br />

new approach is the use of vesicular stomatitis<br />

virus (VSV)-based systems <strong>for</strong> glioma-targeted<br />

oncolytic virotherapy and/or gene therapy. To<br />

improve glioma specificity and to abrogate the<br />

VSV G-mediated VSV-inherent neurotropism, we<br />

pseudotyped VSV with the LCMV glycoprotein<br />

(LCMV GP). Retargeting to glioma cells while<br />

sparing healthy neurons was successfully shown<br />

in vitro, using a panel of different brain tumor cell<br />

lines, human neural as well as brain tumor stem<br />

cells and human primary neurons. The reduction<br />

of neurotropism was confirmed in vivo in a rat<br />

model after intracranial injection of replicationdeficient<br />

VSV-WT and LCMV-GP pseudotype<br />

vectors, encoding the eGFP gene. Furthermore,<br />

we investigated different systems to address viral<br />

spread and microdistribution of LCMV GPpseudotyped<br />

VSV within the tumor. Besides the<br />

generation of a replication-competent LCMV GP<br />

pseudotype rVSV(GP) as a pure virotherapy<br />

approach, we were able to show that tumorinfiltrating<br />

adult stem cells can be used as a<br />

delivery system and packaging cell line <strong>for</strong> noncytopathic,<br />

replication-deficient VSV vectors<br />

encoding the HSV-TK as suicide gene. Efficacy<br />

of rVSVΔG–TK(GP) producing progenitor cells in<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong><br />

| 34<br />

mediating tumor killing after ganciclovir treatment<br />

was shown in vitro in a G62 spheroid model. If<br />

the reduced neurotoxicity can be confirmed in our<br />

current studies in vivo, rVSV(GP) would be an<br />

extremely promising candidate <strong>for</strong> oncolytic<br />

virotherapy of brain cancer.<br />

Session: Cancer <strong>Gene</strong> <strong>Therapy</strong><br />

Or 12<br />

TBA<br />

Jennifer Altomonte 1 , Enrico DeToni 2 , Irene<br />

Esposito 1 , Claus Hellerbrand 3 , Sabrina Marozin 1 ,<br />

Oliver Ebert 1<br />

1II. Medizinische Klinik und Poliklinik, Klinikum<br />

rechts der Isar, TU München, <strong>Munich</strong>, <strong>German</strong>y;<br />

2 Klinikum Großhadern, LMU; München, <strong>Munich</strong>,<br />

<strong>German</strong>y; 3 Universita¨tsklinikum Regensburg,<br />

<strong>Munich</strong>, <strong>German</strong>y<br />

Session: Cancer <strong>Gene</strong> <strong>Therapy</strong><br />

Or 13<br />

An α-Fetoprotein Promoter Driven,<br />

Conditionally Replicating Adenovirus that<br />

Expresses the Sodium Iodide Symporter (NIS)<br />

<strong>for</strong> Radiovirotherapy of HCC<br />

Geoffrey K Grünwald 1 , Kathrin Klutz 1 , Michael J<br />

Willhauck 1 , Nathalie Wunderlich 1 , Reingard<br />

Senekowitsch-Schmidtke 2 , Manfred Ogris 3 , Per S<br />

Holm 4 , Burkhard Göke 1 , Christine Spitzweg 1<br />

1 Department of Internal Medicine II, Klinikum<br />

Grosshadern, Ludwig-Maximilians-University<br />

<strong>Munich</strong>, <strong>Munich</strong>, <strong>German</strong>y; 2 Department of<br />

Nuclear Medicine, Technical University <strong>Munich</strong>,<br />

<strong>Munich</strong>, <strong>German</strong>y; 3 Pharmaceutical<br />

Biotechnology, Department of Pharmacy,<br />

Ludwig-Maximilians-University, <strong>Munich</strong>,<br />

<strong>German</strong>y; 4 Institute of Experimental Oncology<br />

and <strong>Therapy</strong> Research, Klinikum rechts der Isar,<br />

TU <strong>Munich</strong>, <strong>Munich</strong>, <strong>German</strong>y<br />

We recently reported iodide accumulation and<br />

therapeutic efficacy of 131 I in hepatocellular<br />

carcinoma (HCC) cells stably expressing the NIS<br />

gene under control of the tumor-specific αfetoprotein<br />

promoter (AFP) <strong>for</strong> transcriptional<br />

targeting. As a next crucial step towards clinical<br />

application of this promising targeting technology


35 |<br />

we investigated in the current study the in vitro<br />

and in vivo efficacy of NIS gene transfer using a<br />

replication-selective oncolytic adenovirus. For<br />

this purpose NIS gene transfer was per<strong>for</strong>med in<br />

vitro and in vivo in human HCC cell (HepG2)<br />

xenografts, using a replication-selective oncolytic<br />

adenoviral vector carrying the NIS gene linked to<br />

a mouse AFP-promoter construct (Ad5-E3-AFP-<br />

NIS). In vitro experiments with HepG2 cells and<br />

control cell lines demonstrated high transduction<br />

efficiency and tumor selectivity of Ad5-E3-AFP-<br />

NIS. Functional NIS protein expression was<br />

confirmed by measurement of in vitro uptake of<br />

125<br />

I in HCC cells showing a 30-fold increase in<br />

perchlorate-sensitive iodide uptake activity as<br />

compared to control cell lines. A clonogenic<br />

assay with HCC cells infected with Ad5-E3-AFP-<br />

NIS (20 MOI) showed only 6.45 % survival after<br />

incubation with 0.8 mCi (29.6 MBq)<br />

131 I as<br />

compared to 100% survival of untreated HCC<br />

cells representing the high therapeutic potential<br />

of this gene therapy strategy. qPCR of NIS and<br />

fiber protein of the infected cells, a cytopathic<br />

effect assay and a spread assay showed<br />

corresponding results clearly demonstrating the<br />

replication selectivity due to the AFP promoter. In<br />

first in vivo experiments functional NIS protein<br />

expression after adenoviral NIS gene transfer<br />

was confirmed by 123 I gamma-camera imaging as<br />

well as qPCR. HepG2 cell xenografts in nude<br />

mice injected intratumorally with 5 × 10 8 PFU<br />

Ad5-E3-AFP-NIS accumulated approx. 11% ID/g<br />

(percentage injected dose per gram tumor tissue)<br />

48 h after virus injection with an average<br />

biological half-life of 5.5 h. These results clearly<br />

demonstrate the high potential of an α-fetoprotein<br />

promoter driven, conditionally replicating<br />

adenovirus <strong>for</strong> tumor-selective NIS gene delivery<br />

in liver cancer.<br />

Session: Cancer <strong>Gene</strong> <strong>Therapy</strong><br />

Or 14<br />

Tumor Stroma-Specific NIS <strong>Gene</strong> Delivery<br />

Using Mesenchymal Stem Cells<br />

Kerstin Knoop 1 , Marie Kolokythas 1 , Kathrin<br />

Klutz 1 , Michael J Willhauck 1 , Nathalie<br />

Wunderlich 1 , Dan Draganovici 2 , Christian Zach 3 ,<br />

Franz Josef Gildehaus 3 , Guido Böning 3 , Burkhard<br />

Göke 1 , Ernst Wagner 4 , Peter J Nelson 2 , Christine<br />

Spitzweg 1<br />

1Department of Internal Medicine II, Klinikum<br />

Grosshadern, Ludwig-Maximilians-University<br />

<strong>Munich</strong>, <strong>Munich</strong>, <strong>German</strong>y; 2 Clinical Biochemistry<br />

Group, Medical Policlinic, Ludwig-Maximilians-<br />

University <strong>Munich</strong>, <strong>Munich</strong>, <strong>German</strong>y;<br />

3 Department of Nuclear Medicine, Klinikum<br />

Großhadern, Ludwig-Maximilians-University<br />

<strong>Munich</strong>, <strong>Munich</strong>, <strong>German</strong>y; 4 Pharmaceutical<br />

Biotechnology, Department of Pharmacy,<br />

Ludwig-Maximilians-University, <strong>Munich</strong>, <strong>German</strong>y<br />

The tumor-homing property of mesenchymal<br />

stem cells (MSC) has suggested their potential<br />

use as a delivery vehicle <strong>for</strong> cancer therapeutics.<br />

Tracking the delivery and engraftment of MSCs<br />

into human tumors is a crucial prerequisite <strong>for</strong><br />

safety and effectiveness of MSCs as promising<br />

gene delivery vehicles. Due to its dual role as<br />

reporter and therapy gene, the sodium iodide<br />

symporter (NIS) allows non-invasive imaging of<br />

functional NIS expression by 123 I-scintigraphy and<br />

124 I-PET imaging, and thereby provides an<br />

excellent means to elucidate the capacity of<br />

MSCs <strong>for</strong> tumor targeting of therapeutic genes. In<br />

the current study we stably transfected human<br />

bone marrow derived CD34 − MSCs with a NISexpressing<br />

plasmid (CMV-NIS-pcDNA3) (NIS-<br />

MSC) followed by analysis of NIS expression and<br />

cytotoxicity of 131 I. In a human hepatocellular<br />

cancer (HCC) xenograft model (Huh7) we further<br />

investigated distribution and tumor recruitment of<br />

NIS-MSCs by I-123-scintigraphy and 124 I-PET<br />

imaging. NIS-MSCs revealed a 12-fold increase<br />

in perchlorate-sensitive iodide uptake activity as<br />

compared to wild-type MSCs. Further, Western<br />

Blot analysis confirmed NIS protein expression in<br />

NIS-MSCs. In mixed populations of NIS-MSC<br />

and HCC cells (ratio 1:1) a clonogenic assay<br />

showed a 50% reduction of HCC cell survival<br />

after 131 I application induced by its crossfire<br />

effect. After establishment of subcutaneous Huh7<br />

xenografts in nude mice, NIS-MSCs were<br />

injected via the tail vein. Gamma camera and<br />

PET imaging revealed active MSC recruitment<br />

into the tumor stroma as shown by tumorselective<br />

iodide accumulation (9.5% ID/g 123 I,<br />

biol. half-life 4h). Immunhistochemistry and ex<br />

vivo 123 I biodistribution analysis by gamma<br />

counter analysis confirmed active recruitment of<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong>


NIS-MSCs into the tumor stroma while non-target<br />

organs showed no significant MSC recruitment.<br />

Taken together, our results convincingly show<br />

tumor-specific radioiodine accumulation after<br />

MSC-mediated NIS gene delivery demonstrating<br />

tumor stroma-selective MSC recruitment. These<br />

results open the exciting prospect of tumorspecific<br />

NIS-mediated radionuclide therapy of<br />

thyroidal or extrathyroidal tumors using MSCs as<br />

gene delivery vehicles.<br />

Session: Tumor Biology and (Cancer) Stem Cells<br />

Or 15<br />

Monitoring and Excising Reprogramming<br />

Factors: A Novel Lentiviral Expression<br />

System <strong>for</strong> Reprogramming Strategies<br />

Axel Schambach 1 , Eva Warlich 1 , Johannes<br />

Kühle 1 , Tobias Cantz 2 , Martijn Brugman 1 , Tobias<br />

Mätzig 1 , Melanie Galla 1 , Adam Filipczyk 3 ,<br />

Hannes Klump 4 , Hans Schöler 5 , Timm<br />

Schroeder 3 , Christopher Baum 1<br />

1 Department of Experimental Hematology,<br />

Hannover Medical School, Hannover, <strong>German</strong>y;<br />

2 Junior Research Group Stem Cell Biology,<br />

Hannover Medical School, Hannover, <strong>German</strong>y;<br />

3 Institute of Stem Cell Research, Helmholtz<br />

Zentrum München, <strong>Munich</strong>, <strong>German</strong>y; 4 Institute<br />

<strong>for</strong> Transfusion Medicine, University Hospital<br />

Essen, Essen, <strong>German</strong>y; 5 Department of Cell<br />

and Developmental Biology, MPI <strong>for</strong> Molecular<br />

Biomedicine, Münster, Münster, <strong>German</strong>y<br />

Cellular identity is largely determined by<br />

transcription factor networks. The concerted<br />

action of 4 reprogramming factors (RFs: Oct4,<br />

Sox2, Klf4, c-Myc) leads to generation of induced<br />

pluripotent stem cells (iPSC), a promising<br />

resource <strong>for</strong> regenerative medicine and disease<br />

modelling. Here we constructed a modular<br />

versatile lentiviral vector system <strong>for</strong> mono- and<br />

polycistronic expression of these RFs (of either<br />

murine or human origin and optionally codonoptimized).<br />

To drive the reprogramming factors,<br />

we used the strong retroviral promoter SFFV,<br />

which enables high RF expression in most<br />

somatic cells and is frequently silenced in ES<br />

cells. To avoid the risks of residual permanent RF<br />

expression and insertional mutagenesis, we<br />

further improved our system by the inclusion of<br />

FRT, lox or rox sites to be able to remove the RF<br />

cassette through transient recombinase<br />

expression. Having the choice of 3 different<br />

recombinases offers the targeted shut off of RF<br />

at multiple desired time points. Using this system<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong><br />

| 36<br />

and taking advantage of an Oct4-GFP reporter<br />

model, we were able to generate murine and<br />

human iPS cells with high efficiency. To learn<br />

more about the kinetics of iPSC reprogramming<br />

and to understand better the underlying<br />

mechanisms, we developed color-coded RF<br />

vectors to follow expression kinetics qualitatively<br />

and quantitatively in relation to Oct4-GFP.<br />

Thereby, we could document the dynamic<br />

conversion of fibroblasts to pluripotent cells by<br />

fluorescence microscopy combined with longterm<br />

single cell tracking and alternatively highdefinition<br />

structural analysis of single iPSC<br />

colonies. Filming the “birth” of iPSC generation,<br />

we obtained movies, which show the potential<br />

genetic mosaic of early iPSC colonies and also<br />

indicate the necessity of stochastic epigenetic<br />

changes during the process of iPSC<br />

development. With the growing insights in cellspecific<br />

transcription factor networks our versatile<br />

vector system will be a good candidate <strong>for</strong> direct<br />

reprogramming approaches into somatic cells<br />

(e.g., neurons). In summary, with the described<br />

vector system we have an efficient tool in hand<br />

<strong>for</strong> “excisable” and safe reprogramming<br />

strategies in regenerative medicine.<br />

Session: Tumor Biology and (Cancer) Stem Cells<br />

Or 16<br />

MHC-Restricted Fratricide of Recipient<br />

Lymphocytes Expressing Transgenic T Cell<br />

Receptors Specific <strong>for</strong> the Apoptosis-Inhibitor<br />

Protein Survivin<br />

Matthias Leisegang 1 , Susanne Wilde 2 , Stefani<br />

Spranger 2 , Slavoljub Milosevic 2 , Bernhard<br />

Frankenberger 2 , Dolores J Schendel 2 , Wolfgang<br />

Uckert 1<br />

1 Max-Delbrück-Center <strong>for</strong> Molecular Medicine,<br />

Berlin, <strong>German</strong>y; 2 Helmholtz Zentrum München,<br />

<strong>Munich</strong>, <strong>German</strong>y<br />

Expression of T cell receptors (TCR) as<br />

transgenic proteins in peripheral blood<br />

lymphocytes (PBL) enables T cells with defined<br />

tumor specificity to be generated in high numbers<br />

<strong>for</strong> patient-individualized therapy (TCR gene<br />

therapy). Hereby, the selection of tumorassociated<br />

antigens (TAA) that can be effectively<br />

targeted on tumor cells is of central importance.<br />

Survivin, a well-characterized inhibitor of<br />

apoptosis, has been suggested as a candidate<br />

TAA because of its potential role in oncogenicity<br />

and its broad expression in most tumors but<br />

absence from most normal adult tissues. To


37 |<br />

explore use of survivin as a target antigen <strong>for</strong><br />

TCR gene therapy, we generated HLA-A2 allorestricted<br />

survivin-specific T cells with high<br />

functional avidity. We isolated several high<br />

affinity TCR recognizing peptides derived from<br />

survivin presented by HLA-A2 molecules.<br />

Following transfer of these TCR into HLA-A2-<br />

recipient lymphocytes, we found that the effector<br />

cells displayed excellent specific killing of HLA-<br />

A2 + survivin + tumor cells, indicating their<br />

therapeutic potential. Surprisingly, when these<br />

TCR were expressed as transgenic proteins in<br />

HLA-A2 + recipient lymphocytes, as they would<br />

be applied in the clinical setting, we discovered<br />

that the recipient lymphocytes underwent<br />

extensive apoptosis over time. This demise was<br />

caused by HLA-A2-restricted fratricide that<br />

occurred due to survivin expression in recipient<br />

lymphocytes, which created ligands <strong>for</strong><br />

transgenic TCR recognition. Furthermore, we<br />

could demonstrate that cytotoxic T cell clones of<br />

various specificities derived from HLA-A2 +<br />

donors were directly killed by survivin-specific<br />

TCR-modified PBL. There<strong>for</strong>e, survivin-specific<br />

TCR gene therapy would be limited to application<br />

in HLA-A2-mismatched stem cell transplantation.<br />

However, these results raise a general question<br />

regarding cancer vaccines targeting proteins that<br />

are also expressed in activated lymphocytes,<br />

since induction of high avidity T cells that expand<br />

in lymph nodes following vaccination or later<br />

accumulate at tumor sites might limit themselves<br />

by self-MHC-restricted fratricide and also<br />

eliminate neighboring T cells of other<br />

specificities.<br />

Session: Cancer Immune <strong>Therapy</strong> and T-Cell<br />

<strong>Therapy</strong><br />

Or 17<br />

Integration Site Analysis of Reprogrammed T<br />

Cells in a Mouse Model of T Cell Receptor<br />

<strong>Gene</strong> <strong>Therapy</strong> Developing Graft Versus Host<br />

Disease<br />

Eliana Ruggiero 1 , Gavin M Bendle 2 , Ton N.<br />

Schumacher 2 , Hanno Glimm 1 , Christof von<br />

Kalle 1 , Manfred Schmidt 1 , Carsten Linnemann 1 ,<br />

Laura Bies 1<br />

1<strong>German</strong> Cancer Research Center and National<br />

Center <strong>for</strong> Tumor Diseases, Heidelberg,<br />

<strong>German</strong>y; 2 The Netherlands Cancer Institute,<br />

Amsterdam, The Netherlands, Amsterdam, The<br />

Netherlands<br />

The genetic modification of immune cells and the<br />

generation of recombinant vectors encoding<br />

cancer antigens have highlighted the great<br />

potential of immunotherapy in treating cancer.<br />

Since the ability of a T cell to specifically<br />

recognize an antigen relies on the T cell receptor<br />

(TCR), a tumor reactive T cell repertoire can be<br />

generated by the genetic introduction of<br />

transgenes encoding TCR directed towards<br />

specific tumor antigens. We applied our<br />

experience in the comprehensive genome wide<br />

retrieval of gammaretroviral insertions sites (IS)<br />

to analyze the integration site distribution and the<br />

potential influences on the clonal fate of<br />

transduced cells after TCR gene transfer in order<br />

to assess their safety <strong>for</strong> clinical trials. We<br />

per<strong>for</strong>med IS analysis combined with highthroughput<br />

sequencing on mouse CD4 and CD8<br />

T cells transduced with a gamma retroviral vector<br />

encoding an ovalbumin-specific OT-I TCR. Mice<br />

receiving IL-2 administration after OT-I TCR<br />

transplantation developed Graft-versus-Host-<br />

Disease (GvHD). In total, we found 841 unique IS<br />

in OT-I transduced mice and 1063 unique IS in<br />

control GFP transduced mice with a higher<br />

clustering of IS in genes involved in the immune<br />

response (e.g., Cd28, Ctla4) in OT samples<br />

compared to the control samples. Results from<br />

the Ingenuity Pathways Analysis indicated the<br />

presence of specific gene classes, Hematological<br />

disease and Inflammatory response,<br />

preferentially targeted in mice transduced with<br />

the OT-I encoding vector compared to the mice<br />

transduced with the GFP vector (p = 7.84*10 −4<br />

and p = 2.13*10 −2 , respectively). In GvHD<br />

developing mice we observed a higher frequency<br />

of IS located in gene and in the surrounding 10 kb<br />

compared to the control counterpart (74% and<br />

65%, respectively; p = 3*10 −2 ) and an increased<br />

contribution of integration clones to genecorrected<br />

hematopoiesis, indicating the<br />

occurrence of a potential in vivo skewing. Further<br />

sequencing and statistical analyses are ongoing.<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong>


Our results provide important insights in IS<br />

distribution, clonal diversity and identification of<br />

genes preferentially targeted by TCR specific<br />

gamma retroviral vectors.<br />

Session: Cancer Immune <strong>Therapy</strong> and T-Cell<br />

<strong>Therapy</strong><br />

Or 18<br />

Luciferase-Based Dual Bioluminescence<br />

Imaging of Tumor Metastases After Systemic<br />

Transgene Delivery with a Synthetic <strong>Gene</strong><br />

Carrier<br />

Katarina Farkasova 1 , Arzu Cengizeroglu 1 , Rudolf<br />

Haase 1 , Martina Anton 2 , Joachim Ellwart 3 , Ernst<br />

Wagner 1,4 , Manfred Ogris 1,4<br />

1 Pharmaceutical Biotechnology, Department of<br />

Pharmacy, Ludwig-Maximilians-University,<br />

<strong>Munich</strong>, <strong>German</strong>y; 2 Institute of Experimental<br />

Oncology and <strong>Therapy</strong> Research, Klinikum<br />

rechts der Isar, TU <strong>Munich</strong>, <strong>Munich</strong>, <strong>German</strong>y;<br />

3 Institute of Molecular Immunology, Helmholtz<br />

Center <strong>Munich</strong>, <strong>Munich</strong>, <strong>German</strong>y; 4 Center <strong>for</strong><br />

Nanoscience (CeNS), Ludwig-Maximilians-<br />

University, <strong>Munich</strong>, <strong>German</strong>y<br />

Noninvasive, biooptical in vivo imaging is a<br />

powerful method to study the biodistribution and<br />

pharmacodynamics of labeled drugs in the living<br />

organism. For studies on nucleic acid delivery,<br />

the use of luciferase gene allows the highly<br />

sensitive detection of transgene expression by<br />

bioluminescence imaging (BLI). Here we<br />

describe a method, which allows both monitoring<br />

the growth of tumor metastases and the<br />

visualization of transgene expression after<br />

intravenous delivery of nonviral gene vectors in a<br />

syngeneic mouse tumor model. Murine<br />

neuroblastoma cells (Neuro2A) were stably<br />

transduced with a firefly luciferase and enhanced<br />

green fluorescent protein (EGFP) encoding<br />

lentiviral vector. EGFP sorted cells were injected<br />

either intravenously or intrasplenic into A/J mice<br />

and the tumor growth was monitored by BLI after<br />

intraperitoneal injection of luciferin, the substrate<br />

<strong>for</strong> firefly luciferase. After development of well<br />

vascularized tumor metastases, polyplexes<br />

containing a novel plasmid encoding <strong>for</strong><br />

membrane-bound gaussia luciferase (Santos et<br />

al., 2009, Nature Medicine 15, 338–344) and a<br />

polycationic, biodegradable gene carrier were<br />

applied via the tail vein. Gaussia transgene<br />

expression was monitored after intravenous<br />

injection of its substrate coelenterazine. Due to<br />

the different substrates applied <strong>for</strong> firefly and<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong><br />

| 38<br />

gaussia luciferases, we could clearly distinguish<br />

between the firefly luciferase signal from the<br />

labeled tumor cells and the gaussia luciferase<br />

signal from cells expressing the gaussia<br />

transgene. After intravenous injection of<br />

polyplexes, gaussia transgene was only<br />

observed in areas were firefly luciferase labeled<br />

tumors were located, whereas in tumor-free<br />

animals no significant gaussia signal was<br />

observed. Due to a time lag between luciferase<br />

and coelenterazine injection, crosstalk between<br />

firefly and gaussia BLI signal was avoided. This<br />

study clearly shows, that polyplexes based on<br />

biodegradable polycations can selectively<br />

transfect well vascularized tumor metastases and<br />

dual BLI allows to distinguish between tumor<br />

localization and transgene expression.<br />

Session: Pharmacology and Toxicology<br />

Or 19<br />

Retroviral and Transposon-Based Tet-<br />

Regulated All-In-One Vectors with Reduced<br />

Background Expression and Improved<br />

Dynamic Range<br />

Niels Heinz 1 , Rainer Loew 2 , Christopher Baum 1 ,<br />

Bernhard Schiedlmeier 1<br />

1 Experimental Hematology, Hannover Medical<br />

School, Hannover, <strong>German</strong>y; 2 EUFETS GmbH,<br />

Idar-Oberstein, <strong>German</strong>y<br />

The constitutive expression of therapeutic genes<br />

might interfere with cell fate in vivo. There<strong>for</strong>e,<br />

regulated transgene expression is of major<br />

interest <strong>for</strong> gene therapy. Such a system needs<br />

to provide tightly controlled inducible promoters,<br />

as shown <strong>for</strong> the tetracycline regulatory system<br />

(tet-system). However, its application requires the<br />

introduction of two components into the target<br />

cell genome: the tet-responsive transactivator<br />

and the regulated expression cassette. In order<br />

to render the tet-system valuable <strong>for</strong> approaches<br />

in gene therapy, preselection of transactivator<br />

expressing cells has to be avoided, thus both<br />

components <strong>for</strong> regulated transgene expression<br />

have to be provided by a single vector. Up to<br />

date, published All-In-One vectors led to<br />

regulatory windows of around 500-fold, even after<br />

selection <strong>for</strong> single cell clones, thereby displaying<br />

a relatively high signal-to-noise ratio. In this<br />

study, we develop optimized All-In-One vectors<br />

which display tight transgene regulation without<br />

the need <strong>for</strong> clonal selection. Improvements in<br />

the dynamic range could be achieved through<br />

modifications of the vector architecture combined


39 |<br />

with the introduction of newly developed tetresponsive<br />

promoters, Ptet. These optimizations<br />

increased the dynamic range to levels up to<br />

14,000-fold <strong>for</strong> retroviral and 25,000-fold <strong>for</strong><br />

nonviral transposon vectors in non-clonal human<br />

cell lines, and up to 2,800-fold in murine<br />

hematopoietic cell lines. This improved regulation<br />

was based on a strong reduction of background<br />

expression in the off-state, even if cells were<br />

transduced at high multiplicity of infection, while<br />

induction remained at high levels. Our results<br />

indicate that an optimized signal-to-noise ratio of<br />

transgene expression in different target cells<br />

depends on vector architecture, the choice of the<br />

inducible promoter and the integration pattern.<br />

Session: Pharmacology and Toxicology<br />

Or 20<br />

High-Throughput Integration Site Analysis <strong>for</strong><br />

Vector Biosafety Assessment in CGD <strong>Gene</strong><br />

<strong>Therapy</strong><br />

Simone J. Scholz 1 , Margarita Diaz 2 , Stefan<br />

Stein 2 , Axel Schambach 3 , Hanno Glimm 1 ,<br />

Christof von Kalle 1 , Manuel Grez 2 , Manfred<br />

Schmidt 1<br />

1<strong>German</strong> Cancer Research Center and National<br />

Center <strong>for</strong> Tumor Diseases, Heidelberg,<br />

<strong>German</strong>y; 2 Institute <strong>for</strong> Biomedical Research,<br />

Georg-Speyer-Haus, Frankfurt am Main,<br />

<strong>German</strong>y; 3 Experimental Hematology, Hannover<br />

Medical School, Hannover, <strong>German</strong>y<br />

In several (pre)clinical gene therapy trials<br />

insertional mutagenesis by integrating<br />

gammaretroviral vectors was observed. In a<br />

clinical trial <strong>for</strong> the treatment of X-linked chronic<br />

granulomatous disease (X-CGD) insertional<br />

activation of MDS1/EVI1 has led to the clonal<br />

expansion of gene marked myeloid progenitor<br />

cells, triggering the development of monosomy 7<br />

and a myelodysplastic syndrome in both patients<br />

enrolled. There<strong>for</strong>e, a new self-inactivating (SIN)<br />

vector containing an internal cellular promoter (cfes)<br />

and a codon-optimized transgene<br />

(SINfes.gp91s) was developed <strong>for</strong> the treatment<br />

of X-CGD improving the safety and efficacy of<br />

gene delivery (Manuel Grez, Georg-Speyer-<br />

Haus, Frankfurt). As part of the preclinical testing<br />

we per<strong>for</strong>med high-throughput integration site<br />

(IS) analysis and deep sequencing (454/Roche)<br />

with associated bioin<strong>for</strong>matical tools on mice<br />

transplanted with gene modified lineage markernegative<br />

hematopoietic stem cells. As a control a<br />

full LTR SFFV-promoter driven vector<br />

(SF91eGFP) – as it was used in the previous trial<br />

– was analyzed in parallel. In total, we found<br />

1159 unique IS in SF91eGFP transduced mice (n<br />

= 4) and 2261 unique IS in SINfes.gp91s<br />

transduced mice (n = 5). While we did not<br />

observe any significant differences in the<br />

integration site distribution between the vectors,<br />

high-throughput sequencing revealed a less<br />

polyclonal IS pattern in SF91eGFP mice<br />

compared to the SINfes.gp91s transplanted mice<br />

(in average 281 vs. 442 unique IS/mouse).<br />

Furthermore, we found an increased frequency of<br />

(pre)dominant clones in mice transduced with the<br />

SF91eGFP vector than with SINfes.gp91s.<br />

Moreover, SF91eGFP integrants are more<br />

frequently found in/near EVI1/MDS1 than<br />

SINfes.gp91s integrants (9 of 1065 and 2 of<br />

2115, respectively; p = 6.7*10 −4 ). Finally, we could<br />

observe an increased in vivo skewing in<br />

secondary recipients of SF91eGFP transduced<br />

mice compared to SINfes.gp91s (p = 2.9*10 −6 ).<br />

Overall, our results showed no signs of obvious<br />

vector-induced side-effects (no loss of<br />

polyclonality, no obvious skewing) <strong>for</strong> the<br />

SINfes.gp91s vector. * contributed equally<br />

Session: Pharmacology and Toxicology<br />

Or 21<br />

Modification of the Factor X Serine Protease<br />

Domain Ablates Heparan Sulfate<br />

Proteoglycan Engagement by Ad5-FX<br />

Complexes<br />

Margaret R. Duffy 1 , A.C. Bradshaw 1 , A.L.<br />

Parker 1 , S.A. Nicklin 1 , J.H. McVey 2 , A.H. Baker 1<br />

1Glasgow Cardiovascular Research Centre,<br />

University of Glasgow, Glasgow, UK;<br />

2 Thrombosis Research Institute, London, UK<br />

Adenoviruses are commonly used vectors in<br />

gene therapy clinical trials, the majority of which<br />

are based on serotype 5. Following intravascular<br />

administration in vivo Ad5 exhibits high<br />

sequestration to the liver and spleen. Recent<br />

studies show that hepatocyte transduction by<br />

Ad5 is mediated by a high affinity interaction<br />

between coagulation factor X (FX) and the<br />

adenovirus 5 hexon proteins bridging the<br />

adenovirus capsid to the cell hepatocyte surface<br />

putatively through interaction with heparan<br />

sulfate proteoglycan receptors (HSPGs).<br />

Pharmacological blockade of the heparin-binding<br />

exosite in the serine protease (SP) domain of FX<br />

prevents cell surface binding and gene transfer<br />

mediated through FX1. However the exact<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong>


HSPG-interacting residues in the SP domain of<br />

FX require further elucidation. Previous studies<br />

have indicated that the basic residues Arg-316,<br />

Lys-319, Arg-349, Arg-390, Lys-394, Lys-463 and<br />

Arg-467 of FX constitute the exosite in the SP<br />

domain2. We there<strong>for</strong>e sought to generate a FX<br />

mutant and assess the importance of this exosite.<br />

A human FX plasmid construct with mutations at<br />

Arg-316, Lys-319, Arg-349, Arg-390, Lys-394,<br />

Lys-463 and Arg-467 was generated and<br />

confirmed by sequence analysis. Recombinant<br />

FX (mutant and wildtype) was produced by<br />

plasmid transfection into 293T cells in the<br />

presence of vitamin K with collection of the<br />

supernatant. Enzyme-linked immunosorbent<br />

assay was used to quantify the rFX. In vitro<br />

assays demonstrated that the exosite mutations<br />

blocked the ability of FX to enhance transduction<br />

of Ad5 thus confirming the importance of this<br />

exosite in HSPG engagement. [1] Waddington<br />

2008 Cell 132,397-409; [2] Rezaie 2000 JBC<br />

132,3320-3327.<br />

Session: Pharmacology and Toxicology<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong><br />

| 40


41 |<br />

DG-GT 2010 Poster Presentations<br />

P 1<br />

Development of a Stable Lyophilized<br />

Formulation <strong>for</strong> LPEI Polyplexes<br />

Julia C Kasper 1 , David Schaffert 2 , Manfred<br />

Ogris 2 , Ernst Wagner 2 , Wolfgang Frieß 1<br />

1 Department of Pharmacy, Pharmaceutical<br />

Technology and Biopharmaceutics, Ludwig-<br />

Maximilians-University <strong>Munich</strong>, <strong>Munich</strong>,<br />

<strong>German</strong>y; 2 Department of Pharmacy,<br />

Pharmaceutical Biotechnology, Ludwig-<br />

Maximilians-University <strong>Munich</strong>, <strong>Munich</strong>, <strong>German</strong>y<br />

Polyplexes based on linear polyethylenimine<br />

(LPEI) are efficient non-viral gene delivery<br />

systems but the requirement <strong>for</strong> freshly prepared<br />

<strong>for</strong>mulations prior to administration due to the<br />

instability in aqueous suspension is a major<br />

drawback. There<strong>for</strong>e, the aim was to establish an<br />

up-scaled and reproducible method <strong>for</strong> the<br />

preparation of standardized batches of welldefined<br />

LPEI polyplexes and to develop isotonic<br />

lyophilized polyplex <strong>for</strong>mulations with long-term<br />

stability. Polyplexes consisting of pCMVLuc<br />

plasmid and 22 kDa LPEI were prepared by<br />

classical hand pipetting or using an up-scaled<br />

micro-mixer method at different mixing speeds.<br />

Freeze-thawing and lyophilization were<br />

per<strong>for</strong>med on a laboratory scale freeze-drier.<br />

Several excipients at varying concentrations were<br />

tested <strong>for</strong> their cryo- and lyoprotectant potential.<br />

To evaluate long-term stability, lyophilized<br />

samples were stored at 2-8°C, 20°C and 40°C <strong>for</strong><br />

6 weeks. The z-average diameter and<br />

polydispersity index (PdI) of the polyplexes was<br />

measured by dynamic light scattering.<br />

Transfection efficiency was evaluated in murine<br />

neuroblastoma (Neuro 2A) cells and metabolic<br />

activity as an indicator <strong>for</strong> cell toxicity was<br />

analyzed. By using the up-scaled micro-mixer<br />

method with varied mixing speeds, size (59–197<br />

nm) and PdI (0.05-0.19) of the polyplexes can be<br />

directly controlled. When <strong>for</strong>mulated with 14%<br />

lactosucrose, 10% HP-beta-CD/6.5% sucrose or<br />

10% povidone/6.3% sucrose at isotonic<br />

concentrations, the increase in polyplex size was<br />

less than 50% after freeze-thawing, freeze-drying<br />

and during storage. Transfection efficiency and<br />

metabolic activity was high when polyplexes were<br />

<strong>for</strong>mulated with lactosucrose or HP-beta-<br />

CD/sucrose but decreased when PVP/sucrose<br />

was used as stabilizer. The possibility to<br />

reproducibly manufacture large standardized<br />

batches of well-defined, transfection efficient<br />

polyplexes with long-term stability by using the<br />

established up-scaled preparation method<br />

followed by lyophilization is an important step<br />

closer from promising technology to clinical<br />

application.<br />

Session: Vector Development<br />

P 2<br />

Recombinant Influenza C Virus: A New<br />

Prototype of a Melanoma-Specific Oncolytic<br />

Virus<br />

Karin Pachler, Juliane Mayr, Reinhard Vlasak<br />

University of Salzburg, Department of Molecular<br />

Biology, Salzburg, Austria<br />

The ability to genetically engineer viruses<br />

contributes greatly to our understanding of their<br />

life cycle and allows creation of virus-derived<br />

vectors <strong>for</strong> vaccine development, gene therapy or<br />

oncolysis. In the past decade, several reversegenetics<br />

systems have been developed to<br />

manipulate the genomes of influenza A and B<br />

viruses. Here, we introduce a system <strong>for</strong> the<br />

generation of recombinant influenza C viruses<br />

from seven plasmids.The seven full-length viral<br />

cDNAs were cloned into an RNA pol I/pol IIbased<br />

bidirectional vector. Transfection of Vero<br />

cells and subsequent amplification on MDCK<br />

cells yielded viral HA titres of 128. With this<br />

efficient method, mutations or <strong>for</strong>eign genes may<br />

be easily incorporated into the influenza C virus<br />

genome. Next, the influenza C virus protein NS1<br />

was identified as a factor antagonising the<br />

cellular interferon (IFN) signalling, and the<br />

regions of NS1 necessary <strong>for</strong> IFN inhibition were<br />

mapped. Several melanoma cell lines have a<br />

defective IFN signalling and express the cellular<br />

receptor <strong>for</strong> influenza C virus, which is 9-O-acetyl<br />

sialic acid. Thus NS1-deficient recombinant<br />

influenza C viruses could be engineered to serve<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong>


as gene delivery vectors and/or tumour-specific<br />

oncolytic viruses in the future.<br />

Session: Vector Development<br />

P 3<br />

Live-Cell Imaging and Single-Particle<br />

Tracking of Polyplex Internalization<br />

Nadia Ruthardt 1 , Karla de Bruin 1 , Ernst<br />

Wagner 2,3 , Christoph Bräuchle 1,3<br />

1Physical Chemistry, Department of Chemistry,<br />

Ludwig-Maximilians-University, <strong>Munich</strong>,<br />

<strong>German</strong>y; 2 Pharmaceutical Biotechnology,<br />

Department of Pharmacy, Ludwig-Maximilians-<br />

University, <strong>Munich</strong>, <strong>German</strong>y; 3 Center <strong>for</strong><br />

Nanoscience (CeNS), Ludwig-Maximilians-<br />

University, <strong>Munich</strong>, <strong>German</strong>y<br />

We investigate the uptake of targeted and<br />

untargeted polymeric gene vectors (polyplexes)<br />

by highly sensitive fluorescence microscopic<br />

methods by live cell imaging on a single cell<br />

level. The epidermal growth factor receptor<br />

(EGFR) is overexpressed on a high percentage<br />

of human carcinomas and is there<strong>for</strong>e an<br />

attractive therapeutic target <strong>for</strong> tissue-specific<br />

targeting by non-viral vectors in cancer gene<br />

therapy. By comparing uptake kinetics and<br />

internalization dynamics, single particle tracking<br />

in combination with quenching experiments<br />

revealed typical three-phase dynamics of the<br />

uptake process independent of targeting. Phase I<br />

was characterized by slow, actin-cytoskeletonmediated<br />

movement of the particles with drift and<br />

included the internalization process. During<br />

phase II, particles displayed increased velocities<br />

with confined and anomalous diffusion in the<br />

cytoplasm. Phase III was characterized by fast<br />

active transport along microtubules. Targeting of<br />

polyplexes <strong>for</strong> receptor-mediated endocytosis by<br />

the EGF receptor resulted in shortening of phase<br />

I and strongly accelerated internalization.<br />

Targeted as well as untargeted particles were<br />

transported in early endosomes marked by Rab5-<br />

GFP and accumulated in late endosomes marked<br />

by Rab9-GFP. The endosomal release dynamics<br />

of polyplexes consisting of DNA condensed with<br />

the cationic polymers linear polyethyleneimine<br />

(LPEI), poly-(l)-lysine (PLL) or poly-(d)-lysine<br />

(PDL) were studied by photochemical release in<br />

living cells. Using double-labeled polyplexes,<br />

DNA and polymer were imaged simultaneously<br />

by dual-color fluorescence microscopy. Our<br />

results demonstrate that the characteristics of the<br />

cationic polymer significantly influence the<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong><br />

| 42<br />

release behavior of the polyplexes. For<br />

LPEI/DNA particles, LPEI quickly spread<br />

throughout the cytosol, whereas DNA was<br />

released slowly and remained immobile<br />

thereafter. In case of PLL particles, both DNA<br />

and polymer showed quick release. PDL particles<br />

remained condensed upon photosensitizer<br />

activation.<br />

Session: Vector Development<br />

P 4<br />

<strong>Gene</strong>ration of the Novel, Synthetic Hybrid<br />

SCE-Promoter <strong>for</strong> <strong>Gene</strong>-Therapeutical<br />

Applications<br />

Rudolf Haase, Terese Magnusson, Ernst<br />

Wagner, Manfred Ogris<br />

Pharmaceutical Biotechnology, Department of<br />

Pharmacy, Ludwig-Maximilians-University,<br />

<strong>Munich</strong>, <strong>German</strong>y<br />

A major factor <strong>for</strong> successful gene therapy is<br />

strong and enduring expression of the transgene.<br />

Commonly used promoters <strong>for</strong> expression of<br />

transgenes are the CMV-immediate early (CMV-<br />

IE), chicken β-actin, ubiquitin C or the EF1α<br />

promoter, each with their own advantages and<br />

disadvantages. Here we present the generation<br />

of novel synthetic hybrid promoter, based on the<br />

CMV-IE and the EF1α promoter. It was obtained<br />

by generating shuffled consensus sequences of<br />

both promoters, which were analyzed by<br />

promoter prediction software. One sequence, the<br />

shuffle CMV/EF1α promoter (SCE), was<br />

synthesized and tested in vitro and in vivo. The<br />

SCEP sequence features an identity of 83% with<br />

the CMV and 85% with the EF1α promoter. The<br />

sequence was CpG-free designed to circumvent<br />

inflammatory reactions towards CpG isles and<br />

promoter methylation leading to silencing. In<br />

several cell lines a 2-7 fold stronger expression of<br />

the SCE promoter compared to the EF1α was<br />

observed; when compared to CMV-IE, SCE<br />

promoter activity was almost similar. All<br />

promoters were within a CpG-free plasmid<br />

featuring a human CMV-IE enhancer and<br />

luciferase as transgene. In immunocompetent<br />

Balb/c mice we observed a ≈3 fold stronger<br />

transgene expression of SCEP compared to the<br />

EF1α promoter after hydrodynamic gene delivery<br />

to the liver <strong>for</strong> a time period of 30 days. Hence<br />

the SCE promoter is qualified <strong>for</strong> further<br />

applications in biotechnology or gene therapy,<br />

when strong and enduring transgene expression<br />

and evasion of promoter silencing is needed.


43 |<br />

Session: Vector Development<br />

P 5<br />

pH-Labile Degradable Pseudodendritic<br />

Polycations <strong>for</strong> In Vitro and In Vivo DNA<br />

Delivery into Cancer Cells<br />

Christian Dohmen, Thomas Fröhlich, Ruggero<br />

Foralosso, Ernst Wagner<br />

Pharmaceutical Biotechnology, Department of<br />

Pharmacy, Ludwig-Maximilians-University,<br />

<strong>Munich</strong>, <strong>German</strong>y<br />

Polycationic dendritic structures show good<br />

properties <strong>for</strong> DNA delivery into cancer cells. For<br />

example branched polyethylenimines (bPEI) are<br />

known to have a good transfection ability but also<br />

high toxicity. Former work of our group (Russ et<br />

al., <strong>Gene</strong> <strong>Therapy</strong> 2008) indicated that<br />

pseudodendrimers containing degradable ester<br />

linkages have similar transfection efficiency but<br />

lower toxicity. Within this work we designed novel<br />

pseudodendrimers based on different polyamines<br />

such as polypropylenimine and oligoethylenimine<br />

as core structure. They were modified with the<br />

acid degradable bisacrylate acetal linker BAA<br />

and oligoethylenimine (OEI 800Da) as<br />

polycationic surface structure. We could show<br />

that these dendritic molecules have the ability to<br />

<strong>for</strong>m stable polyplexes with DNA and are<br />

degradable under incubation at endosomal pH.<br />

Cell testing confirmed that this degradability<br />

leads to a lower intracellular toxicity, while<br />

keeping the transfection ability both in vitro as<br />

well as in vivo in a neuroblastoma mouse tumor<br />

model.<br />

Session: Vector Development<br />

P 6<br />

Biodegradable Pseudodendritic Oligoamines<br />

<strong>for</strong> siRNA Delivery<br />

Thomas Fröhlich, Alexander Philipp, Verena<br />

Ruß, Ernst Wagner<br />

Pharmaceutical Biotechnology, Department of<br />

Pharmacy, Ludwig-Maximilians-University,<br />

<strong>Munich</strong>, <strong>German</strong>y<br />

Searching <strong>for</strong> efficient synthetic vectors <strong>for</strong> siRNA<br />

delivery, a series of pseudodendritic oligoamines<br />

with different hydrophilic/lipophilic properties was<br />

synthesized. The polymers consist of an 800 Da<br />

oligoethylenimine (OEI) core and various surface<br />

molecules (spermine, spermidine, OEI, etc.),<br />

attached to the core via ester bond containing<br />

linkers. Out of this series, the HD-S polymer most<br />

effectively facilitates siRNA delivery, silencing<br />

luciferase expression in stably transfected<br />

Neuro2A-luc cells. All presented polymers are<br />

biodegradable because of the incorporated ester<br />

bonds, which can be cleaved by esterases<br />

ubiquitously present in living organisms.<br />

Ineffective polymers like the HD-O can be<br />

trans<strong>for</strong>med into successful delivery agents by<br />

crosslinking the surface amines of the polyplexes<br />

with DSP (Dithiobis-succinimidyl-propionate).<br />

This lateral stabilization of polyplexes boosts the<br />

delivery efficiency of HD-O, which is<br />

demonstrated on Neuro2A-luc cells and HUH-7luc<br />

hepatoma cells. Enhanced cellular uptake of<br />

the crosslinked particles is proven by FACS<br />

analysis and gene down regulation is analyzed<br />

on mRNA level by qPCR. The disulfide bond in<br />

DSP maintains the biodegradability of the<br />

polymers and ensures sufficient release of the<br />

siRNA out of the stabilized polyplexes due to<br />

cleavage of disulfide bonds in the reductive<br />

cytosolic environment.<br />

Session: Vector Development<br />

P 7<br />

Succinylated PEI as Carrier <strong>for</strong> siRNA<br />

Delivery in Tumor Xenograft Models:<br />

Evaluation by RT-qPCR<br />

Daniel Edinger, Alexander Philipp, Katarina<br />

Farkasova, Ernst Wagner<br />

Pharmaceutical Biotechnology, Department of<br />

Pharmacy, Ludwig-Maximilians-University,<br />

<strong>Munich</strong>, <strong>German</strong>y<br />

Developing polymers <strong>for</strong> tumor targeted delivery<br />

of siRNA, succinylated 25 kDa PEI has emerged<br />

as highly effective carrier system. Mechanistic in<br />

vitro study demonstrated that lower toxicity of the<br />

polymer compared to standard 25 kDa PEI is<br />

mainly responsible <strong>for</strong> the enhanced siRNA<br />

delivery efficacy. Higher amounts of polymer can<br />

be applied, which result in improved endosomal<br />

escape characteristics. For in vivo knockdown<br />

studies in xenograft tumor models the<br />

establishment of accurate and reliable mRNA<br />

detection methods is important. For this purpose<br />

we optimized Reverse Transcription (RT)<br />

quantitative Polymerase Chain Reaction (qPCR).<br />

Variations in expression levels of reporter genes<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong>


(luciferase), therapeutic genes (RAN, KSP) and<br />

housekeeping genes (ACTB, GAPDH, AHA)<br />

were evaluated in tumor samples in vitro and in<br />

vivo. Human and mouse specific primer probe<br />

sets were designed. Those species specific<br />

primer probe sets offer the possibility to<br />

accurately detect levels of the human tumor<br />

target RNA in xenograft tumor models in the<br />

presence of mouse tissue. In our studies ACTB<br />

has emerged to be a more reliable housekeeping<br />

gene than GAPDH due to less variation in mRNA<br />

levels. Knockdown of therapeutic genes (such as<br />

KSP), which are cell cycle dependant, showed<br />

higher variations in analysis of cell culture and in<br />

vivo tumor samples. In addition silencing target<br />

genes which affect tumor survival complicate the<br />

evaluation due to cell death of transfected cells.<br />

Thus, targeting of non-essential gene targets<br />

seems to be more feasible <strong>for</strong> measuring<br />

silencing efficiency.<br />

Session: Vector Development<br />

P 8<br />

Magnetic Microbubbles: Magnetically<br />

Targeted and Ultrasound-Triggered Vectors<br />

<strong>for</strong> <strong>Gene</strong> Delivery In Vitro<br />

Dialechti Vlaskou 1 , Olga Mykhaylyk 1 , Pallab<br />

Pradhan 1 , Christian Bergemann 2 , Alexander L.<br />

Klibanov 3 , Karin Hensel 4 , Georg Schmitz 4 ,<br />

Christian Plank 1<br />

1 Klinikum rechts der Isar, TU München, Institute<br />

für Experimentelle Onkologie, <strong>Munich</strong>, <strong>German</strong>y;<br />

2 Chemicell GmbH, Berlin, Berlin, <strong>German</strong>y;<br />

3 Department of Biomedical Engineering, Division<br />

of Cardiovascular Medicine, University of<br />

Virginia, Charlottesville, USA; 4 Institute of<br />

Medical Engineering, Dept. <strong>for</strong> Electrical<br />

Engineering and In<strong>for</strong>mation Sciences, Ruhr-<br />

University Bochum, Bochum, <strong>German</strong>y<br />

In recent years microbubbles technology has<br />

gained lot of interest in the field of gene and drug<br />

delivery. Based on the “Magnetofection” concept<br />

[1], besides the development of magnetic<br />

acoustically active lipospheres [2, 3], we have<br />

been able to prepare lipid monolayer shelled<br />

microbubbles loaded with highly positively<br />

charged naked magnetic nanoparticles<br />

(composed of iron oxide) through electrostatic<br />

and matrix affinity interactions. These magnetic<br />

microbubbles show strong ultrasound contrast.<br />

Treatment of cancer cells with these<br />

microbubbles using ultrasound exhibited strong<br />

dose-dependent cytotoxic effects, although<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong><br />

| 44<br />

ultrasound alone, lipid microbubbles alone,<br />

magnetic nanoparticles or magnetic<br />

microbubbles alone at the corresponding<br />

concentrations did not affect the cell viability. On<br />

the other hand, when these magnetic<br />

microbubbles were mixed with plasmid DNA<br />

encoding a reporter gene, we achieved gene<br />

delivery to cultured adherent cells only when<br />

ultrasound was applied. <strong>Gene</strong> transfer efficiency<br />

was strongly dependent on the application of a<br />

gradient magnetic field to sediment the<br />

microbubbles on the target cell membranes.<br />

From the preliminary experiments we conclude<br />

that magnetic microbubbles could be used as<br />

magnetically targeted diagnostic agents <strong>for</strong> realtime<br />

ultrasound as well as magnetic resonance<br />

imaging. At the same time, such magnetic<br />

microbubbles may be useful <strong>for</strong> therapeutic<br />

purposes such as in cancer therapy, vascular<br />

thrombolysis and gene therapy. However, further<br />

improvements are required to control their<br />

cytotoxicity. References [1] C. Plank, U.<br />

Schillinger, F. Scherer, C. Bergemann, J-S.<br />

Remy, F. Krötz, M. Anton, J. Lausier, J.<br />

Rosenecker. The magnetofection method: using<br />

magnetic <strong>for</strong>ce to enhance gene delivery. Biol.<br />

Chem. 2003; 384 (5): 737–747. [2] Plank C,<br />

Vlaskou D, Schillinger U, Mykhaylyk O, Brill T,<br />

Rudolph C, Huth S, Krötz F, Hirschberger J,<br />

Bergemann C. Localized nucleic acid delivery<br />

using magnetic nanoparticles. European Cells<br />

and Materials. 2005; 10(Suppl. 5):8. [3] Vlaskou<br />

D, Mykhaylyk O, Giunta R, Neshkova I, Hellwig<br />

N, Krötz F, Bergemann C, Plank C. Magnetic<br />

Microbubbles: New Carriers <strong>for</strong> Localized <strong>Gene</strong><br />

and Drug Delivery. Mol Ther. 2006; 13(Suppl.<br />

1):S290.<br />

Session: Vector Development<br />

P 9<br />

Sleeping Beauty-Mediated Transposition in<br />

Human Cells Is Enhanced in RNA Interference<br />

Knockdown Cells<br />

Christina Rauschhuber, Anja Ehrhardt<br />

Department of Virology, Max von Pettenkofer-<br />

Institute, LMU, <strong>Munich</strong>, <strong>German</strong>y<br />

Integrating nonviral vectors based on<br />

transposable elements and bacteriophage<br />

derived integrases are widely used <strong>for</strong> genetically<br />

engineering mammalian cells, in functional<br />

genomics and therapeutic gene transfer. For the<br />

Sleeping Beauty (SB) transposase system it was<br />

demonstrated that convergent transcription


45 |<br />

driven by the SB transposase inverted repeats<br />

(IRs) in eukaryotic cells occurs. This may lead to<br />

<strong>for</strong>mation of double-stranded RNAs potentially<br />

presenting targets <strong>for</strong> the RNA interference<br />

(RNAi) machinery and finally to silencing of the<br />

transgene. There<strong>for</strong>e, in the present study we<br />

generated and characterized RNAi knockdown<br />

mammalian cell lines and analyzed transposition<br />

in these cells. RNAi knockdown HEK293 cell lines<br />

were generated by stably introducing the RNAi<br />

suppressor protein P19 from the tomato bushy<br />

stunt virus which binds and inhibits 21 nucleotide<br />

long double stranded siRNAs. In order to<br />

characterize our RNAi knockdown cell lines we<br />

used the marker gene HoxB8 which, in<br />

differentiated cells, is suppressed by the miRNA<br />

miR196a. Using a quantitative Real-Time PCR<br />

based approach we detected up to 10-fold higher<br />

HoxB8 mRNA levels in the RNAi knockdown cell<br />

lines compared to the parental cell line clearly<br />

demonstrating that P19 is a sufficient inhibitor of<br />

the RNAi pathway. After per<strong>for</strong>ming colony<br />

<strong>for</strong>ming assays measuring SB transposition<br />

events we could show that transposition was<br />

increased up to 4.5-fold in our RNAi knockdown<br />

cell lines compared to parental HEK293 cells,<br />

providing clear evidence <strong>for</strong> the first time that SB<br />

transposition is regulated by the RNAi machinery<br />

in mammalian cells. In contrast another<br />

transposable element, the Frog Prince<br />

transposon that displays only modest<br />

transcriptional activity, a 1.2-fold increase in<br />

transposition events was observed. To<br />

investigate integration of non-viral vectors in a<br />

more global manner, we also quantified<br />

bacteriophage-derived integrase PhiC31<br />

mediated integration events, and found that<br />

integration was enhanced up to 3-fold. In<br />

conclusion, our results indicate that transposition<br />

in mammalian cells and potentially also the<br />

PhiC31 integrase system are regulated by the<br />

endogenous RNAi machinery.<br />

Session: Vector Development<br />

P 10<br />

Micro RNA Knockdown Significantly<br />

Enhances Adenovirus Replication and Vector<br />

Production<br />

Christina Rauschhuber, Martin Hausl, Anja<br />

Ehrhardt<br />

Department of Virology, Max von Pettenkofer-<br />

Institute, LMU, <strong>Munich</strong>, <strong>German</strong>y<br />

The use of microRNAs (miRNAs) fundamentally<br />

improved the generation of viral vectors <strong>for</strong> gene<br />

therapy. For instance the introduction of miRNA<br />

target sequences into lentiviral vectors excluded<br />

expression of the respective protein from an<br />

unwanted tissue (Brown et al., 2006). However,<br />

the effect of the RNA interference (RNAi)<br />

pathway itself on gene therapy vectors remains<br />

an obstacle. In this study we used our recently<br />

established RNAi knockdown cell line B6<br />

(Rauschhuber and Ehrhardt, submitted) to<br />

analyze the influence of the RNAi pathway on<br />

adenoviral vectors. We first investigated<br />

replication of wildtype adenovirus 5 (wtAd5) and<br />

first generation adenovirus (FgAd) on a genome<br />

level. An up to 10-fold increase in viral genome<br />

copy numbers 24 hours and 48 hours post<br />

infection could demonstrate that there is<br />

significantly enhanced viral DNA replication<br />

under RNAi knockdown conditions. To analyze<br />

whether we can also achieve higher viral titers in<br />

the B6 cell line, we infected with a commonly<br />

used FgAd expressing firefly luciferase<br />

(FgAdluc). After re-infection of HEK293 cells we<br />

measured up to 5-fold increased luciferase<br />

activity indicating a higher titer of FgAdluc<br />

derived from B6 cells. To reach even stronger<br />

effects on virus replication we hypothesized that<br />

the amount of the RNAi suppressor protein P19<br />

may be the limiting factor. Thus, we applied our<br />

novel adenovirus cloning technology based on<br />

bacterial artificial chromosomes and generated a<br />

recombinant adenovirus expressing p19 under<br />

the control of the fiber promoter <strong>for</strong> which the<br />

amount of P19 directly correlates with adenovirus<br />

genome replication. We detected up to 10-fold<br />

increased genome copy numbers compared to<br />

wtAd5 and the corresponding control virus.<br />

Following our new method of adenovirus type<br />

switch we also generated a helper-virus<br />

expressing p19 <strong>for</strong> helper-dependent adenovirus<br />

(HC-AdV) production. In contrast to a<br />

conventionally used helper-virus we detected up<br />

to 12-fold increased HC-AdV titers. In conclusion,<br />

we provide new insights into regulation of<br />

adenoviral vectors by RNAi and our RNAi<br />

knockdown cell line B6 could be used as a novel<br />

producer cell line <strong>for</strong> adenoviral vectors.<br />

Session: Vector Development<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong>


P 11<br />

A Transient Cell Cycle Arrest Enhances AAV<br />

and ZFN-Mediated <strong>Gene</strong> Targeting<br />

Shamim H Rahman, Katharina Gellhaus, Kaweh<br />

Montazami-Astaneh, Regine Heilbronn, Toni<br />

Cathomen<br />

Department of Experimental Hematology,<br />

Hannover Medical School, Hannover, <strong>German</strong>y<br />

Vectors based on adeno-associated virus (AAV)<br />

are efficient tools to modify complex genomes by<br />

gene targeting. <strong>Gene</strong> targeting is based on the<br />

homologous recombination (HR) pathway, which<br />

is mainly available during the G2 phase of the<br />

cell cycle. We have shown that insertion of a<br />

DNA double strand break (DSB) in the target<br />

locus stimulates gene targeting up to 1000-fold,<br />

most likely by activating the HR repair pathway.<br />

To assess the effect of the cell cycle on AAV and<br />

DSB-mediated gene targeting, we established<br />

three human cell lines (HeLa, HT-1080, U-2 OS)<br />

that carry a copy of a mutated eGFP gene<br />

flanked by recognition sites <strong>for</strong> the meganuclease<br />

I-SceI and zinc-finger nucleases (ZFNs). After<br />

treatment with different cytostatic drugs (including<br />

hydroxyurea, vinblastine, indirubin), cells were<br />

transduced with AAV vectors that encode the<br />

nuclease and an HR donor with the purpose of<br />

rescuing eGFP expression by gene targeting.<br />

The cell cycle profile, the extent of cytotoxicity<br />

and the frequency of gene targeting were<br />

assessed by flow cytometry. We show that a<br />

transient cell cycle arrest be<strong>for</strong>e the creation of a<br />

DSB in the target locus increased AAV-mediated<br />

gene targeting up to 6-fold. Although this effect<br />

could be partially attributed to an increase in AAV<br />

transduction, a significant increase in gene<br />

targeting was also observed by plasmid-based<br />

eGFP rescue, implying that a drug-induced<br />

transient cell cycle arrest is a general means to<br />

augment DSB-mediated gene targeting.<br />

Depending on the cell line and the AAV vector<br />

dose, we achieved targeted gene editing in up to<br />

12% of transduced cells. Interestingly, the ratio<br />

between AAV vectors used <strong>for</strong> gene targeting vs.<br />

illegitimate integration was strongly cell type<br />

dependent and ranged between 30:1 and 1:9.<br />

Also, drug-induced transient cell cycle arrest had<br />

only minor effects on this ratio. In conclusion,<br />

cytostatic drugs can be used to enhance DSBmediated<br />

gene targeting in different cell lines.<br />

The combined beneficial effects of site-specific<br />

DSBs and transient cell cycle arrest allowed us to<br />

lower the AAV vector dose – and hence<br />

illegitimate integration of the vectors – without<br />

compromising on the gene targeting efficacy.<br />

Session: Vector Development<br />

P 12<br />

Novel Heterospecific FRTs Enable <strong>Gene</strong><br />

Targeting of Several Loci by Flp-Multiplex-<br />

RMCE<br />

Sören Turan, Juergen Bode, Johannes Kuehle,<br />

Axel Schambach<br />

Hannover Medical School (MHH), Experimental<br />

Hematology, Hannover, <strong>German</strong>y<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong><br />

| 46<br />

Since 48 bp Flp-recombinase sites (FRTs)<br />

represent unique targets in all eukaryotic<br />

genomes recombinase-mediated cassette<br />

exchange (Flp-RMCE) is not hampered by the<br />

occurence of genomic pseudosites. We explored<br />

the feasibility of exchanging two distinct<br />

genomically anchored cassettes, each flanked by<br />

an unique pair of heterospecific FRT sites. For<br />

successful RMCE novel constructed mutant<br />

FRTs had to meet two major prerequisites (a)<br />

given self-recognition potential comparable to a<br />

pair of FRT wildtype sites (b) negligible<br />

crossinteraction among heterospecific sites. We<br />

applied a two-step strategy to characterize<br />

various newly created FRT spacer mutants <strong>for</strong><br />

these properties. As a result of our screening<br />

steps we identified combinations F3/F and<br />

F13/F14 of sites, which were successfully applied<br />

to simultaneous Flp-mediated genomic targeting<br />

(“multiplexing”) reactions. The emergence of<br />

novel heterospecific FRTs can play a key role <strong>for</strong><br />

specified gene targeting approaches, <strong>for</strong> instance<br />

the sequential elaboration of larger gene domain<br />

subunits by RMCE. Reference: Turan S, Kuehle<br />

J, Schambach A, Baum, C and Bode, J (2010)<br />

RMCE-multiplexing: versatile extensions of the<br />

Flp-Recombinase-Mediated Cassette-Exchange<br />

technology. J. Mol. Biol. In press.<br />

Session: Vector Development


47 |<br />

P 13<br />

Chemical Modification of the Adenovirus<br />

Capsid <strong>for</strong> Enhanced Viral Uptake into Target<br />

Cells<br />

Alexandra Vetter 1 , Kulpreet Singh Virdi 2 , Sigrid<br />

Espenlaub 3 , Wolfgang Rödl 1 , Ernst Wagner 1,4 ,<br />

Per S. Holm 5 , Florian Kreppel 3 , Christina Scheu 2 ,<br />

Christine Spitzweg 6 , Manfred Ogris 1,4<br />

1 Pharmaceutical Biotechnology, Department of<br />

Pharmacy, Ludwig-Maximilians-University,<br />

<strong>Munich</strong>, <strong>German</strong>y; 2 Physical Chemistry,<br />

Department of Chemistry, Ludwig-Maximilians-<br />

University, <strong>Munich</strong>, <strong>German</strong>y; 3 Division of <strong>Gene</strong><br />

<strong>Therapy</strong>, University of Ulm, Ulm, <strong>German</strong>y;<br />

4 Center <strong>for</strong> Nanoscience (CeNS), Ludwig-<br />

Maximilians-University, <strong>Munich</strong>, <strong>German</strong>y;<br />

5 Institute of Experimental Oncology and <strong>Therapy</strong><br />

Research, Technische Universität München,<br />

<strong>Munich</strong>, <strong>German</strong>y; 6 Department of Internal<br />

Medicine II, Ludwig-Maximilians-University,<br />

<strong>Munich</strong>, <strong>German</strong>y<br />

Recombinant adenovirus type 5 (Ad5) is a highly<br />

efficient vector <strong>for</strong> cancer gene therapy<br />

approaches, but due to its natural tropism <strong>for</strong> the<br />

coxsackie- and adenovirus receptor (CAR) it<br />

lacks specificity <strong>for</strong> tumor cells and can induce<br />

severe inflammatory responses. Although<br />

present on a broad range of tissues, CAR is often<br />

down-regulated in several tumor types.<br />

Combining the high transfection efficiency of Ad<br />

with synthetic polymers can potentially allow detargeting<br />

from CAR positive tissues and retargeting<br />

to tumor cells. We have coated Ad5<br />

with cationic polymers like PAMAM dendrimer or<br />

linear polyethylenimine (LPEI) by electrostatic<br />

interaction with the negatively charged Ad capsid.<br />

Size and surface charge of polymer coated Ad5<br />

were measured by laser light scattering, particle<br />

shape by transmission electron microscopy<br />

(TEM). Uptake and intracellular transport was<br />

studied by fluorescence microscopy and flow<br />

cytometry using fluorescently labeled Ad<br />

(Alexa488) and polymer (Cy5). Transgene<br />

expression studies in vitro were carried out using<br />

either luciferase gene or the sodium iodide<br />

symporter gene (NIS), were the latter leads to<br />

cellular uptake of radioactive iodine. With TEM it<br />

was possible to show that the virus is<br />

encapsulated by the polymer <strong>for</strong>ming a shell<br />

around the virus with a rough surface structure.<br />

The virus surface charge changed from a net<br />

negative value of naked virus −17mV) ( to + 4 mV<br />

when coated with PAMAM dendrimer, and the<br />

average particle size increased with increasing<br />

amounts of polycation added. Recharging the<br />

virus led to pronounced cellular binding and<br />

uptake of virus. On high CAR expressing cells<br />

transfection efficiency was reduced after polymer<br />

coating, whereas on cell lines with low CAR<br />

expression increased transgene expression was<br />

measured. In addition, we covalently attached a<br />

peptide ligand to the polymer binding to the<br />

epidermal growth factor receptor (EGFR). This<br />

further promoted uptake in EGFR positive tumor<br />

cells. The obtained data suggests that a<br />

combination of viral and nonviral gene vector<br />

systems could compensate the limitations each<br />

approach has on its own and lead to superior<br />

success in cancer gene therapy.<br />

Session: Vector Development<br />

P 14<br />

<strong>Gene</strong>ration of microRNA-Sensitive VSV<br />

Vectors <strong>for</strong> Safer Treatment of Hepatocellular<br />

Carcinoma and Hepatic Metastasis of<br />

Colorectal Cancer<br />

Sibylle Apfel 1 , Jennifer Altomonte 1 , Michael<br />

Stiess 2 , Frank Bradke 2 , Oliver Ebert 1<br />

1 II. Medizinische Klinik und Poliklinik, Klinikum<br />

rechts der Isar, TU München, <strong>Munich</strong>, <strong>German</strong>y;<br />

2 Independent Junior Research Group Axonal<br />

Growth and Regeneration, Max Planck Institute<br />

of Neurobiology, Martinsried, <strong>German</strong>y<br />

Vesicular stomatits virus (VSV) is an oncolytic<br />

virus, which shows promise as a therapeutic<br />

agent <strong>for</strong> cancer, however, toxic collateral effects<br />

on the brain and the liver occur when it is<br />

administered at high doses. We hypothesized<br />

that by employing a novel strategy, termed<br />

microRNA-engineering, we could eliminate offtarget<br />

replication and improve the therapeutic<br />

index. This approach involves the incorporation<br />

of target sequences of a specific miRNA (miRTs)<br />

into the viral genome, resulting in degradation of<br />

viral gene transcripts in cells in which this miRNA<br />

is highly expressed, without interfering with virus<br />

replication in cells not expressing the relevant<br />

miRNA. We screened a panel of candidate<br />

miRNAs to select <strong>for</strong> those with high expression<br />

levels in the brain and/or liver and low levels in<br />

primary human HCC and CRC and cell lines. We<br />

then confirmed the miRNA-mediated regulation of<br />

the candidates in a reporter gene assay. Four<br />

tandem repeats of perfect complementary miRTs<br />

were cloned into the ′ 3 -UTR of a luciferase<br />

reporter gene, and these plasmids were<br />

transfected into a panel of normal and tumor<br />

cells. We then selected only those miRTs which<br />

resulted in a reduction of luciferase expression in<br />

primary hepatocytes and neurons but not in<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong>


tumor cells. Based on these preliminary<br />

experiments, we chose three miRTs which best<br />

fit our selection criteria, and created recombinant<br />

VSV vectors containing four tandem repeats of<br />

the miRTs in the 3′ -UTRs of crucial endogenous<br />

viral genes. After testing of these vectors in tumor<br />

and non-tumor cells in vitro, thorough toxicity and<br />

efficacy studies will be per<strong>for</strong>med in mouse<br />

models of HCC and CRC. We ultimately aim to<br />

engineer next-generation viruses with one or<br />

more viral gene regulated by multiple miRTs.<br />

Because saturation of endogenous miRNAs in<br />

the target tissues could result in “leaky” virus<br />

replication, we hypothesize that the incorporation<br />

of multiple miRTs could overcome this limitation<br />

and result in an even safer vector. Thus, this<br />

strategy has the potential to provide a basis <strong>for</strong><br />

clinical application of VSV vectors <strong>for</strong> the<br />

treatment of primary and metastatic liver disease,<br />

as well as other cancers, in the future.<br />

Session: Vector Development<br />

P 15<br />

Multifunctional Magnetic Liposomes <strong>for</strong><br />

Cancer <strong>Therapy</strong><br />

Pallab Pradhan 1 , Jyotsnendu Giri 2 , Andreas<br />

Steingoetter 3 , Christian Koch 1 , Olga Mykhaylyk 1 ,<br />

Rinti Banerjee 4 , Dhirendra Bahadur 4 , Christian<br />

Plank 1<br />

1Institut für Experimentelle Onkologie und<br />

Therapie<strong>for</strong>schung, Klinikum rechts der Isar,<br />

TUM, <strong>Munich</strong>, <strong>German</strong>y; 2 Dept. of Chemical<br />

Engineering, Cali<strong>for</strong>nia Institute of Technology,<br />

Pasadena, USA; 3 Dept. of Nuclear Medicine,<br />

Klinikum rechts der Isar, Technical University of<br />

<strong>Munich</strong>, <strong>Munich</strong>, <strong>German</strong>y; 4 Indian Institute of<br />

Technology, Bombay, Powai, Mumbai, India<br />

A multifunctional magnetic liposome <strong>for</strong>mulation,<br />

which is designed to combine features of<br />

biological (folate receptor mediated) and physical<br />

(magnetic field assisted) drug targeting <strong>for</strong> use in<br />

magnetic hyperthermia triggered drug release <strong>for</strong><br />

cancer therapy has been developed. The<br />

magnetic liposomes (MagFolDox) composed of<br />

DPPC/Cholesterol/DSPE-PEG2000/DSPE-<br />

PEG2000-Folate at 80:20:4.5:0.5 molar ratio<br />

released 52% doxorubicin at 43°C after 60<br />

minutes incubation in 50% fetal bovine serum.<br />

The MagFolDox, when physically targeted to<br />

tumor cells in culture by a permanent magnetic<br />

field yielded a several fold increase in cellular<br />

uptake of doxorubicin as compared to Caelyx ® (a<br />

stealth liposomal <strong>for</strong>mulation of doxorubicin),<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong><br />

| 48<br />

nonmagnetic folate-targeted liposomes and free<br />

doxorubicin in folate receptor expressing tumor<br />

cell lines. Consequently, the MagFolDox<br />

liposomes enabled improved tumor cell killing.<br />

Moreover, MagFolDox and magnetic<br />

hyperthermia (at 42.5-43.5°C) showed synergistic<br />

cytotoxic effect in KB cells. Also, the distribution<br />

of magnetic liposomes in vivo can be monitored<br />

by MRI due to their contrast enhancement<br />

properties. In vivo real time MRI monitoring in<br />

mice confirmed significantly higher blood<br />

circulation time <strong>for</strong> MagFolDox compared with<br />

free magnetic particles. This also confirmed that<br />

the biodistribution of the MagFolDox can be<br />

monitored in vivo by MRI. Thus, the<br />

multifunctionality of the magnetic liposomes has<br />

been successfully demonstrated.<br />

Session: Vector Development<br />

P 16<br />

Melanoma-Killing Adenovirus Derived by<br />

Direct Evolution<br />

Inés Fernández Ulibarri, Dominik Dorer, Dirk M<br />

Nettelbeck<br />

DKFZ, Heidelberg, <strong>German</strong>y<br />

Viral oncolysis is a strategy <strong>for</strong> targeted treatment<br />

of cancer by tumor-restricted virus infection.<br />

Human Adenoviruses (Ad) are promising<br />

oncolytic agents, but showed insufficient<br />

therapeutic efficiency in clinical trials. There<strong>for</strong>e,<br />

the key challenge <strong>for</strong> virotherapy research is to<br />

develop Ad with enhanced viral lysis. In this<br />

regard, I focus on developing effective oncolytic<br />

Ad <strong>for</strong> malignant melanoma by directed evolution<br />

in order to derive adenoviruses with increased<br />

lytic activity. First, we investigated the efficiency<br />

of a panel of Ad serotypes from species B, C,<br />

and D in melanoma cells in comparison to the<br />

most potent virus <strong>for</strong> this type of cancer, Ad5/3<br />

(Ad5 with the cell-binding domain of Ad3). Our<br />

results indicate that none of those serotypes are<br />

more potent than Ad5/3 in melanoma cells.<br />

However, the oncolysis of the melanoma cells is<br />

still strongly attenuated in comparison with<br />

bronchial epithelial cells, which are the natural<br />

host cells. There<strong>for</strong>e, although Ad5/3 is the most<br />

potent Ad in melanoma an improvement is still<br />

needed. To enhance the lytic activity of Ad5/3,<br />

the viruses were treated with nitrous acid sodium<br />

(NaNO2) which induces random mutations in the<br />

virus genome. Subsequently, the originated<br />

mutations were fixed and the individual mutants<br />

were amplified in melanoma cells. Finally, with


49 |<br />

the objective of harvesting the most rapid spread<br />

viruses, the bioselection rounds of the<br />

mutagenized Ad5/3 were per<strong>for</strong>med by<br />

passaging the supernatant (at early stage of<br />

infection) repeatedly on the melanoma cells. After<br />

the 20th round of bioselection, our results show<br />

that mutagenized Ad5/3 were 10-fold more potent<br />

in melanoma cells in comparison with the<br />

parenteral virus. This result suggests that the<br />

most potent virus generated after NaNO2<br />

treatment replicate, lyse the host and are<br />

released into the supernatant be<strong>for</strong>e the majority<br />

of virus population. Currently, I am characterizing<br />

the mutants to restrict their replication in<br />

melanoma cells by a tumor-specific promoter. In<br />

summary, our results indicated that directed<br />

evolution is a strategy which allows the<br />

enrichment of viruses' mutants with enhanced<br />

replication and spread <strong>for</strong> malignant melanoma<br />

cells.<br />

Session: Vector Development<br />

P 17<br />

Versatile and Efficient Genome Editing in<br />

Human Cells by Combining Zinc-Finger<br />

Nucleases with AAV Vectors<br />

Eva M Händel 1 , Katharina Gellhaus 2 , Kafaitullah<br />

Khan 1 , Christien Bednarski 1 , Tatjana I Cornu 2 ,<br />

Felix Müller-Lerch 2 , Regine Heilbronn 2 , Toni<br />

Cathomen 1<br />

1 Department of Experimental Hematology,<br />

Hannover Medical School, Hannover, <strong>German</strong>y;<br />

2 Institute of Virology (CBF), Charité Medical<br />

School, Berlin, <strong>German</strong>y<br />

Zinc-finger nucleases (ZFNs) are an innovative<br />

tool <strong>for</strong> targeted genome engineering. They<br />

consist of a catalytic nuclease domain and a<br />

designed DNA binding domain and can be<br />

employed <strong>for</strong> either targeted deletion or gene<br />

disruption by harnessing the non-homologous<br />

end joining DNA repair pathway or, in the<br />

presence of a properly designed donor DNA,<br />

gene targeting by activating homology directed<br />

repair (HDR). Here, we demonstrate the<br />

versatility of ZFN-mediated genome editing when<br />

combined with vectors derived from adenoassociated<br />

virus type 2 (AAV2). For proof-ofconcept<br />

studies, we generated four different<br />

human reporter cell lines. For assessing gene<br />

disruption and targeted deletion of an integrated<br />

provirus, a lentiviral EGFP vector containing ZFN<br />

target sites in the LTRs was used to generate an<br />

EGFP-positive cell line. To evaluate therapeutic<br />

gene correction, cells harboring a mutated EGFP<br />

or Luciferase reporter were produced. The<br />

frequencies of targeted genome editing and the<br />

extent of cytotoxicity were determined by flow<br />

cytometry and/or luminometry at different time<br />

points after transduction. Disruption of the EGFP<br />

locus was achieved in 18% of cells infected with<br />

vectors encoding EGFP-specific ZFNs. Upon<br />

transduction with AAV vectors coding <strong>for</strong> LTRspecific<br />

ZFNs, the lentiviral provirus was excised<br />

in 10% of cells. Furthermore, after co-infection of<br />

cells with vectors that encode locus-specific<br />

ZFNs or serve as donors <strong>for</strong> gene targeting,<br />

AAV-mediated correction of the mutated marker<br />

genes restored reporter activity in up to 6% of<br />

cells. The frequency of genome editing was<br />

depending on the vector dose and the specificity<br />

of the ZFNs. Significant signs of cell death were<br />

not observed, suggesting minor toxicity<br />

associated with these approaches. In conclusion,<br />

the combined ZFN–AAV vector technology is a<br />

flexible tool to efficiently modify the human<br />

genome ad libitum. Because AAV vectors can<br />

transduce multiple human tissues, including<br />

induced pluripotent stem cells and mesenchymal<br />

stem cells, the ex vivo delivery of ZFNs and HDR<br />

donors via AAV vectors will be of great interest<br />

<strong>for</strong> the treatment of several disorders in<br />

regenerative medicine.<br />

Session: Vector Development<br />

P 18<br />

Progress in Minicircle Manufacturing and<br />

Per<strong>for</strong>mance Testing<br />

Marco Schmeer, Markus Blaesen, Ruth Baier,<br />

Martin Schleef<br />

PlasmidFactory GmbH & Co. KG, Bielefeld,<br />

<strong>German</strong>y<br />

Plasmid DNA is commonly used in vaccination,<br />

gene- or cell therapy but also as a basic<br />

substance in viral vector production. The<br />

dissemination of antibiotic resistance genes, as<br />

well as the uncontrolled expression of backbone<br />

sequences present in such plasmids may have<br />

profound detrimental effects. Additionally,<br />

unmethylated CpG motifs have been shown to<br />

contribute to silencing of episomal transgene<br />

expression. There<strong>for</strong>e, an important goal in<br />

vector development is to produce supercoiled<br />

DNA lacking bacterial backbone sequences:<br />

Minicircle DNA. PlasmidFactory's minicircle<br />

production technology facilitates the large-scale<br />

production of highly pure minicircle DNA <strong>for</strong><br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong>


applications in gene therapy and vaccination as<br />

well as, e.g., virus production. The production<br />

technology is based on two processes: (1) An<br />

inducible, sequence specific, and very efficient in<br />

vivo recombination process; (2) A<br />

chromatographic purification technology <strong>for</strong> the<br />

isolation of the minicircle DNA. The resulting<br />

minicircle DNA only consists of the gene of<br />

interest and a tiny residual sequence stretch<br />

including one of the two recombination sites. The<br />

chromatographic purification results in an<br />

exceptional purity, proven by various QC tests,<br />

which is extremely important since even small<br />

amounts of contaminants can produce dramatic<br />

experimental artefacts. For first efficacy studies,<br />

reporter genes <strong>for</strong> different types of analyses<br />

within various tissues, cells, animals and <strong>for</strong><br />

testing the mode of administration (e.g., electro<br />

gene transfer, sonoporation, lipofection,<br />

magnetofection, etc.) have been used. Also<br />

biodistribution studies using these constructs<br />

have been per<strong>for</strong>med. Additionally, minicircle<br />

constructs <strong>for</strong> vaccines, virus production, etc., are<br />

currently investigated. The results demonstrate a<br />

significant increase of gene expression of<br />

minicircle DNA in comparison to a standard<br />

plasmid. As a result of the work presented here,<br />

PlasmidFactory has launched several<br />

commercially available minicircle DNA products.<br />

Session: Vector Development<br />

P 19<br />

Minicircle – An Overview<br />

Martin Schleef, Marco Schmeer, Ruth Baier,<br />

Markus Blaesen<br />

PlasmidFactory GmbH & Co. KG, Bielefeld,<br />

<strong>German</strong>y<br />

For future gene therapy and even genetic<br />

vaccination approaches it is crucial to develop<br />

safe and highly efficient vector systems to be<br />

transferred into the target cells. Currently, viral<br />

and non-viral vectors are used, both having their<br />

advantages and limitations. The dissemination of<br />

antibiotic resistance genes, as well as the<br />

uncontrolled expression of backbone sequences<br />

present in plasmid DNA may have profound<br />

detrimental effects. Additionally, unmethylated<br />

CpG motifs have been shown to contribute to<br />

silencing of episomal transgene expression.<br />

Hence, it seems obvious that the removal of<br />

bacterial backbone DNA can greatly improve the<br />

safety and efficiency of plasmid DNA used in<br />

gene therapy and vaccination. Here, we give an<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong><br />

| 50<br />

overview on earlier and recent approaches <strong>for</strong><br />

the production, purification, and application of<br />

such minimal constructs, devoid of bacterial<br />

backbone sequences. Different approaches have<br />

been described so far, from plasmids where the<br />

antibiotics resistance gene has been replaced by<br />

another marker to minicircle DNA consisting only<br />

of the gene of interest and a tiny residual<br />

sequence stretch from the recombination.<br />

Minicircles can be produced by in vivo sitespecific<br />

recombination of a so-called parental<br />

plasmid resulting in a miniplasmid and the<br />

minicircle. This recombination can be achieved<br />

using different enzymes which need specific<br />

target sequences as recombination sites. The<br />

most important difference between these<br />

approaches is the efficiency of the recombination<br />

step as well as the purification procedure used in<br />

order to remove the miniplasmid (with the<br />

bacterial sequence motifs) and residual amounts<br />

of the parental plasmid if necessary. In addition<br />

to their improved safety profile, minicircles have<br />

been shown to greatly increase the efficiency of<br />

transgene expression in various in vitro and in<br />

vivo studies. Scale-up of such technology was<br />

recently possible in fermentation and results in an<br />

extremely pure DNA product.<br />

Session: Vector Development<br />

P 20<br />

<strong>Gene</strong>tic Manipulation of High-Capacity<br />

Adenoviral Vectors Utilizing Bacterial<br />

Artificial Chromosomes and Rapid Small-<br />

Scale Vector Preparations<br />

Martin Hausl, Christina Rauschhuber, Richard<br />

Voigtlander, Zsolt Ruzsics, Anja Ehrhardt<br />

Department of Virology, Max von Pettenkofer-<br />

Institute, LMU, <strong>Munich</strong>, <strong>German</strong>y<br />

First-generation adenoviral vectors (FG-AdVs)<br />

are easy to produce and there<strong>for</strong>e representing<br />

the best option to analyse capsid-modifications or<br />

new expression systems. On the other hand<br />

high-capacity adenoviral vectors (HC-AdVs)<br />

lacking all viral coding sequences are the most<br />

attractive option <strong>for</strong> therapeutic approaches.<br />

There<strong>for</strong>e, there is a great interest in developing<br />

a method <strong>for</strong> adoption of evaluated vectormodifications<br />

or expression systems in FG-AdVs<br />

towards HC-AdVs including fast generation of<br />

small scales of purified HC-AdV <strong>for</strong> initial<br />

experiments and subsequent large-scale<br />

amplification. Utilizing a novel plat<strong>for</strong>m based on<br />

homologous recombination of bacterial artificial


51 |<br />

chromosomes (BACs) we generated a BAC from<br />

a plasmid containing an HC-AdV genome by<br />

replacing the plasmid-backbone with the BACbackbone<br />

in a single cloning step. After this<br />

backbone-exchange we incorporated an eGFPexpression<br />

cassette, generated the respective<br />

HC-AdV and showed eGFP-expression. To adopt<br />

FG-AdVs to the BAC-plat<strong>for</strong>m we demonstrated<br />

that BACs with FG-AdV genomes can be<br />

constructed either by homologous recombination<br />

with adenoviral DNA isolated from purified virions<br />

or by backbone-exchange. Furthermore, we<br />

showed that our BAC technology can be used to<br />

switch from FG-AdVs to BACs with a helper virus<br />

(HV) genome <strong>for</strong> HC-AdV amplification, wild-type<br />

adenovirus or oncolytic adenovirus in a single<br />

cloning step. To highlight the impact of our BAC<br />

plat<strong>for</strong>m on adenovirus vectorology we<br />

exemplified adenovirus vector type switches by<br />

generating a HV with the established fiber mutant<br />

fib5/35 as well as a HV with an eGFP/Fluc<br />

expression cassette in the E3 region and a new<br />

hexon mutant with precisely exchanged<br />

hypervariable regions from human adenovirus<br />

serotype 12. All viruses were reconstituted and<br />

amplified. Notably, we observed that HC-AdVs<br />

can be first amplified in small amounts (4-6 × 20<br />

ml) and purified by column (Vivapure<br />

AdenoPACK 20, Sartorius). Yielded HC-AdV was<br />

used <strong>for</strong> initial proof-of-principle experiments and<br />

to optimize large-scale amplification making this<br />

technology amenable also <strong>for</strong> researchers<br />

without experiences in HC-AdV production.<br />

Session: Vector Development<br />

P 21<br />

A Novel Adenoviral Hybrid-Vector System<br />

Carrying a Plasmid Replicon <strong>for</strong> the Safe and<br />

Efficient <strong>Gene</strong> <strong>Therapy</strong> of Human Diseases<br />

Richard Voigtlander, Rudolf Haase, Martin Hausl,<br />

Armin Baiker, Anja Ehrhardt<br />

Department of Virology, Max von Pettenkofer-<br />

Institute, LMU, <strong>Munich</strong>, <strong>German</strong>y<br />

In dividing cells the two primary aims a gene<br />

therapy approach should accomplish are defined<br />

as the nuclear distribution and retention of<br />

therapeutic DNA. Because conventional<br />

monosystems fail to fulfil both tasks with equal<br />

efficiency, hybrid-vector systems are most<br />

promising in facing the challenges of modern<br />

molecular medicine. Our hybrid-vector system<br />

HDAdV-pEPito synergizes the helper-dependent<br />

adenoviral vector technology (HDAdV) with the<br />

plasmid pEPito containing the therapeutic gene<br />

and special DNA sequence<br />

SMAR(Scaffold/Matrix Attachment Region) <strong>for</strong><br />

episomal retention and replication. Thus, our<br />

technique provides a powerful tool <strong>for</strong> stable<br />

maintenance of the transgene reducing the risk of<br />

insertional mutagenesis. Adenoviral hybridvectors<br />

HDAdV-pEPito and the respective SMAR<br />

deleted control (HDAdV-pEPito-ΔSMAR) were<br />

generated with a novel BAC-technology<br />

established in our laboratory (Hausl et al., in<br />

preparation). Both contain an eGFP-IRES-BSDcassette<br />

under control of promoter hCMV/EF1. In<br />

FACS analysis 83% of A549-cells infected with a<br />

MOI of 50 were positive <strong>for</strong> eGFP-expression.<br />

Since adenoviral vectors carry linear DNA,<br />

pEPito has to be released from their genomes<br />

with Flpe-recombinase to function as a plasmid.<br />

A specific PCR proved this recombination event<br />

<strong>for</strong> both constructs. The episomal status of<br />

recombined plasmids could be verified with<br />

bacterial rescue experiments at day 4 after<br />

coinfecting A549-cells with our constructs and<br />

Flpe-expressing virus HDAdV-mSB-Flpe. More<br />

bacterial colonies grew after trans<strong>for</strong>mation of<br />

genomic DNA containing HDAdV-pEPito with<br />

Flpe (n = 281) than of DNA containing HDAdVpEPito-ΔSMAR<br />

with Flpe (n = 25), indicating<br />

replication of excised plasmids. Ongoing<br />

experiments are the comparison of HDAdVpEPito<br />

and HDAdV-pEPito-ΔSMAR concerning<br />

their long-term persistence in mice and their in<br />

vitro establishment efficiency in a colony <strong>for</strong>ming<br />

assay (CFA). Previous CFAs per<strong>for</strong>med with the<br />

original plasmids inserted into our two viruses<br />

seemed to show a tendency towards SMARpositive<br />

pEPito (48 surviving colonies) compared<br />

to SMAR deleted pEPito-ΔSMAR (27 surviving<br />

colonies) after 4 weeks of blasticidin selection.<br />

Session: Vector Development<br />

P 22<br />

Transient Cold Shock Enhances Zinc-Finger<br />

Nuclease-Mediated <strong>Gene</strong> Disruption – Rule or<br />

Exception?<br />

Nadine Dannemann, Christien Bednarski, Anne-<br />

Kathrin Dreyer, Toni Cathomen<br />

Department of Experimental Hematology,<br />

Hannover Medical School, Hannover, <strong>German</strong>y<br />

Zinc-finger nucleases (ZFNs) are a promising tool<br />

<strong>for</strong> precise editing of the human genome and<br />

hence of great interest in the field of gene<br />

therapy. ZFNs are designer nucleases consisting<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong>


of the catalytic domain of the FokI endonuclease<br />

and an engineered DNA binding domain. Three<br />

major plat<strong>for</strong>ms are available to generate zincfinger<br />

based DNA-binding domains: modular<br />

assembly, context-specific selection (OPEN), and<br />

the proprietary Sangamo plat<strong>for</strong>m. Recently,<br />

Doyon et al. (Nat Methods 7, 459-60; 2010)<br />

reported that transient hypothermia generally and<br />

robustly increased the frequency of ZFN-induced<br />

gene disruption. Here, we aimed to work out the<br />

mechanism behind enhanced ZFN activity at<br />

30°C and to elucidate whether enhanced ZFN<br />

activity upon cold shock is a generally applicable<br />

phenomenon. To this end, we employed six<br />

different ZFN pairs – Sangamo-based ZFNs<br />

directed against the human AAVS1, CCR5 and<br />

IL2RG loci and OPEN-based ZFN that recognize<br />

the CFTR, EGFP and HOXB13 genes – and<br />

compared their activities at 30°C and 37°C in<br />

vitro and in human cell lines. In vitro cleavage<br />

assays revealed no difference in ZFN activity<br />

when comparing ZFNs of the different plat<strong>for</strong>ms<br />

at 30°C vs. 37°C, implying that the temperature<br />

does neither affect DNA binding nor the catalytic<br />

activity. On the other hand, incubation of the cells<br />

at 30°C induced an M phase arrest and<br />

assessment of the ZFN levels by immunoblotting<br />

revealed increased protein levels, suggesting that<br />

the cold shock either enhances ZFN expression<br />

or stabilizes the enzyme. However, the higher<br />

ZFN levels in transfected cells did not correlate<br />

with a significant increase in gene disruption at<br />

30°C at the analyzed genomic loci, neither in<br />

HeLa nor in 293T cells. Moreover, analysis of<br />

ZFN-mediated EGFP disruption by flow<br />

cytometry in a U2OS.EGFP cell line did not<br />

reveal augmented knockout efficiency <strong>for</strong> cells<br />

that underwent transient cold shock treatment. In<br />

summary, our data suggest that although mild<br />

hypothermic conditions let to a robust increase in<br />

ZFN protein levels, enhanced gene disruption<br />

activity of ZFNs at 30°C may not be a general<br />

phenomenon but rather dependent on the cell<br />

type and the genomic target locus.<br />

Session: Vector Development<br />

P 23<br />

Development of Scaffold/Matrix Attachment<br />

Region (S/MAR) Minicircles <strong>for</strong> Enhanced and<br />

Persistent Transgene Expression in the<br />

Mouse Liver<br />

Orestis Argyros 1 , Suet-Ping Wong 1 , Constantinos<br />

Fedonidis 1 , Oleg Tolmachov 1 , Simon<br />

Waddington 2 , Steven Howe 3 , Charles Coutelle 1 ,<br />

Richard Harbottle 1<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong><br />

| 52<br />

1 <strong>Gene</strong> <strong>Therapy</strong> Research Group, Section of<br />

Molecular and Medicine, National Heart and Lung<br />

Institute, Imperial College London, London, UK;<br />

2 Department of Haematology, Haemophilia<br />

Centre and Haemostasis Unit, Royal Free and<br />

University College Medical School, London, UK;<br />

3 Molecular Immunology Unit, Institute of Child<br />

Health, University College London, London, UK<br />

We previously developed a Scaffold Matrix<br />

Attachment Region (S/MAR) plasmid system <strong>for</strong><br />

in vivo application (1). We demonstrated about<br />

40% hepatocyte transfection and sustained in<br />

vivo luciferase transgene expression in the<br />

mouse liver <strong>for</strong> at least six months following a<br />

single hydrodynamic administration.<br />

Subsequently, we observed sustained transgene<br />

expression <strong>for</strong> the lifetime of the animal, but this<br />

dropped appreciably over time. It has been<br />

shown that bacterial sequences present in<br />

plasmid DNA are responsible <strong>for</strong> vector toxicity<br />

and silencing of expression in vivo. We there<strong>for</strong>e<br />

hypothesised that by eliminating these<br />

extraneous bacterial components from our<br />

vectors we might improve their per<strong>for</strong>mance in<br />

vivo. We describe here the development of a<br />

minimally sized S/MAR vector using Cre/Lox<br />

recombinase (2), to generate a minicircle devoid<br />

of bacterial sequences and comprising only an<br />

expression cassette and an S/MAR moiety. Upon<br />

administration to mice by hydrodynamic delivery<br />

we found it to be maintained episomally<br />

(Southern blot analysis) and to provide sustained<br />

levels of luciferase expression <strong>for</strong> up to three<br />

months (end of experiment), as measured by a<br />

bioluminescent imager (XENOGEN). In addition,<br />

luciferase expression did not decline over time,<br />

as seen with our original S/MAR vector (1)<br />

containing the bacterial backbone. At the final<br />

time point, luciferase expression from the<br />

minicircle vector was approximately two orders of<br />

magnitude higher than both the control minicircle<br />

vector (no S/MAR) and the original S/MAR vector<br />

containing the bacterial backbone. In a further<br />

experiment, we per<strong>for</strong>med partial hepatectomy<br />

on S/MAR minicircle treated mice, and observed<br />

a rapid drop of expression, indicative of absence<br />

of plasmid replication. These promising results<br />

demonstrate the utility of minimally sized S/MAR<br />

vectors <strong>for</strong> persistent, atoxic gene expression in<br />

the liver. (1) Argyros et al., <strong>Gene</strong> Ther. (2008),<br />

15:1593-1605. (2) Bigger et al., J. Biol. Chem.<br />

(2001), Jun 22;276(25):23018-23027.<br />

Session: Vector Development


53 |<br />

P 24<br />

Systemic Administration of Nonviral Vectors<br />

to Neonatal Mice<br />

Suet-Ping Wong 1 , Argyros Orestis 1 , Steven<br />

Howe 2 , Charles Coutelle 1 , Richard Harbottle 1<br />

1 <strong>Gene</strong> <strong>Therapy</strong> Research Group, Section of<br />

Molecular and Medicine, National Heart and Lung<br />

Institute, Imperial College London, London, UK;<br />

2 Molecular Immunology Unit, Institute of Child<br />

Health, University College London, London, UK<br />

Nonviral vectors have not been extensively<br />

investigated in neonatal mice due to the poor<br />

efficiency of the delivery methods available.<br />

Understanding the effects of nonviral vectors<br />

during early development is vital to develop safe<br />

gene therapy treatments where irreversible<br />

pathological processes may be avoided by early<br />

gene reconstitution. Here we describe a simple<br />

and effective method <strong>for</strong> the systemic<br />

administration of nonviral vectors via the superior<br />

temporal vein of mouse pups at 1.5 days of age.<br />

We show that injection of polyethylenimine (PEI)<br />

complexed plasmid DNA (pDNA) intravenously<br />

results in effective transfection in the liver, lung,<br />

heart and spleen. We also investigate the specific<br />

targeting of transgene expression to the<br />

proliferating neonate liver using a liver-specific<br />

S/MAR plasmid, which has previously been<br />

shown to confer long-term expression in adult<br />

mouse liver. Using bioluminescent imaging, a<br />

gradual increase in transgene expression was<br />

observed peaking at days 11–12, be<strong>for</strong>e the<br />

reduction of expression to background levels by<br />

day 25, suggestive of vector copy number loss.<br />

We conclude that nonviral vectors can<br />

successfully be used <strong>for</strong> systemic delivery to<br />

neonatal mice but that further optimization of the<br />

S/MAR nonviral systems is required <strong>for</strong> longerterm<br />

maintenance of expression during neonatal<br />

growth and development.<br />

Session: Vector Development<br />

P 25<br />

Therapeutic Angiogenesis – Nonviral <strong>Gene</strong><br />

<strong>Therapy</strong> <strong>for</strong> Patients with Critical Limb<br />

Ischemia<br />

Alex Slobodianski 1 , Astrid Kathöfer 2 , Christoph<br />

Hartog 2 , Ziyang Zhang 1 , Jessica Frenz 2 , Peter<br />

Mailänder 2 , Hans-Günther Machens 1<br />

1 Klinikum rechts der Isar, TU München, <strong>Munich</strong>,<br />

<strong>German</strong>y; 2 University Hospital of Schleswig-<br />

Holstein, Kiel, <strong>German</strong>y<br />

Background: Therapeutic angiogenesis using<br />

angiogenic growth factors is expected to be a<br />

new treatment <strong>for</strong> patients with chronic limb<br />

ischemia. In our studies, we designed a GMPadapted<br />

process of therapeutic angiogenesis <strong>for</strong><br />

medicinal purposes using fibroblasts to<br />

temporarily produce bFGF and VEGF165 local in<br />

ischemic tissue. Two different animal models of<br />

ischemia were used <strong>for</strong> evaluations. We also<br />

considered some important aspects <strong>for</strong> preclinical<br />

evaluation of gene transfer medical products.<br />

Methods: The eukaryotic expression vectors<br />

harboring VEGF and bFGF cDNAs were<br />

transfected into rat primary fibroblasts mediated<br />

by Nucleofection. The transfection efficiency as<br />

well as expression of genes in the transfected<br />

fibroblasts were detected by means of flow<br />

cytometry, immunohistochemistry, enzyme-linked<br />

immunosorbent assay (ELISA) and real-time<br />

PCR. Effects on clinical outcome after<br />

transfected cells administration were determined<br />

in an ischemic flap model and in a hindlimb<br />

ischemia model in rat.<br />

Results: Transfection efficiency of about 60%<br />

and viability nearly 80% were observed by<br />

transfection of rat primary fibroblasts. Specific<br />

assays and real-time PCR af<strong>for</strong>d to determine<br />

any correlation between protein expression in<br />

vitro and local therapeutic effect of angiogenesis<br />

in target tissue. The established reproducible<br />

monitoring system allows to detect the<br />

distribution of gene therapeutics in the target<br />

tissue and to trace it in rats. A reduction in flap<br />

necrosis in flap model by nearly 40% was<br />

detected by planimetric measurements after two<br />

weeks if transfected cells were applied.<br />

Arteriogenesis and angiogenesis were improved<br />

in the rat hindlimb ischemia model two weeks<br />

after the transfected cells administration as it was<br />

confirmed by the computer analysis.<br />

Summary: Thus, in our studies we established a<br />

healing method Therapeutic Angiogenesis and<br />

demonstrated that transient expression of bFGF<br />

and VEGF165 induced therapeutic effects in<br />

iscemic tissue and improved tissue survival. Use<br />

of monitoring systems helps to recognize<br />

potential risks of gene therapeutics and to identify<br />

relations between the dosage and therapeutic<br />

effects in the target tissue <strong>for</strong> dose-finding<br />

studies.<br />

Session: On the Route to Clinical Application<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong>


P 26<br />

Lentiviral Vector-Induced Dendritic Cells<br />

Coexpressing GM-CSF, IFN-α and NS3 <strong>for</strong><br />

Immunotherapy of Hepatitis C<br />

Anusara Daenthanasanmak 1 , Rakesh Bakshi 1 ,<br />

Sandra Kuhs 1 , Bala Sai Sundarasetty 1 , Gustavo<br />

Salguero 1 , Helmut Diepolder 2 , Michael Ott 3 ,<br />

Heiner Wedemeyer 3 , Renata Stripecke 1<br />

1Department of Hematology, Hemostasis,<br />

Oncology and Stem Cell Transplantation,<br />

Hannover Medical School, Hannover, <strong>German</strong>y;<br />

2 Klinikum der Universitaet Muenchen, <strong>Munich</strong>,<br />

<strong>German</strong>y; 3 Department of Gastroenterology,<br />

Hannover Medical School, Hannover, <strong>German</strong>y<br />

Hepatitis C virus (HCV) is the main cause of<br />

chronic hepatitis and an effective therapeutic<br />

approach should effectively result in long-term<br />

immunologic eradication of the virus.<br />

Recombinant viruses and DNA vaccines are in<br />

the development <strong>for</strong> adjunct therapy along with<br />

pegylated interferon alpha (IFN-α) plus ribavirin.<br />

We propose to develop potent antigen-presenting<br />

cells (APCs), such as dendritic cells (DCs)<br />

capable to directly mimic the IFN-α therapy<br />

concurrently with optimized antigen presentation.<br />

We developed a novel technology <strong>for</strong> production<br />

of DC consisting of one overnight lentiviral vector<br />

(LV) transduction of GM-CSF and IL4 plus fulllength<br />

antigens into DC precursors that results<br />

into induction of “SMART-DCs” capable to selfdifferentiate<br />

directly into long-lived DCs in vivo,<br />

and thus bypass in vitro culturing. This concept<br />

has been tested and validated in preclinical<br />

human and mouse tumor models <strong>for</strong> potency<br />

(Koya et al., Molecular <strong>Therapy</strong>, 2007). Recently,<br />

we have demonstrated that monocyte-derived<br />

DCs transduced with lentiviral vectors expressing<br />

the HCV nonstructural cluster result into<br />

multiantigenic anti-HCV responses in vitro (Jirmo<br />

et al., Vaccine, 2010). There<strong>for</strong>e, we are currently<br />

validating multicistronic LVs that coexpress GM-<br />

CSF, IFN-α and a conserved and potent HCV<br />

antigen (NS3) in inducing a novel modality of<br />

engineered APCs, namely self-differentiated<br />

myeloid derived and lentivirus-induced DCs<br />

(SMYLE-DCs). Cotransduction of GM-CSF and<br />

IFN-α genes into CD14 + monocytes with a<br />

bicistronic LV resulted high levels of GM-CSF (1-<br />

2 ng/ml) and IFN-α (4 ng/ml) production, which<br />

were persistently detectable <strong>for</strong> 21 days. The<br />

SMYLE-DCs showed similar morphological and<br />

immunophenotypic features of DCs (MHCII + ,<br />

CD80 + CD86 + and CCR5+) and autonomous<br />

viability <strong>for</strong> 21 days of culture. The SMYLE-DCs<br />

coexpressing NS3 will be subsequently tested in<br />

in vitro assays and in humanized mouse models<br />

<strong>for</strong> the stimulation of CD4 + T cell responses<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong><br />

| 54<br />

against NS3 class II epitopes, which have been<br />

correlated with virus clearance. Ultimately, we<br />

propose clinical development of programmed DC<br />

vaccines <strong>for</strong> HCV immunotherapy with high<br />

capabilities of standardization.<br />

Session: On the Route to Clinical Application<br />

P 27<br />

Resolution of Chronic Ischemia by Regional<br />

Application of Embryonic eEPCs or Thymosin<br />

β4 in a Preclinical Pig Model<br />

Rabea Hinkel 1 , Alexander Wuchrer 1 , Jan C<br />

Horstkotte 1 , Corinna Lebherz 1 , Antonis K<br />

Hatzopoulos 1 , Ildiko Bock-Marquette 2 , Michael<br />

DiMaio 2 , Peter Boekstegers 1 , Christian Kupatt 1<br />

1Internal medicine I, Klinikum Großhadern, LMU<br />

München, <strong>Munich</strong>, <strong>German</strong>y; 2 Southwestern<br />

Medical Center, Dallas, USA<br />

In patients suffering from ischemic<br />

cardiomyopathy, lacking interventional or surgical<br />

treatment options, induction of neovascularisation<br />

via vascular growth factor application and cell<br />

therapy is a novel therapeutic approach. In a<br />

preclinical pig model model of chronic ischemia,<br />

we investigated the efficacy of regional<br />

application of murine eEPCs or an AAV-mediated<br />

longterm Thymosin β4 (Tβ4) overexpression.<br />

Methods: In vitro, human microvascular<br />

endothelial cells (HMECs) were embedded in<br />

matrigel and tube <strong>for</strong>mation was obtained. Cells<br />

were stimulated with conditioned media (CM)<br />

from eEPCs ± Tβ4shRNA. Tubes like structures<br />

per low power field were evaluated. In pigs (n = 6)<br />

a reduction stent graft was implanted into the<br />

circumflex artery, leading to complete occlusion<br />

at day 28. Retroinfusion of saline, 5 × 10 6 murine<br />

eEPCs or recombinant AAV2/9 rAAV.Tβ4 (5 ×<br />

10 12 viral particles) was per<strong>for</strong>med at d28. At<br />

days 28 and 56, global myocardial function<br />

(LVEDP) was assessed. Regional myocardial<br />

function (SES) and post mortem angiography<br />

were obtained at day 56. Tissue samples from<br />

the ischemic and non-ischemic tissue were<br />

harvested <strong>for</strong> capillary staining. Results: In vitro,<br />

HMECs incubated with conditioned media of the<br />

eEPCs revealed an increase of tube <strong>for</strong>mation<br />

which was comparable to the stimulation with<br />

VEGF (16 ± 1 control vs. 34 ± 2 VEGF stimulation<br />

and 31 ± 2 with eEPC CM tube-like structures/low<br />

power field). The reduction of Tβ4 expression in<br />

the eEPCs via shRNA abolished this effect (11 ±<br />

1). In the pig model, retroinfusion of eEPCs<br />

induced an increase of capillaries (200 ± 12 vs.


55 |<br />

112 ± 6 C/F in controls) and collaterals (4.4 ± 0.4<br />

vs. 1.2 ± 0.2 in controls), followed by improvement<br />

of the global and regional myocardial function<br />

(LVEDP: 14 ± 1 vs. 19 ± 1.2 mmHg in control<br />

group; SES 44 ± 4 vs. 10 ± 6% in control group).<br />

rAAV.Tβ4 displayed a similar gain of<br />

neovascularization and myocardial function. (223<br />

± 14 C/F; 6.4 ± 0.5 collaterals; 14 ± 0.5mmHg<br />

LVEDP; 56 ± 7% SES). Murine embryonic EPCs<br />

are capable of inducing angiogenesis and<br />

arteriogenesis in a Tβ4 dependent manner. The<br />

combination of AAV vector and Tβ4 may offer<br />

therapeutic potential in patients with ischemic<br />

cardiomyopathy.<br />

Session: On the Route to Clinical Application<br />

P 28<br />

AAV-Mediated VEGF-A and PDGF-B<br />

Overexpression Resolves Mal-perfusion in<br />

Chronic Ischemia: Role of Vessel Maturation<br />

Rabea Hinkel 1 , Achim Pfosser 1 , Michael<br />

Thormann 1 , Corinna Lebherz 1 , Alexander<br />

Wuchrer 1 , Franziska Globisch 1 , Chiraz El-Aouni 1 ,<br />

Andrea Banfi 2 , Peter Boekstegers 1 , Christian<br />

Kupatt 1<br />

1 Internal medicine I, Klinikum Groβhadern, LMU<br />

München, <strong>Munich</strong>, <strong>German</strong>y; 2 Cell and <strong>Gene</strong><br />

<strong>Therapy</strong>, Institute <strong>for</strong> surgical Research and<br />

Hospital Management, Basel, Switzerland<br />

In patients suffering from ischemic<br />

cardiomyopathy or peripheral artery disease,<br />

lacking interventional or surgical treatment<br />

options, induction of neovascularization via<br />

vascular growth factor application is a novel<br />

therapeutic concept. Here we tested an approach<br />

by combining hVEGF-A with hPDGF-B and<br />

prolonging the expression period by using a AAV<br />

as viral vector.<br />

Methods: In rabbits (n = 5/group), 0.5 × 10 11<br />

rAAV.VEGF-A ± 1 × 10 12 rAAV.hPDGF-B were<br />

retroinfused at d7 of femoral artery excision.<br />

Angiography of both hindlimbs was per<strong>for</strong>med <strong>for</strong><br />

collateral score and frame count score (% d7).<br />

Capillary density was determined at d35. In pigs,<br />

a reduction stent graft was implanted into the<br />

circumflex artery, leading to complete occlusion<br />

at day 28, when retroinfusion of rAAV.VEGF-<br />

A/PDGF-B (2 × 10 12 particles each), rAAV.VEGF-<br />

A (1 × 10 13 particles) or rAAV.LacZ was<br />

per<strong>for</strong>med. Global and regional myocardial<br />

function and perfusion were assessed at d28 and<br />

d56.<br />

Results: In rabbits, rAAV.hVEGF-A strongly<br />

induced angiogenesis (C/MF 1.32 ± 0.07 vs. 0.96<br />

± 0.08 in controls), but neither collateral growth<br />

(125 ± 7% d7 vs. 95 ± 6% d7) nor perfusion (136 ±<br />

12% vs. 107 ± 9%). hVEGF-A/hPDGF-B<br />

cotransfection, enhanced collateral growth (146 ±<br />

9%) and perfusion (163 ± 8%) at a similar<br />

capillary density (1.44 ± 0.10 CM/F). NG2 staining<br />

revealed a significant increase of pericytes after<br />

VEGFA/PDGF-B overexpression, in contrast to<br />

the VEGF-A group. In the pig model, retroinfusion<br />

of rAAV-VEGF-A/PDGF-B increased myocardial<br />

blood flow (1.5 ± 0.1ml/g vs. 1.0 ± 0,1ml/g in<br />

controls), based on collateral growth (9.5 ± 0.3 vs.<br />

5.5 ± 0.5 in controls) and capillary density (257 ±<br />

23 vs. 135 ± 6/field in controls). VEGF-A alone<br />

failed to enhance myocardial perfusion by the<br />

same capillary density (274 ± 12/field).<br />

rAAV.VEGF-A/PDGF-B improved global<br />

myocardial function (LVEDP: 14.7 ± 0.9 vs. 19.6 ±<br />

0.8mmHg in controls) and regional myocardial<br />

function in the RCx area (11 ± 3% SES vs. 2 ±<br />

6%, controls), an effect lacking in the<br />

rAAV.VEGF-A group (EF 37 ± 2%, SES 6 ± 3%).<br />

Long-term overexpression of hVEGF and PDGF<br />

substantially enhanced angio- and arteriogenesis<br />

in the ischemic region followed by an improved<br />

function, which is superior to hVEGF alone.<br />

Session: On the Route to Clinical Application<br />

P 29<br />

Inhibition of G-Protein Coupled Receptor<br />

Kinase 2 Prevents Myocardial Hypertrophy in<br />

a Mouse Model <strong>for</strong> Dystrophin-Deficient<br />

Cardiomyopathy<br />

Ralf Bauer, Stefanie Schinkel, Christian Volz,<br />

Hugo A Katus, Oliver J Müller<br />

Medizinische Klinik Heidelberg, Abteilung<br />

Kardiologie, Angiologie und Pneumologie,<br />

Heidelberg, <strong>German</strong>y<br />

Severe cardiomyopathy is a common feature in<br />

Duchenne muscular dystrophy (DMD) and has<br />

critical impact on mortality of these patients.<br />

Although, to date efficient treatment strategies to<br />

prevent cardiac complications in DMD remain<br />

elusive, cardiac-specific adeno-associated virus<br />

(AAV)-based gene therapy strategies represent a<br />

promising tool. In DMD, absence of dystrophin<br />

causes instability of the dystrophin-glycoproteincomplex<br />

and increased vulnerability of<br />

cardiomyocytes towards contraction-induced<br />

damage. It is unclear to what extend impairment<br />

of the cardiac beta-adrenergic receptor (BAR)<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong>


system contributes to progressive cardiac<br />

damage in DMD. It has been shown in transgenic<br />

mice that the inhibition of G-protein coupled<br />

receptor kinase 2 (GRK2) through the carboxylterminus<br />

of GRK2 (βARKct) has beneficial effects<br />

with an attenuation of heart failure symptoms.<br />

There<strong>for</strong>e, we have investigated the efficiency of<br />

AAV9- mediated cardiac overexpression of<br />

βARKct to prevent the development of<br />

cardiomyopathy in dystrophin-deficient (mdx)<br />

mice. AAV-9 vectors containing βARKct cDNA<br />

under transcriptional control of the CMV-MLC<br />

promoter (AAV9/βARKct) were created. 10 12<br />

AAV9/βARKct vector particles were intravenously<br />

injected into the tail vein of 8 week-old mdx mice<br />

be<strong>for</strong>e the onset of any signs of cardiomyopathy.<br />

In some mice AAV9 with enhanced green<br />

fluorescent protein (AAV9/EGFP) was injected as<br />

control vector. At the age of 12 months<br />

echocardiography to assess contractility<br />

(fractional shortening (FS)) and diastolic left<br />

ventricular posterior wall thickness (PWTd) was<br />

per<strong>for</strong>med. Untreated and AAV9/EGFP-treated<br />

mdx mice showed extensive myocardial<br />

hypertrophy and reduced contractility (PWTd<br />

16.5 + 0.04 and 16.3 + 0.14 mm; FS 45 + 9% and<br />

45 + 8%, respectively (values + standard error)).<br />

Treatment with AAV9/βARKct attenuated<br />

myocardial hypertrophy and improved<br />

contractility (PWTd 12.6 + 0.08 mm and FS 71 +<br />

3%) in mdx mice. We suggest that cardiac<br />

inhibition of GRK2 through AAV-mediated<br />

overexpression of βARKct is a valuable strategy<br />

limiting cardiac damage in muscular dystrophyassociated<br />

cardiomyopathy.<br />

Session: On the Route to Clinical Application<br />

P 30<br />

Non-Integrating Lentiviral <strong>Gene</strong> Transfer of<br />

E2F2 to Induce Corneal Endothelial Cell<br />

Replication<br />

Daniel Kampik, Alexander Smith, Ulrich F.O.<br />

Luhmann, Prateek Buch, Daniel F.P. Larkin,<br />

Robin R. Ali<br />

UCL Institute of Ophthalmology, London, UK<br />

The osmotic activity of corneal endothelial cells<br />

(CEC) maintains the clarity of the eye's cornea.<br />

CECs being non-replicative in humans, a<br />

decrease of CEC density due to aging, injury or<br />

inherited disease causes corneal swelling and<br />

loss of transparency. E2F2 is a transcription<br />

factor regulating progression from G1 to S phase<br />

of the cell cycle. We investigate whether gene<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong><br />

| 56<br />

transfer of E2F2 is able to induce cell proliferation<br />

in CEC. We constructed a non-integrating HIVbased<br />

lentiviral vector delivering E2F2 under a<br />

CMV promoter (LNT-E2F2). A lentiviral vector<br />

delivering GFP (LNT-GFP) served as control. In<br />

293T cells, exposure to LNT-E2F2 resulted in a ><br />

200-fold increase of E2F2 mRNA compared to<br />

uninfected or LNT-GFP infected controls, as<br />

detected by quantitative PCR. E2F2<br />

overexpression was detected by Western blot<br />

and immunohistochemistry. Samples of human<br />

corneal tissue were exposed to either LNT-E2F2<br />

or LNT-GFP or left uninfected. Exogenous E2F2<br />

protein was detectable only in LNT-E2F2 infected<br />

samples by Western blot and<br />

immunohistochemistry. 5-bromodeoxyuridine<br />

(BrdU) incorporation and Ki-67 immunostaining<br />

was used to detect G1 to S phase progression or<br />

cells in any active cell cycle phase, respectively.<br />

CECs infected with LNT-E2F2 showed both BrdU<br />

and Ki67 nuclear staining. Lentivirus mediated<br />

E2F2 overexpression could be used to increase<br />

CEC density in donor corneas ex vivo be<strong>for</strong>e<br />

transplantation. Safety measures are now being<br />

investigated to enable clinical use.<br />

Session: On the Route to Clinical Application<br />

P 31<br />

Membrane-Anchored and Secreted C<br />

Peptides as HIV Entry Inhibitors<br />

Lisa Egerer 1 , Janine Kimpel 1 , Andreas Volk 2 ,<br />

Joerg Kahle 3 , Marianne Hartmann 2 , Felix G.<br />

Hermann 3 , Sebastian Newrzela 2 , Dorothee von<br />

Laer 1<br />

1 Dept. of Hygiene, Microbiology and Social<br />

Medicine, Section Virology, Medical University<br />

Innsbruck, Innsbruck, Austria; 2 Applied Virology<br />

and <strong>Gene</strong> <strong>Therapy</strong>, Institute <strong>for</strong> Biomedical<br />

Research, Georg-Speyer-Haus, Frankfurt am<br />

Main, <strong>German</strong>y; 3 Vision 7 GmbH, Frankfurt am<br />

Main, <strong>German</strong>y<br />

C peptides derived from the HIV envelope<br />

glycoprotein gp41 (e.g., T-20, C46) are highly<br />

efficient inhibitors of virus entry. Here, we<br />

analysed a membrane-anchored (maC46) as well<br />

as a secreted (sC46) variant of the peptide C46<br />

<strong>for</strong> gene therapy of HIV infection. Membraneanchored<br />

or secreted C46 peptides were<br />

expressed from gammaretroviral or lentiviral<br />

vectors in B and T cell lines as well as in primary<br />

human T cells. Cultures were analysed <strong>for</strong><br />

expression of the peptides and antiviral effect.<br />

Furthermore, a humanized mouse model was


57 |<br />

estabished to test these antiviral peptides in vivo.<br />

The sC46 peptides were expressed and secreted<br />

in different lymphoid cells and exerted high<br />

antiviral activity against a variety of HIV envelope<br />

glycoproteins. In mixed cell cultures peptides<br />

secreted from transduced cells produced a<br />

bystander effect and prevented infection of nonmodified<br />

cells. In the mouse model, we observed<br />

a substantial increase of maC46 + CD4 + T cells<br />

in blood as well as in spleen, apparently due to<br />

the selective pressure of ongoing HIV infection.<br />

This increase of CD4 + T cells was neither seen<br />

in uninfected control mice nor with a control<br />

vector. The clear accumulation of maC46 + cells<br />

in HIV-infected humanized mice indicates that<br />

these cells are protected from HIV infection in<br />

vivo. Further studies in this model with maC46<br />

and sC46 will allow us to analyze the conditions<br />

that determine efficacy of T cell based<br />

immuno/gene therapy <strong>for</strong> HIV-infection.<br />

Session: <strong>Gene</strong> <strong>Therapy</strong><br />

P 32<br />

Self-Complementary rAAV8-iSAVE Vectors <strong>for</strong><br />

Liver-Targeted Entry-Inhibitory HIV-1 <strong>Gene</strong><br />

<strong>Therapy</strong><br />

Andreas Volk 1 , Janine Kimpel 1 , Lisa M Egerer 1 ,<br />

Hanna Janicki 2 , Hildegard Büning 2 , Dorothee von<br />

Laer 1<br />

1 Department of Virology, Medical University<br />

Innsbruck, Innsbruck, Austria; 2 Department I of<br />

Internal Medicine and Center <strong>for</strong> Molecular<br />

Medicine Cologne, Cologne, <strong>German</strong>y<br />

Antiviral entry-inhibitory C-peptides, a novel class<br />

of antivirals in HIV standard therapy (HAART),<br />

are derived from the C-heptad motif in the<br />

extracellular domain of the gp41 transmembrane<br />

protein. C-peptides (such as T-20 or C46) bind to<br />

a fusion intermediate state of gp41 and thereby<br />

block viral entry with an IC50 in the low nM range.<br />

T-20 is the only marketed C-peptide and is<br />

administered twice daily via subcutaneous<br />

injection. Major drawbacks of this drug are (i) the<br />

inconvenient route of administration and (ii) rapid<br />

viral resistance emergence. To circumvent these<br />

drawbacks, our group focuses on the<br />

development of an entry-inhibitory gene therapy<br />

based on an elongated variant of T-20, the ‘in<br />

vivo secreted antiviral entry inhibitory C-peptide’<br />

(iSAVE). In comparison to T-20, iSAVE also<br />

targets a very conserved membrane proximal<br />

region, which cannot be mutated without massive<br />

viral replication deficiencies. Additionally, a gene<br />

therapy ideally requires only one administration<br />

of antiviral and thus reduces the need <strong>for</strong><br />

constant patient adherence. In our gene<br />

therapeutical model, iSAVE expression can be<br />

targeted to several sites of production. Besides<br />

the lymphatic tissue as sites of viral replication,<br />

the liver is a very promising organ <strong>for</strong> iSAVE<br />

expression, since (i) hepatocytes are prone to<br />

secrete high amounts of protein and (ii) the liver<br />

is known to be able to induce a tolerogenic<br />

response towards transgenes. Here, we present<br />

first results of iSAVE delivered by a selfcomplementary<br />

rAAV8 vector to the liver. iSAVE<br />

is expressed to moderate nM plasma<br />

concentrations, while its IC50 is in the low nM<br />

range promising a sustained antiviral effect upon<br />

in vivo challenge. Furthermore, no humoral<br />

response against the iSAVE peptide could be<br />

detected in immunocompetent mice, pointing to<br />

an potential tolerogenic transgene response.<br />

Currently, we are investigating the antiviral effect<br />

in a humanized mouse model <strong>for</strong> HIV-infection. In<br />

conclusion, scAAV8-iSAVE is a promising<br />

candidate <strong>for</strong> HIV gene therapy and will be<br />

further analyzed <strong>for</strong> its antiviral activity and<br />

transgene-induced immunologic responses.<br />

Session: <strong>Gene</strong> <strong>Therapy</strong><br />

P 33<br />

<strong>Gene</strong> Vector Loaded Biphasic Calcium<br />

Phosphate Ceramic Induces Ectopic Bone<br />

Formation in Rats<br />

Christian Koch 1 , Andreas Kolk 2 , Balasz Vigh 1 ,<br />

Bernd Gänsbacher 1 , Christian Plank 1<br />

1Institute of Experimental Oncology and <strong>Therapy</strong><br />

Research, Technische Universität München,<br />

<strong>Munich</strong>, <strong>German</strong>y; 2 Department of Oral and<br />

Maxillofacial Surgery, Technische Universität<br />

München, <strong>Munich</strong>, <strong>German</strong>y<br />

TricOs is a resorbable bone substitute which is<br />

already used in clinical applications. This material<br />

is a biphasic calcium phosphate ceramic<br />

composed of a mixture of hydroxyapatite and βtricalcium<br />

phosphate which combines structural<br />

stability with osteogenic capacity. For faster and<br />

better healing of large bone defects the<br />

optimization of the biological properties of this<br />

substitute would be an advantage. This could be<br />

achieved by localized gene delivery using vectors<br />

coding, <strong>for</strong> e.g., a growth factor such as BMP-2.<br />

Based on our knowledge of localized gene<br />

delivery systems using copolymer protected gene<br />

vectors (COPROGs) and organic matrices such<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong>


as collagen sponges or fibrin glue, we examined<br />

whether a combination of COPROGs and the<br />

inorganic calcium ceramic <strong>for</strong> matrix mediated<br />

gene delivery is possible. For the in vitro studies<br />

we used a novel reporter gene, Metridia<br />

luciferase, a protein which is secreted in the<br />

tissue culture supernatant. We could<br />

demonstrate that our gene vector <strong>for</strong>mulation<br />

immobilized on TricOs revealed Metridia<br />

luciferase expression over a period of 48 days<br />

with a maximum at day 5. Detection of GFP<br />

expression exhibited a bright fluorescene of cells<br />

which colonized the TricOs granules. For in vivo<br />

studies we implanted TricOs granules loaded<br />

with COPROGs coding either <strong>for</strong> BMP-2 or firefly<br />

luciferase as control in the musculus latisimus<br />

dorsi in rats. At different time points the bone<br />

substitutes were explanted and the <strong>for</strong>mation of<br />

newly <strong>for</strong>med bone was examined by micro-CT,<br />

conventional CT and PETscan. In contrast to the<br />

luciferase implants BMP-2 gene vector loaded<br />

granules exhibited a better turnover of the bone<br />

substitute into newly <strong>for</strong>med bone and a higher<br />

bone density. The finding that ectopic bone<br />

<strong>for</strong>mation in the muscle of rats suggests that<br />

matrix mediated gene delivery is useful in bone<br />

regeneration applications.<br />

Session: <strong>Gene</strong> <strong>Therapy</strong><br />

P 34<br />

Bioactivation of Collagen Scaffolds with<br />

Nonviral <strong>Gene</strong> Vectors to Enhance the<br />

Release of Proangiogenic Molecules in Tissue<br />

Regeneration<br />

Ann K Reckhenrich 1 , Christian Koch 1 , Ursula<br />

Hopfner 2 , Hans G Machens 2 , Christian Plank 1 ,<br />

Tomas J Egana 2<br />

1Institute für Experimentelle Onkologie, Klinikum<br />

rechts der Isar, TU München, <strong>Munich</strong>, <strong>German</strong>y;<br />

2 . Klinik für Plastische Chirurgie und<br />

Handchirurgie, Klinikum rechts der Isar, TU<br />

München, <strong>Munich</strong>, <strong>German</strong>y<br />

Background: Biodegradable scaffolds serve as<br />

structural matrix <strong>for</strong> cell guiding, promoting cell<br />

growth and proliferation. Moreover, scaffolds can<br />

also be used as carriers <strong>for</strong> bioactive molecules.<br />

The combination of nanobiotechnology and<br />

tissue engineering offers the opportunity to<br />

temporarily modify cells by nucleic acid<br />

transporting nanoparticles. Under such<br />

conditions, infiltrating cells will be temporarily<br />

modified to express genes of interest. In this<br />

study we developed a new approach to activate<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong><br />

| 58<br />

biomaterials to release proangiogenic molecules<br />

by combining their use with a new class of nonviral<br />

gene vectors so called Copolymer-protected<br />

gene vectors (COPROGs).<br />

Methods: COPROGs were incorporated into<br />

commercially available collagen scaffolds, and<br />

the optimal concentration of DNA was<br />

determined in vitro by following the release of<br />

reporter proteins. After cell seeding into the<br />

scaffolds, distribution and attachment of the cells<br />

were evaluated by fluorescence microscopy and<br />

cell viability and proliferation by metabolic MTT<br />

assays. Finally, scaffolds were activated with<br />

vectors encoding <strong>for</strong> relevant growth factors (e.g.,<br />

VEGF165) and their release was quantified by<br />

ELISA.<br />

Results: We have determined that the optimal<br />

amount of COPROGs per scaffold was 0.4 μg<br />

(relative to DNA) per milligram of scaffold. After<br />

seeding, results showed that cells were<br />

homogeneously distributed <strong>for</strong>ming focal<br />

adhesions with the scaffold. Moreover, the<br />

presence of COPROGs did not affect cell viability<br />

and proliferation of seeded cells. The release of a<br />

reporter protein was detected <strong>for</strong> at least 2 weeks<br />

in vitro, with a peak of expression between days<br />

3 to 5. Similar results were obtained by the use of<br />

DNA encoding <strong>for</strong> VEGF, where a transient<br />

release of the protein was observed <strong>for</strong> at least 2<br />

weeks with a peak of expression at day 5.<br />

Conclusions: Combine use of collagen scaffolds<br />

and COPROGs, allows inducing local release of<br />

therapeutic proteins without cell toxicity in vitro.<br />

Such technologies may have tremendous impact<br />

in the field of tissue engineering and regenerative<br />

medicine. Preclinical studies are under current<br />

evaluation.<br />

Session: <strong>Gene</strong> <strong>Therapy</strong>


59 |<br />

P 35<br />

Evaluation of a Novel AAV/Transposase<br />

Hybrid-Vector System <strong>for</strong> Somatic Integration<br />

In Vitro and In Vivo<br />

Nadine Müther 1 , Lajos Mates 2 , Zsuzsanna<br />

Izsvak 2 , Zoltan Ivics 2 , Wenli Zhang 1 , Martin<br />

Hausl 1 , Nina Harms 1 , Nicola M. Wolf 1 , Anja<br />

Ehrhardt 1<br />

1Max von Pettenkofer-Institute, Department of<br />

Virology, Ludwig-Maximilians-University of<br />

<strong>Munich</strong>, <strong>Munich</strong>, <strong>German</strong>y; 2 Max Delbrück<br />

Center <strong>for</strong> Molecular Medicine, Berlin, <strong>German</strong>y<br />

Recombinant adeno-associated viral (rAAV)<br />

vectors predominantly persist as<br />

extrachromosomal genomes. However, in<br />

dividing cells, rAAV vector genome copy<br />

numbers and transgene expression levels decline<br />

rapidly. Herein, we inserted the hyperactive<br />

Sleeping Beauty (SB) transposase variants HSB5<br />

and the novel mutant SB100 into our AAV-based<br />

two-viral-vector system displaying 10- and 100fold<br />

increased integration efficiencies compared<br />

to wild-type SB, respectively. Our previous work<br />

showed that transposition only works sufficiently<br />

from circular substrates. However, once inside<br />

the cell rAAV genomes <strong>for</strong>m various episomal<br />

DNA <strong>for</strong>ms including circular and linear DNA<br />

molecules. Thus, in initial experiments we<br />

addressed whether Flp recombinase-mediated<br />

circularization of the transposon from rAAV<br />

genome is a precondition of our system. Our data<br />

suggested that Flp-mediated circularization may<br />

not be required <strong>for</strong> AAV-delivered transposase<br />

activity significantly reducing the complexity of<br />

our system. In order to avoid co-transduction of<br />

two AAV-vectors into the same cell we generated<br />

a stably SB100 expressing cell line. To analyze<br />

integration efficiencies we per<strong>for</strong>med colony<br />

<strong>for</strong>ming assays and found that after infection with<br />

an rAAV with a neomycin encoding transposon<br />

(MOI 10.000), the integration efficiencies in<br />

SB100 cells was 5-fold increased compared to<br />

the control cell line with inactive SB (mSB). This<br />

demonstrated that inserting the SB transposase<br />

system into rAAV increases integration efficiency<br />

significantly. We are in the process to determine<br />

sites of insertion after SB100-mediated<br />

integration from the rAAV vector into the host<br />

genome by using a plasmid rescue strategy. To<br />

address the question whether the new version of<br />

the rAAV hybrid-vector system also results into<br />

stable transgene expression levels in vivo, we<br />

coinjected C57Bl/6 mice with the rAAV<br />

transposon donor vector expressing the<br />

coagulation factor IX and HSB5 delivered by an<br />

adenoviral vector. After induction of rapid cell<br />

cycling in mouse liver, transgene expression<br />

levels were more stable in mice which received<br />

HSB5 compared to the control mSB group<br />

indicating that transposition works from rAAV.<br />

Session: <strong>Gene</strong> <strong>Therapy</strong><br />

P 36<br />

AAV2.9 Thymosin β4 Regional Application<br />

Enhances Therapeutic Neovascularization<br />

and Requires AKT Activation and Capillary<br />

Sprouting in the Calf Muscle<br />

Rabea Hinkel 1 , Teresa Trenkwalder 1 , Elena<br />

Gottlieb 1 , Achim Pfosser 1 , Shahana Sultana 1 ,<br />

Franziska Globisch 1 , Georg Stachel 1 , Corinna<br />

Lebherz 1 , Ildiko Bock-Marquette 2 , Christian<br />

Kupatt 1<br />

1<br />

Internal medicine I, Klinikum Großhadern, LMU<br />

2<br />

München, <strong>Munich</strong>, <strong>German</strong>y; Southwestern<br />

Medical Center, Dallas, Texas, USA<br />

Thymosin β4, an endogenously occurring peptide<br />

of 5 kDa, is cardioprotective after ischemia and<br />

reperfusion in a Protein Kinase B (AKT)<br />

dependent manner. Furthermore it is essential <strong>for</strong><br />

coronary vessel development and provides<br />

angiogenesis during wound healing of the adult<br />

organism. Here we tested if long-term<br />

overexpression of Thymosin β4 with an adenoassociated<br />

virus is beneficial <strong>for</strong> therapeutic<br />

neovascularisation in a chronic in vivo model of<br />

hindlimb ischemia.<br />

Methods: In rabbits (n = 5/group), femoral artery<br />

excision was per<strong>for</strong>med at d0 and 5 × 10 12<br />

AAV2/9 expressing Tβ4 or Lac-Z were applied<br />

through intramuscular injection. Tβ4 was either<br />

injected over the whole limb or only into the calf<br />

muscles. An additional group received Tβ4 whole<br />

limb and 1 × 10 13 AAV 2/9 AKT-DN into the lower<br />

limb only (i.m.). At day 7 and day 35 angiography<br />

was per<strong>for</strong>med <strong>for</strong> collateral quantification (% of<br />

day 7) and frame count score (% of day 7) and<br />

fluorescent microspheres were applied to<br />

determine the regional blood flow. At day 35<br />

tissue was harvested <strong>for</strong> determination of<br />

capillary density via immunostaining <strong>for</strong> PECAM-<br />

1 (c/mf).<br />

Results: rAAV-Tβ4 significantly increasd<br />

capillary density in the ischemic limb (1.6 ± 0.2<br />

c/mf) compared to the control group (0.96 ±<br />

0.1c/mf). Quantification of collateral growth (175 ±<br />

15% vs. 95 ± 6% in controls) and microspheres<br />

(157 ± 10 vs. 110 ± 6 in controls) yielded similar<br />

results after Tβ4 transfection. However<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong>


cotransfection of AKT-DN blunted the effect (c/mf<br />

1.3 ± 0.2, collaterals 126 ± 11%; microspheres<br />

107 ± 3%). Moreover, calf muscle transduction of<br />

Tβ4 sufficed to induce collateral growth (176.84 ±<br />

4.1%) and gain of perfusion (160 ± 12.1%) to the<br />

same extent as whole limb Tβ4 transduction.<br />

Conclusion: Thymosin β4 long-term<br />

overexpression exerts therapeutic<br />

neovascularisation in chronic ischemia. The<br />

down-regulation of activated AKT in the lower<br />

limb only suppresses capillary <strong>for</strong>mation and<br />

subsequent collateral growth. In contrast,<br />

stimulation of microcirculatory growth using Tβ4<br />

in calf muscles only sufficed to promote<br />

arteriogenesis in the upper leg. These results<br />

suggest a backward signalling from<br />

microcirculation to conductance vessel <strong>for</strong>mation.<br />

Session: <strong>Gene</strong> <strong>Therapy</strong><br />

P 37<br />

Extensive Methylation of SFFV Promoter<br />

Sequences Does Not Allow Lentiviral<br />

Transgene Expression In Vivo<br />

Friederike Herbst 1 , Claudia R Ball 1 , Francesca<br />

Tuorto 2 , Wei Wang 2 , Ulrich Kloz 2 , Franciscus van<br />

der Hoeven 2 , Frank Lyko 2 , Manfred Schmidt 1 ,<br />

Christof von Kalle 1 , Hanno Glimm 1<br />

1 National Center <strong>for</strong> Tumour Diseases (NCT) &<br />

<strong>German</strong> Cancer Research Center (DKFZ),<br />

Heidelberg, <strong>German</strong>y; 2 <strong>German</strong> Cancer<br />

Research Center (DKFZ), Heidelberg, <strong>German</strong>y<br />

Lentiviral vectors (LV) possess a high stability of<br />

expression in vivo and are often used to<br />

investigate genes and their functions. Lentiviral<br />

gene transfer is considered to be a promising<br />

less time consuming technology to facilitate the<br />

generation of transgenic mice with a higher yield<br />

of transgenic offspring as compared to the<br />

commonly used DNA microinjection. We applied<br />

LV to generate a mouse model transgenic <strong>for</strong><br />

SETBP1 and eGFP. Lentiviral particles were<br />

injected into the perivitelline space of early stage<br />

embryos. LV integration was detected in newborn<br />

animals (F0) using PCRs specific <strong>for</strong> either the<br />

SETBP1 transgene or <strong>for</strong> the WPRE element of<br />

the construct. Lentiviral integration sites were<br />

detected in 65% of 31 analyzed F0 mice by<br />

highly sensitive LAM-PCR. 5 out of 9 F0 mice<br />

showed germline transmission, revealing a total<br />

of 33% vector positive offspring. However, no<br />

ectopic transcription and overexpression of<br />

neither SETBP1 nor eGFP could be detected in<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong><br />

| 60<br />

transgenic mice. We there<strong>for</strong>e analyzed the<br />

methylation status of the internal SFFV promoter<br />

(SFFVp) by bisulfite sequencing. 18 of 18<br />

analyzed CpGs within the promoter region were<br />

extensively methylated in F0 animals and in all<br />

progeny determined (n = 12). To exclude<br />

transgene effects on epigenetic silencing of<br />

SFFVp sequences in self-inactivating LVs, we<br />

transduced mES cells with<br />

LV.SFFV.Setbp1.IRES.eGFP or the<br />

corresponding eGFP-expressing control vector.<br />

eGFP expression decreased 1.8 fold and 3.5 fold<br />

after differentiation of ES cells infected with the<br />

transgene vector and SFFV driven control vector,<br />

respectively. Further, we compared the<br />

methylation status of SFFVp sequences of<br />

gammaretroviral (RV) and LV in peripheral blood<br />

of bone marrow transplanted mice (n = 7) 3<br />

months after transplantation. Strikingly, LV<br />

transplanted mice showed a higher degree of<br />

methylation. Here, we demonstrate that the<br />

commonly used SFFV promoter is highly<br />

methylated with remarkable strength and<br />

frequency during development in vivo and<br />

differentiation in vitro. We conclude that LV using<br />

an internal SFFV promoter are not suitable <strong>for</strong><br />

the generation of transgenic mice or constitutive<br />

expression studies in hematopoietic cells.<br />

Session: <strong>Gene</strong> <strong>Therapy</strong><br />

P 38<br />

Effect of Nogo-B Overexpression in Vascular<br />

Smooth Muscle Cell Migration and<br />

Proliferation Mediated by Integration-Deficient<br />

Lentiviral Vectors<br />

Helen Chick 1 , RA McDonald 1 , AB Kritz 1 , NM<br />

Kane 1 , R Alba 1 , WC Sessa 2 , AJ Thrasher 3 , AH<br />

Baker 1<br />

1BHF Glasgow Cardiovascular Research Centre,<br />

Division of Cardiovascular and Medical Sciences,<br />

University of Glasgow, Glasgow, UK;<br />

2 Department of Pharmacology and Program in<br />

Vascular Signaling and Therapeutics, Boyer<br />

Centre <strong>for</strong> Molecular Medicine, Yale University<br />

School of Medicine, New Haven, Connecticut,<br />

USA; 3 Molecular Immunology Unit, UCL, Institute<br />

of Child Health, London, UK<br />

Proliferation and migration of vascular smooth<br />

muscle cells (VSMCs) is a hallmark of neointima<br />

<strong>for</strong>mation (NI) associated with acute vascular<br />

injury. Nogo-B is a member of the reticulon 4<br />

family of proteins. Nogo-B is highly expressed in<br />

vascular endothelial cells and VSMCs and


61 |<br />

regulates vascular remodeling. Previous studies<br />

have demonstrated that adenoviral-mediated<br />

overexpression of Nogo-B reduced NI after acute<br />

vascular injury in animal models. This effect was<br />

mediated through inhibition of VSMC migration<br />

and proliferation. Lentiviral vectors (LVs) are<br />

efficient at targeting VSMCs and the recent<br />

development of integration-deficient lentiviral<br />

vectors (IDLVs) offers additional potential <strong>for</strong> LVs<br />

in vascular gene delivery. Here, we have<br />

assessed Nogo-B overexpression in VSMCs<br />

mediated by IDLVs in vitro, as a potential<br />

therapeutic strategy <strong>for</strong> the prevention of NI in<br />

vivo. Immunofluorescence and western blot<br />

analysis indicated that IDLVs expressing Nogo-B<br />

efficiently increased Nogo-B expression in<br />

VSMCs, compared to IDLVs expressing GFP and<br />

no virus controls. Assessment of proliferation<br />

demonstrated that IDLVs overexpressing Nogo-<br />

B, led to a significant decrease in VSMC<br />

proliferation compared to IDLVs expressing GFP<br />

(multiplicity of infection (MOI) 25: 0.14 ± 0.0084<br />

absorbance (abs) at 570 nm vs. 0.28 ± 0.01 abs, p<br />

< 0.05). A wound-mediated cell migration assay<br />

demonstrated that IDLV delivery of Nogo-B<br />

significantly reduced the migration of VSMCs,<br />

compared to the IDLVs expressing GFP (MOI 25:<br />

55 ± 4.9 μm vs. 113 ± 7.6 μm, p < 0.05). Taken<br />

together, our study demonstrates that IDLVs are<br />

efficient in mediating overexpression of Nogo-B<br />

in VSMCs, leading to phenotypic effects on<br />

migration and proliferation. This has important<br />

implications <strong>for</strong> the use of IDLVs as a potential<br />

therapeutic vector <strong>for</strong> the prevention of NI during<br />

acute vascular injury.<br />

Session: <strong>Gene</strong> <strong>Therapy</strong><br />

P 39<br />

Combining Oncolytic Adenoviruses with<br />

Expression of Therapeutic Antibodies<br />

Dominik E Dorer 1 , Roland Kontermann 2 , Dirk M<br />

Nettelbeck 1<br />

1 Helmholtz University Group Oncolytic<br />

Adenoviruses, DKFZ and Department of<br />

Dermatology, Heidelberg University, Heidelberg,<br />

<strong>German</strong>y; 2 Institute <strong>for</strong> Cell Biology and<br />

Immunology, University of Stuttgart, Stuttgart,<br />

<strong>German</strong>y<br />

Oncolytic viruses which selectively kill tumor cells<br />

are emerging tools <strong>for</strong> cancer treatment. We seek<br />

to combine adenoviral oncolysis and therapeutic<br />

antibody expression in order to further increase<br />

anti-cancer efficacy through antibody-mediated<br />

toxicity. There<strong>for</strong>e, we created a model system<br />

by arming our adenoviruses with a recombinant<br />

single-chain antibody directed against the well<br />

established carcinoembryonic cancer antigen<br />

(CEA) fused to the constant domain of IgG2a<br />

(scFvCEA-Fc). To ensure cancer cell specific<br />

replication, replication-competent viruses were<br />

engineered to bear a 24 bp deletion in the E1A<br />

region and a chimeric 5/3 fiber to further increase<br />

infectivity <strong>for</strong> various cancer entities.<br />

Furthermore, we used different genomic positions<br />

and genetic tools <strong>for</strong> our scFvCEA-Fc transgene<br />

insertion. Among these we investigated splice<br />

acceptor sites derived from the beta-actin gene,<br />

the adenoviral pIII gene, and the Ad40 long fiber<br />

gene and compared resulting transgene levels to<br />

expression via established tools such as an<br />

internal ribosomal entry site or a nonreplicating<br />

adenovirus containing a cytomegalovirus early<br />

promoter. Infection of various CEA + and CEA −<br />

cell lines with any of our recombinant<br />

adenoviruses led to detectable amounts of<br />

scFvCEA-Fc in cell culture supernatants and cell<br />

pellets. However, transgene levels were strongly<br />

varying depending on the genomic loci and<br />

genetic tool used, giving the highest amount with<br />

the Ad40 splice acceptor. The expressed<br />

scFvCEA-Fc was then further characterized in<br />

flow cytometry, SDS-page, ELISA, and<br />

cytotoxicity assays. Our results show that<br />

scFvCEA-Fc binds only to cells expressing CEA<br />

or to purified antigen in a dose-dependent way<br />

and that transgene expression/antibody mediated<br />

cytotoxicity does not interfere with adenoviral<br />

replication and hence oncolysis. Currently, we<br />

are investigating anti-tumoral efficacy of immune<br />

cells in presence of scFvCEA-Fc in vitro and<br />

eventually in vivo. In the end, we want to create a<br />

highly efficient oncolytic adenovirus plat<strong>for</strong>m that<br />

combines virus-induced tumor cell killing and<br />

expression of therapeutic antibodies with<br />

systemic activity directed against various cancer<br />

markers.<br />

Session: Cancer <strong>Gene</strong> <strong>Therapy</strong><br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong>


P 40<br />

TNF Alpha <strong>Gene</strong> <strong>Therapy</strong> Synergizes with<br />

Liposomal Doxorubicin in the Treatment of<br />

Experimental Neuroblastoma and Hepatoma<br />

in Mice<br />

Baowei Su 1 , Ernst Wagner 1,2 , Manfred Ogris 1,2<br />

1 Pharmaceutical Biotechnology, Department of<br />

Pharmacy, Ludwig-Maximilians-University,<br />

<strong>Munich</strong>, <strong>German</strong>y; 2 Center <strong>for</strong> Nanoscience<br />

(CeNS), Ludwig-Maximilians-University, <strong>Munich</strong>,<br />

<strong>German</strong>y<br />

Tumor necrosis factor alpha (TNF) is a highly<br />

potent cytokine involved in endothelial cell<br />

activation, inflammation and tumor cell killing. As<br />

systemic application of TNF protein can be highly<br />

toxic, tumor localized expression of the cytokine<br />

after gene delivery is a potentially suited<br />

approach. We could recently show that systemic<br />

TNF gene delivery promoted tumor accumulation<br />

of liposomally encapsulated doxorubicine<br />

(Caelyx). Here we present the therapeutic effect<br />

of this treatment on subcutaneous Neuro2A<br />

murine neuroblastoma and enhanced Caelyx<br />

accumulation in HUH7 human hepatocellular<br />

carcinoma. A/J mice bearing subcutaneous,<br />

syngeneic Neuro2A tumors were treated with<br />

polyplexes based on a biodegradable polymer<br />

(G3-HD-OEI), HUH7 hepatoma xenografts in<br />

SCID mice received TNF polyplexes based on<br />

linear polyethylenimine carrying a peptide (GE11)<br />

selectively binding to the epidermal growth factor<br />

receptor, which is highly overexpressed in HUH7.<br />

When pretreating mice with TNF polyplexes,<br />

improved tumor accumulation of Caelyx was<br />

observed in both tumor models as compared to<br />

control polyplexes plus Caelyx. With the help of<br />

live bioluminescense imaging, enhanced tumor<br />

accumulation of Caelyx fluorescently labeled with<br />

the lipid DiR emitting in the near infrared was<br />

observed in living animals. This suggests a TNF<br />

mediated increase of tumor blood vessel<br />

permeability augmenting the accumulation of<br />

liposomal chemotherapeutics. In the Neuro2A<br />

model, three rounds of systemic TNF polyplex<br />

application and subsequent Caelyx treatment led<br />

to tumor growth inhibition lasting until eight days<br />

after the last treatment. Regrowth of treated<br />

tumors could be inhibited by a second treatment<br />

cycle indicating that no resistance towards<br />

combined TNF/Caelyx treatment occurred.<br />

Histological staining of cryosection showed<br />

significant decrease of CD31 expression in the<br />

re-treated group, which indicates a decreased<br />

vessel density caused by TNF/Caelyx treatment.<br />

In conclusion, combining liposomal<br />

chemotherapeutic with TNF gene therapy leads<br />

to a synergistic effect on tumor reduction in vivo.<br />

Session: Cancer <strong>Gene</strong> <strong>Therapy</strong><br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong><br />

| 62<br />

P 41<br />

Harnessing Molecular Targets in Human<br />

Colon Cancer Cells <strong>for</strong> Tumor Specific RNAi<br />

and pDNA-Based Therapies<br />

Arzu Cengizeroglu 1 , Katarina Farkasova 1 ,<br />

Daniela Deutsch 1 , Rudolf Haase 1 , Martina<br />

Anton 2 , Ernst Wagner 1,3 , Manfred Ogris 1,3<br />

1Pharmaceutical Biotechnology, Department of<br />

Pharmacy, Ludwig-Maximilians-University,<br />

<strong>Munich</strong>, <strong>German</strong>y; 2 Institute of Experimental<br />

Oncology and <strong>Therapy</strong> Research, Technische<br />

Universität München, <strong>Munich</strong>, <strong>German</strong>y; 3 Center<br />

<strong>for</strong> Nanoscience (CeNS), Ludwig-Maximilians-<br />

University, <strong>Munich</strong>, <strong>German</strong>y<br />

A major challenge in cancer therapy is to achieve<br />

high levels of specificity and efficacy preventing<br />

cancer cells to escape apoptosis, which<br />

otherwise leads to uncontrolled proliferation. For<br />

targeted cancer gene therapy a tumor cell<br />

specific promoter would be essential, which is<br />

inactive in nontrans<strong>for</strong>med tissue and at the<br />

same allows <strong>for</strong> high levels of transgene<br />

expression in tumor cells. Moreover, the tumorspecific<br />

activity of such a promoter can be utilized<br />

<strong>for</strong> tumor detection as well as measuring antitumoral<br />

effects. In gastrointestinal tumors, the<br />

constitutive activation of the wnt-pathway leads to<br />

malignancy due to high levels of the transcription<br />

factor beta-catenin in the nucleus, which in turn<br />

constitutively activates target genes causing<br />

enhanced proliferation, invasion and metastasis.<br />

Here we describe the use of the synthetic betacatenin<br />

dependent promoter CTP4 <strong>for</strong> specific<br />

expression of toxic transgenes, like the VP3<br />

subunit of the chicken anemia virus (Apoptin). In<br />

addition, CTP4 driven luciferase expression<br />

allowed us to evaluate the effect of RNAi<br />

mediated knockdown of wnt components on<br />

beta-catenin mediated transcription. Early<br />

passages of human colorectal cancer cell lines<br />

and control cell lines (HeLa and U87MG<br />

glioblastoma) were either transiently transfected<br />

with CTP4-luciferase plasmid or stably<br />

transduced with a lentiviral vector encoding <strong>for</strong><br />

CTP4 driven luciferase. Only cell lines with a<br />

deregulated wnt/beta-catenin signaling showed<br />

transgene expression, whereas in control cell<br />

lines transgene expression was below the level of<br />

the control vector without promoter. Knockdown<br />

of beta-catenin with siRNA clearly reduced CTP4<br />

driven luciferase expression, and in a similar way<br />

other factors were targeted by siRNA


63 |<br />

transfection. For plasmid based therapies we<br />

were able to show that in wnt/beta-catenin<br />

deregulated colon carcinoma cells CTP4 driven<br />

apoptin expression led to more prominent cell<br />

killing when compared to control cells. Taken<br />

together, these results provide rationales <strong>for</strong><br />

further preclinical in vivo testing of<br />

transcriptionally targeted plasmid vectors and<br />

siRNA based therapies <strong>for</strong> colorectal cancer.<br />

Session: Cancer <strong>Gene</strong> <strong>Therapy</strong><br />

P 42<br />

Liver Cancer Protease Activity Supports MMP<br />

Activatable Oncolytic Measles Virus<br />

Michael D. Muehlebach 1 , Thomas Schaser 1 ,<br />

Martina Zimmermann 2 , Sorin Armeanu 2 , Kay-<br />

Martin O. Hanschmann 3 , Michael Bitzer 2 , Ullrich<br />

Lauer 2 , Roberto Cattaneo 4 , Klaus Cichutek 1 ,<br />

Christian J. Buchholz 1<br />

1 Division of Medical Biotechnology, Paul-Ehrlich-<br />

Insitut, Langen, <strong>German</strong>y; 2 Innere Medizin,<br />

Medizinische Universitätsklinik, Tübingen,<br />

<strong>German</strong>y; 3 Biostatistics Section, Paul-Ehrlich-<br />

Institut, Langen, <strong>German</strong>y; 4 Molecular Medicine,<br />

Mayo Clinic, Rochester (MN), USA<br />

Primary and secondary cancers of the liver are a<br />

significant health problem with limited treatment<br />

options. We sought to develop an oncolytic<br />

measles virus (MV) preferentially activated in<br />

liver tumor tissue thus reducing infection and<br />

destruction of healthy tissue. Making cell entry of<br />

measles virus (MV) dependent on tumourassociated<br />

matrix metalloproteases (MMPs)<br />

restricts virus propagation to tumour cells and<br />

improves the virus safety in xenograft tumour<br />

models. We documented with specimen of 44<br />

patients that in primary tumor tissue urokinase<br />

type plasminogen activator (uPA) and especially<br />

matrix metallproteinase-2 (MMP-2) are<br />

significantly more active than in adjacent nontumorous<br />

tissue. When allocated to specific<br />

tumor entities, the overall tendency observed in<br />

the whole collective was confirmed. We then<br />

generated variants of the MV fusion protein by<br />

inserting different MMP-substrate motifs at the<br />

protease cleavage site, and identified the motif<br />

PQGLYA as the most efficient cleavage site as<br />

determined by syncytia <strong>for</strong>mation on protease<br />

positive tumor cells. The corresponding MMPactivatable<br />

oncolytic MV-MMPA1 virus was<br />

rescued and shown to be strongly restricted on<br />

primary human hepatocytes and healthy human<br />

liver tissue, while remaining as effective as the<br />

parental MV in tumor tissue sections on tissue<br />

slices of 10 patients. Thereby, a strong trend (p =<br />

0.09) <strong>for</strong> improved tumor targeting of MV-MMPA1<br />

was revealed, which correlated to the upregulation<br />

of MMP-2 activity in tumorous tissue (r<br />

= 0.7388). These data underline the clinical<br />

potency of the MMP-activation concept to<br />

generate safer oncolytic viruses <strong>for</strong> the treatment<br />

of primary and secondary cancers of the liver.<br />

Session: Cancer <strong>Gene</strong> <strong>Therapy</strong><br />

P 43<br />

Specific LNA-ASOs Against Oncogenic p73<br />

Iso<strong>for</strong>ms Permit Therapeutic Cancer<br />

Targeting In Vivo<br />

Stephan Emmrich 1 , Weiwei Wang 2 , Katja John 1 ,<br />

Wenzhong Li 2 , Brigitte M Pützer 1<br />

1 Department of Vectorology and Experimental<br />

<strong>Gene</strong> <strong>Therapy</strong>, Biomedical Research Center,<br />

University of Rostock, Rostock, <strong>German</strong>y;<br />

2 Department of Cardiac Surgery, University of<br />

Rostock, Rostock, <strong>German</strong>y<br />

Differential mRNA splicing and alternative<br />

promoter usage of the TP73 tumor suppressor<br />

gene results in the expression of multiple NH2truncated<br />

iso<strong>for</strong>ms that act as oncogenes.<br />

Abundant levels of these p73 variants in a variety<br />

of human cancers correlate with adverse clinical<br />

prognosis and response failure to conventional<br />

therapies, underscoring their relevance as<br />

marker <strong>for</strong> disease severity and target <strong>for</strong> cancer<br />

intervention. With respect to an equally important<br />

role <strong>for</strong> amino-truncated p73 splice <strong>for</strong>ms<br />

(DeltaTAp73) and DeltaNp73 (summarized as<br />

DNp73) in the tumorigenic process, we designed<br />

locked nucleic acid (LNA) antisense<br />

oligonucleotide (ASO) gapmers against individual<br />

species that were complementary to DeltaEx2<br />

and DeltaEx2/3 splice junctions and a region in<br />

exon 3B unique <strong>for</strong> ′ DeltaN and DeltaN.<br />

Treatment of cancer cells with these ASOs<br />

resulted in a strong and specific reduction of<br />

tumorigenic p73 transcripts and proteins,<br />

importantly, without abolishing the wild-type p73<br />

tumor suppressor <strong>for</strong>m as observed with p73shRNA.<br />

The specific antisense oligonucleotides<br />

rescued cells from apoptosis inhibition due to<br />

overexpression of their corresponding aminotruncated<br />

p73 iso<strong>for</strong>m and decreased tumor cell<br />

proliferation. Furthermore, ASO-116 against<br />

DeltaEx2/3 coupled to magnetic nanobead<br />

polyethyleneimine (MNB/PEI) carriers<br />

significantly inhibited malignant melanoma<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong>


growth, which correlated with a shift in the<br />

balance between endogenous TAp73 and<br />

DeltaEx2/3 towards apoptotic full-length p73. Our<br />

study demonstrates the successful development<br />

of LNA-ASOs that selectively differentiate<br />

between the closely related p73 oncoproteins,<br />

and provide new tools to further delineate their<br />

biological properties in different human<br />

malignancies and <strong>for</strong> therapeutic cancer<br />

targeting.<br />

Session: Cancer <strong>Gene</strong> <strong>Therapy</strong><br />

P 44<br />

Oncolytic Enhancement of VSV Through<br />

Inhibition of Host Proinflammatory Innate<br />

Immune Responses by Vector-Mediated<br />

Expression of NFκB Super-Repressor<br />

Jennifer Altomonte, Sabrina Marozin, Oliver<br />

Ebert<br />

II. Medizinische Klinik und Poliklinik, Klinikum<br />

rechts der Isar, TU München, <strong>Munich</strong>, <strong>German</strong>y<br />

Vesicular stomatitis virus (VSV) vectors harboring<br />

the MD51 mutation in the endogenous matrix (M)<br />

protein are potent inducers of interferon (IFN) in<br />

non-neoplastic cells, making them safer oncolytic<br />

agents than the wild-type virus. However, we<br />

have observed substantial attenuation of<br />

intratumoral replication of these vectors in our<br />

immune-competent, orthotopic rat model of HCC,<br />

significantly limiting the therapeutic efficacy.<br />

Following a peak in rVSV(MD51) titers in infected<br />

tumors after only 24 hours, a rapid and<br />

logarithmic reduction occurs, which is<br />

coincidental with the massive infiltration of<br />

inflammatory cells to the infected tumor sites. We<br />

hypothesized that intratumoral virus replication,<br />

and hence the oncolytic potency of rVSV(MD51)<br />

could be significantly enhanced by blocking antiviral<br />

inflammatory responses triggered by<br />

induction of the NF-κB pathway following viral<br />

infection without substantially interfering with the<br />

vector's intrinsic interferon (IFN) induction<br />

capacity to protect non-tumor cells. To test this<br />

hypothesis, we incorporated srIkBalpha, the socalled<br />

NF-κB super-repressor into the full-length<br />

VSV(MD51) vector. In vitro characterization<br />

revealed that expression of srIκBalpha as an<br />

additional transcription unit in the rVSV(MD51)<br />

backbone successfully blocked virus-mediated<br />

activation of NF-κB in primary human<br />

hepatocytes. Although reporter assays<br />

demonstrated that srIκBalpha expression partially<br />

interferes with the robust IFN induction and<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong><br />

| 64<br />

response to the rVSV(MD51) vector, the<br />

rVSV(MD51)-srIkB vector is nevertheless able to<br />

induce IFN significantly better than wild-type<br />

VSV. The advantage of such an intermediate<br />

vector is that it would maintain an improved<br />

safety profile over wild-type, while maintaining<br />

high levels of replication within the tumor. We<br />

predict that this strategy will prolong the kinetics<br />

of rVSV(M51R) replication, resulting in an<br />

improved oncolytic agent. We further hypothesize<br />

that a general reduction of inflammation could<br />

potentially allow us to safely administer higher<br />

doses of virus. These questions will be answered<br />

through comprehensive toxicity and efficacy<br />

studies, which are underway in HCC tumorbearing<br />

immune-competent rats.<br />

Session: Cancer <strong>Gene</strong> <strong>Therapy</strong><br />

P 45<br />

Noninvasive In Vivo Monitoring of Tumor<br />

Responses to Oncolytic Viral <strong>Therapy</strong> of<br />

Hepatocellular Carcinoma in Rats by [ 18 F]-<br />

FDG-PET and MRI<br />

Jennifer Altomonte 1 , Andreas K Buck 2 , Rickmer<br />

Braren 3 , Oliver Ebert 1<br />

1 II. Medizinische Klinik und Poliklinik, Klinikum<br />

rechts der Isar, TU München, <strong>Munich</strong>, <strong>German</strong>y;<br />

2 Nuklearmedizinische Klinik und Poliklinik,<br />

Klinikum rechts der Isar, TU München, <strong>Munich</strong>,<br />

<strong>German</strong>y; 3 Institut für Röntgendiagnostik,<br />

Klinikum rechts der Isar, TU München, <strong>Munich</strong>,<br />

<strong>German</strong>y<br />

Oncolytic viruses are currently under intense<br />

development as alternative cancer therapies.<br />

Preclinical and clinical studies will benefit<br />

significantly from the development of noninvasive<br />

imaging modalities to repetitively monitor key<br />

parameters of tumor response. As oncolytic<br />

viruses can kill tumor cells by distinct<br />

mechanisms from conventional therapies, they<br />

require new approaches to accurately evaluate<br />

responses. We hypothesized that the<br />

combination of positron emission tomography<br />

(PET) and magnetic resonance (MR) imaging<br />

could in<strong>for</strong>matively monitor the effects of<br />

vesicular stomatitis virus (VSV) treatment of HCC<br />

tumors. To this end, VSV or control buffer were<br />

infused through the hepatic arteries of multifocal<br />

HCC-bearing rats, and the animals were imaged<br />

by PET and MRI on days − 1, 1, and 3 posttreatment.<br />

In addition, a subset of animals was<br />

euthanized at each time-point in order to validate<br />

the imaging data with histology. For PET


65 |<br />

imaging, we utilized the 18 F-fluorodeoxyglucose<br />

([ 18 F]-FDG) tracer, which is an analog of glucose<br />

that is taken up by metabolically active cells,<br />

such as tumor cells. Analysis of the isocontour 50<br />

(50% of maximum) of tumor intensities revealed<br />

a significant decrease in [ 18 F]-FDG uptake<br />

following VSV treatment, indicating a reduction of<br />

metabolic activity, as compared to buffer controls.<br />

To determine whether this reduction correlated<br />

with increased tumor necrosis, histological<br />

sections of corresponding tumors were subjected<br />

to morphometric analysis of necrotic areas.<br />

Interestingly, a strong correlation of the two<br />

parameters existed only <strong>for</strong> the buffer control<br />

group. Further investigation by analysis of MRI<br />

and histological data revealed complex<br />

processes underway at early time-points postviral<br />

therapy, such as necrosis, inflammation, and<br />

repair of necrotic tissue, all of which result in<br />

changes in [ 18 F]-FDG uptake. Together, these<br />

experiments indicate that [ 18 F]-FDG-PET data<br />

should be carefully considered at early timepoints<br />

after VSV therapy, as changes in tracer<br />

uptake can be easily misinterpreted. This data<br />

strongly supports the rationale <strong>for</strong> multimodal<br />

imaging to assess tumor responses to oncolytic<br />

viral therapy.<br />

Session: Cancer <strong>Gene</strong> <strong>Therapy</strong><br />

P 46<br />

Functional Analysis of Armed Oncolytic<br />

Adenoviruses <strong>for</strong> Nuclear Reporter <strong>Gene</strong><br />

Imaging<br />

Youlia Kostova 1 , Klaus Mantwill 1 , Katja Dumler 1 ,<br />

Anja Wolf 1 , Hans-Jürgen Wester 2 , Bernd<br />

Gansbacher 1 , Per S Holm 1 , Martina Anton 1<br />

1 Klinikum rechts der Isar der TUM, Institut für<br />

Experimentelle Onkologie und<br />

Therapie<strong>for</strong>schung, <strong>Munich</strong>, <strong>German</strong>y; 2 Klinikum<br />

rechts der Isar der TUM, Nuklearmedizinische<br />

Klinik und Poliklinik, <strong>Munich</strong>, <strong>German</strong>y<br />

Oncolytic adenoviruses (AdV) are recently<br />

gaining interest in gene therapy, as they have the<br />

ability to lyse tumour cells. Hence, we<br />

investigated whether a YB-1-dependent oncolytic<br />

AdV armed with the reporter and suicide gene<br />

herpes simplex virus thymidine kinase (HSV1sr39tk)<br />

displays an enhanced tumour cell killing<br />

through the combined action of oncolysis and<br />

HSV1-sr39tk-Ganciclovir (GCV) mediated<br />

cytotoxicity in human U87MG glioma cells, and if<br />

this AdV is suitable <strong>for</strong> in vivo imaging studies. As<br />

a nuclear reporter gene HSV1-sr39tk is favoured<br />

over the wild-type <strong>for</strong>m because of its improved<br />

in vivo imaging properties. YB-1-dependent<br />

oncolytic AdV have so far not been combined<br />

with suicide genes in attempt to boost their<br />

therapeutic effect. U87MG cells were infected<br />

with armed oncolytic AdV expressing HSV1sr39tk<br />

in the E3 region and control AdV under<br />

varying conditions. DNA, RNA and protein levels<br />

of HSV1-sr39tk were determined by Southern<br />

blot, qRT-PCR and Western blot analyses,<br />

respectively. The oncolytic and cytotoxic effects<br />

of the virus were analysed by Sul<strong>for</strong>hodamine B<br />

staining or XTT assay and compared to the<br />

activity of other oncolytic and replication-deficient<br />

AdV. The enzymatic function of HSV1-sr39tk was<br />

measured by uptake of the [18F]-FHBG, a<br />

radiolabelled prodrug-analogue, 2 days after<br />

infection. Increased AdV DNA and HSV1-sr39TK<br />

DNA and protein levels over time were observed.<br />

The virus displayed an enhanced overlapping<br />

oncolytic and HSV1-sr39tk-GCV-mediated<br />

cytotoxic effect (>95%) in glioma cells, even at a<br />

low viral dosage and at GCV concentrations < 0.1<br />

μg/ml. The strongest cell killing was achieved if<br />

GCV was applied 2–3 days after infection. We<br />

also observed an increased bystander effect<br />

when infected cells were mixed with uninfected<br />

cells. Furthermore, HSV1-sr39TK displayed<br />

enhanced accumulation of radioactive tracer as<br />

compared to control AdV expressing HSV1sr39tk<br />

in the E3 region. The results of this study<br />

indicate that the HSV1-sr39TK expressing<br />

oncolytic adenovirus effectively induces tumour<br />

cell killing and radiotracer accumulation ([ 18 F]-<br />

FHBG) and might allow <strong>for</strong> in vivo PET imaging<br />

within a limited time frame.<br />

Session: Cancer <strong>Gene</strong> <strong>Therapy</strong><br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong>


P 47<br />

Radioiodine <strong>Therapy</strong> of HCC Following<br />

Systemic Sodium Iodide Symporter (NIS)<br />

<strong>Gene</strong> Transfer Using EGF-Receptor Targeted<br />

Nonviral <strong>Gene</strong> Delivery Vectors<br />

Kathrin Klutz 1 , David Schaffert 2 , Geoffrey k<br />

Grünwald 1 , Michael J Willhauck 1 , Wolfgang<br />

Rödl 2 , Nathalie Wunderlich 1 , Christian Zach 3 ,<br />

Franz Josef Gildehaus 3 , Reingard Senekowitsch-<br />

Schmidtke 4 , Ernst Wagner 2 , Burkhard Göke 1 ,<br />

Manfred Ogris 2 , Christine Spitzweg 1<br />

1Department of Internal Medicine II, Klinikum<br />

Großhadern, Ludwig-Maximilians-University<br />

<strong>Munich</strong>, <strong>Munich</strong>, <strong>German</strong>y; 2 Pharmaceutical<br />

Biotechnology, Department of Pharmacy,<br />

Ludwig-Maximilians-University, <strong>Munich</strong>,<br />

<strong>German</strong>y; 3 Department of Nuclear Medicine,<br />

Klinikum Groβhadern, Ludwig-Maximilians-<br />

University <strong>Munich</strong>, <strong>Munich</strong>, <strong>German</strong>y;<br />

4 Department of Nuclear Medicine, Technical<br />

University, <strong>Munich</strong>, <strong>Munich</strong>, <strong>German</strong>y<br />

We have recently demonstrated induction of<br />

significant tumor-selective uptake and therapeutic<br />

efficacy of radioiodine in neuroblastoma tumors<br />

after systemic nonviral polyplex-mediated NIS<br />

gene delivery. The aim of the current study was<br />

to evaluate the efficacy of novel nanoparticle<br />

vectors based on linear polyethylenimine (LPEI),<br />

shielded by polyethylene glycol (PEG), and<br />

coupled with the synthetic peptide GE11 as an<br />

epidermal growth factor (EGF) receptor-specific<br />

ligand <strong>for</strong> targeting the NIS gene to EGFRexpressing<br />

human HCC (Huh7) cells. We used<br />

LPEI-PEG-GE11 to <strong>for</strong>m targeted polyplexes with<br />

a NIS-expressing plasmid (pCpG-EF1-NIS) to<br />

transfect Huh7 cells followed by analysis of<br />

functional NIS expression in vitro and in vivo. In<br />

vitro incubation of Huh7 cells with NIS-conjugated<br />

LPEI-PEG-GE11 resulted in a 22-fold increase in<br />

iodide uptake activity. After establishment of<br />

subcutaneous Huh7 tumors in nude mice, NISconjugated<br />

nanoparticle vectors or control<br />

vectors were injected via the tail vein followed by<br />

analysis of radioiodine biodistribution after i.p.<br />

injection of 123 I using γ-camera imaging and ex<br />

vivo γ-counting. While injection of control vectors<br />

did not result in tumoral iodide accumulation,<br />

Huh7 tumors showed a perchlorate-sensitive<br />

iodide uptake of 6–9 % ID/g 123 I with an eff. halflife<br />

of approx. 6 h. In contrast, non-target organs<br />

like liver, lungs and kidneys showed no<br />

significant iodide uptake activity. After application<br />

of the EGFR-specific antibody cetuximab 24 h<br />

prior to administration of NIS-conjugated LPEI-<br />

PEG-GE11 tumoral iodide uptake activity and<br />

NIS mRNA expression were markedly reduced<br />

confirming the specificity of EGFR-targeted<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong><br />

| 66<br />

nanoparticle vectors. After 4 cycles of polymer<br />

application followed by therapeutic application of<br />

131 I, tumor growth was significantly reduced as<br />

compared to control groups. In conclusion, these<br />

results clearly demonstrate that systemic in vivo<br />

NIS gene transfer using novel synthetic<br />

nanoparticle vectors coupled with an EGFRtargeting<br />

ligand is capable of inducing tumorspecific<br />

iodide uptake, which represents a<br />

promising innovative strategy <strong>for</strong> systemic NIS<br />

gene therapy in metastatic cancers.<br />

Session: Cancer <strong>Gene</strong> <strong>Therapy</strong><br />

P 48<br />

MSC-Mediated sTRAIL and CXCR4 Delivery in<br />

Combination with XIAP Knockdown in<br />

Pancreatic Cancer Cells Leads to Inhibition of<br />

Metastatic Growth of Pancreatic Carcinoma<br />

Sina Rupprecht 1 , Andrea Mohr 1 , Stella<br />

Albarenque 1 , Rui Yu 1 , Laura Deedigan 1 , Mairead<br />

Reidy 1 , Simone Fulda 2 , Ralf Zwacka 2<br />

1National Centre <strong>for</strong> Biomedical Engineering<br />

Science, Molecular Therapeutics Group, National<br />

University of Ireland, Galway, Ireland; 2 Children's<br />

Hospital Ulm, Ulm University, Ulm, <strong>German</strong>y<br />

Disseminating tumour cells appear to be one of<br />

the biggest problems in oncological medicine.<br />

Here, we combined the tumour-specific<br />

apoptosis-inducing activity of TRAIL with the<br />

ability of mesenchymal stem cells (MSCs) to<br />

infiltrate both tumour and lymphatic tissues.<br />

Using this approach it was our aim to target<br />

primary tumours as well as disseminated cancer<br />

cells in a human pancreatic cancer mouse model.<br />

Targeting XIAP by applying RNAi inside the<br />

cancer cells further optimized this approach,<br />

because it is known that knocking down XIAP<br />

has an apoptosis sensitizing and anti-metastatic<br />

effect on targeted cancer cells. Furthermore, it<br />

has recently been published that the chemokine<br />

receptor CXCR4 and its ligand, the stromal-cellderived<br />

factor (SDF-1), enhanced the migratory<br />

abilities of MSCs, following the tumour-derived<br />

SDF-1 chemotactic axis. We generated MSCs<br />

expressing a trimeric soluble wild-type TRAIL<br />

(MSC.sTRAIL). MSC.sTRAIL triggered limited<br />

apoptosis in pancreatic cancer cells that were<br />

resistant to soluble recombinant TRAIL, which is<br />

most likely due to the enhanced effect of the<br />

direct, cell-mediated delivery of trimeric TRAIL.<br />

MSC.sTRAIL-mediated cell death was markedly<br />

increased by concomitant knockdown of XIAP by<br />

RNAi in the cancer cells. These findings were


67 |<br />

confirmed in xenograft models. Using<br />

MSC.sTRAIL on XIAP silenced xenografts it was<br />

possible to <strong>for</strong>ce the primary tumour to go into<br />

remission and to block the growth of lung<br />

metastasis in MSC.sTRAIL treated animals.<br />

Furthermore, to enhance the directed migration<br />

of sTRAIL-loaded MSCs towards tumour cells,<br />

these cells were adenovirally transducted to<br />

overexpress the chemokine receptor CXCR4.<br />

The natural secretion of SDF-1 by tumour cells<br />

builds up a chemotactic axis and it could be<br />

demonstrated that CXCR4 overexpressing<br />

MSC.sTRAIL utilize this gradient <strong>for</strong> enhanced<br />

migration towards the tumour cells. In summary,<br />

this is the first demonstration that a combined<br />

approach using systemic MSC-mediated delivery<br />

of sTRAIL and overexpression of CXCR4<br />

together with XIAP inhibition suppresses<br />

metastatic growth of pancreatic carcinoma.<br />

Session: Tumor Biology and (Cancer) Stem Cells<br />

P 49<br />

YB-1 Dependent Oncolytic Adenovirus as a<br />

Strategy <strong>for</strong> the Treatment of Glioma Cancer<br />

Stem Cells<br />

Ulrike Naumann 1 , Per Sonne Holm 2 , Klaus<br />

Mantwill 2 , Janina Seznec 1<br />

1Hertie Institute <strong>for</strong> Clinical Brain Research and<br />

Center Neurology, Laboratory <strong>for</strong> Molecular<br />

Neuro-Oncology, Tübingen, <strong>German</strong>y; 2 Institute<br />

of Experimental Oncology and <strong>Therapy</strong><br />

Research, Klinikum rechts der Isar, TU <strong>Munich</strong>,<br />

<strong>Munich</strong>, <strong>German</strong>y<br />

Recent studies have demonstrated the existence<br />

of a small fraction of glioma cells endowed with<br />

features of primitive progenitor cells and tumorinitiating<br />

function. Such cells have been defined<br />

as glioma-like cancer stem cells (GCSC). GCCS<br />

contribute to glioma progression due o their<br />

properties to overcome cell death induced by<br />

chemo- and radiation-therapy, to self-renew and<br />

to promote angiogenesis. For this reason, GCSC<br />

might never be completely eradicated using<br />

standard therapy and will be responsible <strong>for</strong> the<br />

recurrence of glioma. It has been shown that the<br />

Y-box protein YB-1 is an oncogenic<br />

transcription/translation factor associated with the<br />

development of cancer. By upregulation of the<br />

detoxification membrane channel protein MRP<br />

and MDR, YB-1 induces drug resistance. Again,<br />

YB-1 itself is regulated via the stem cell-specific<br />

transcription factors SLUG, SNAIL and TWIST.<br />

Third, YB-1 is overexpressed in GCSC, but is<br />

barely detectable in normal human stem cells.<br />

Fourth, YB-1 plays an important role in the<br />

adenovirus half-life by targeting the adenoviral<br />

E2-late promoter. This knowledge promoted us to<br />

develop an YB-1 based glioma virotherapy. We<br />

investigated the therapeutic potential of the YB-1<br />

dependent oncolytic adenovirus Ad-Delo3-RGD<br />

in GCSC. We demonstrated that in vitro Ad-<br />

Delo3-RGD replicates in GCSC, kills highly<br />

temozolomide (TMZ)-resistant GCSC and<br />

moderately rendered these cells susceptible<br />

towards TMZ-treatment. In vivo, using an<br />

orthotopic mouse xenograft glioma model,<br />

infection of GCSC-derived tumors with Ad-Delo3-<br />

RGD significantly prolonged survival. Taken<br />

together all data, an YB-1 based adenovirotherapy<br />

might be a promising treatment<br />

strategy against glioma.<br />

Session: Tumor Biology and (Cancer) Stem Cells<br />

P 50<br />

Tet-Regulated, Lentivirally Mediated BMP-2<br />

Expression in Primary Cells<br />

Daniela Wübbenhorst, Katja Dumler, Gabriele<br />

Wexel, Andreas Imhoff, Bernd Gansbacher,<br />

Stephan Vogt, Martina Anton<br />

Institute of Experimental Oncology and <strong>Therapy</strong><br />

Research, Klinikum rechts der Isar, TU <strong>Munich</strong>,<br />

<strong>Munich</strong>, <strong>German</strong>y<br />

<strong>Therapy</strong> of cartilage defects is challenging due to<br />

poor self-healing capacity. A combined gene and<br />

cell therapeutic approach using transduced<br />

primary chondrocytes and pluripotent<br />

mesenchymal stem cells (MSC) was chosen.<br />

MSC are able to differentiate into a variety of cell<br />

types among them osteocytes and chondrocytes<br />

and are there<strong>for</strong>e considered <strong>for</strong> the treatment of<br />

osteochondral defects. Regulated gene<br />

expression of growth factor bone morphogenetic<br />

protein 2 (BMP-2) was achieved using the Tet-on<br />

System, delivered by VSV-G pseudotyped<br />

lentiviral SIN vectors (LV). Primary rabbit<br />

chondrocytes were infected with 1-vector Tet-on<br />

constructs, expressing the reverse Transactivator<br />

(rtTA) and eGFP or BMP 2 under control of the<br />

Tet-responsive element (TRE). Transgene<br />

expression was induced by doxycycline (dox).<br />

The efficacy of the Tet-on induction system was<br />

optimized using eGFP. Using BMP-2-expressing<br />

vectors resulted in secretion of 15–16 ng/ml<br />

BMP-2 into the medium. After withdrawal of dox<br />

BMP-2 expression decreased to background<br />

levels and was re-inducible <strong>for</strong> various cycles<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong>


without loss of efficacy. Additionally, prolonged<br />

tet-regulated BMP-2 expression was feasible.<br />

Produced BMP-2 was functional as evidenced by<br />

proteoglycan synthesis. Transduction efficiency<br />

of rabbit bone marrow derived MSC by eGFP<br />

expressing LV was 65%. After lentiviral infection<br />

cells retained full differentiation potential.<br />

Transduction of MSC with the BMP-2 Tet-on<br />

vectors resulted in inducible BMP-2 expression<br />

with a 36-fold increase of BMP-2 expression on<br />

RNA level as well as a 113-fold increase on<br />

protein level compared to uninduced cells. The<br />

lentivirally delivered Tet-on System allows <strong>for</strong><br />

regulated expression of BMP-2 in primary rabbit<br />

chondrocytes as well as MSCs. The amount of<br />

BMP-2 produced by chondrocytes after induction<br />

in vitro is sufficient <strong>for</strong> proteoglycan synthesis, a<br />

marker <strong>for</strong> chondrogenesis and levels are<br />

comparable to results achieved using a<br />

constitutively expressing retroviral vector.<br />

Session: Tumor Biology and (Cancer) Stem Cells<br />

P 51<br />

Neutrality and Non-Neutrality in Clonal<br />

Development<br />

Lars Thielecke, Ingmar Glauche, Sebastian<br />

Gerdes, Ingo Roeder<br />

Institut für Medizinische In<strong>for</strong>matik und Biometrie,<br />

TU-Dresden, Dresden, <strong>German</strong>y<br />

Tissue regeneration and maintenance are<br />

generally driven by a population of stem cells.<br />

These cells are considered to have closely<br />

similar functional capabilities and contribute to<br />

the production of mature somatic cells by<br />

differentiation and continuous proliferation. Clonal<br />

marking studies in the hematopoietic system<br />

revealed that the peripheral blood is generally<br />

composed of the ancestry of multiple<br />

hematopoietic stem cells at each point in time.<br />

However, in certain pathological situations (i.e., in<br />

leukemias) this balance is upset and the system<br />

converts to a monoclonal, often malign situation.<br />

Similar results are confirmed in vivo and in vitro<br />

using a wide range of viral markers. However, the<br />

general questions remain how this clonal<br />

dominance is generated and how it can be<br />

potentially detected at early stages. Using a<br />

range of mathematical modeling approaches we<br />

characterize the temporal development of<br />

multiple clones under different assumptions<br />

about the relative fitness. In particular we use<br />

these models to study the case of neutral<br />

competition <strong>for</strong> certain in vitro and in vivo settings<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong><br />

| 68<br />

and extend our studies to the characterization of<br />

clonal dominance. Based on our model<br />

simulations we make predictions about the<br />

variability of clone sizes as a function of time <strong>for</strong><br />

the situation of neutral and non-neutral clonal<br />

competition. Our results suggest that clonal<br />

conversion is inevitable even in the neutral<br />

situation and is only accelerated if dominating<br />

clones exist. We furthermore suggest<br />

experimental strategies <strong>for</strong> a quantitative<br />

characterization of the conversion process.<br />

Session: Tumor Biology and (Cancer) Stem Cells<br />

P 52<br />

Efficient Zinc-Finger Nucleases-Mediated<br />

<strong>Gene</strong> Knockout in Pluripotent Stem Cells<br />

Anna Osiak 1 , Frank Radecke 2 , Eva Guhl 1 , Sarah<br />

Radecke 2 , Kafaitullah Khan 3 , Fabienne Lütge 3 ,<br />

Tobias Cantz 4 , Regine Heilbronn 1 , Klaus<br />

Schwarz 2 , Toni Cathomen 3<br />

1 Institute of Virology (CBF), Charité Medical<br />

School, Berlin, <strong>German</strong>y; 2 Institute <strong>for</strong><br />

Transfusion Medicine, University of Ulm, Ulm,<br />

<strong>German</strong>y; 3 Department of Experimental<br />

Hematology, Hannover Medical School,<br />

Hannover, <strong>German</strong>y; 4 REBIRTH Cluster of<br />

Excellence, JRG Group Stem Cell Biology,<br />

Hannover Medical School, Hannover, <strong>German</strong>y<br />

Murine embryonic stem cells (mESCs) have been<br />

an excellent model system <strong>for</strong> introducing<br />

specific modifications into the genome to study<br />

gene function in vitro or to generate knockout<br />

mouse models. However, gene targeting in<br />

mESCs using standard techniques is quite<br />

inefficient and typically reaches frequencies of<br />

10-7 to 10-6. In consequence, the usage of<br />

complex positive/negative selection strategies to<br />

isolate gene targeted clones has been crucial. A<br />

promising strategy to introduce specific changes<br />

into complex genomes is based on zinc-finger<br />

nucleases (ZFNs). A ZFN subunit consists of a<br />

nonspecific endonuclease domain fused to a<br />

specific DNA-binding domain composed of<br />

engineered zinc-finger motifs that tether the<br />

nuclease domain to a preselected chromosomal<br />

site. Upon dimerization of two ZFN subunits at<br />

the target site, the ZFN pair specifically cleaves<br />

the DNA to trigger the ensuing DNA damage<br />

response, which can be harnessed <strong>for</strong> targeted<br />

genome engineering. Here, we used an EGFPpositive<br />

mESC line in order to explore the<br />

potential of ZFNs to generate a genetic knockout<br />

in the absence of antibiotic selection. EGFP-


69 |<br />

specific ZFNs carrying different previously<br />

described endonuclease variants were initially<br />

tested in an EGFP-positive human cell line and in<br />

EGFP + mESC to identify ZFN variants that<br />

combine high activity with minimal toxicity. Under<br />

optimized conditions, the single copy EGFP locus<br />

was disrupted in 9% of EGFP + mESC after<br />

lipofection of the ZFN expression vectors and 3%<br />

upon electroporation. Importantly, the percentage<br />

of knockout cells remained stable over time,<br />

suggesting minor ZFN-associated toxicity. For<br />

two independent clones the EGFP knockout<br />

status was verified on the genome level.<br />

Currently, these clones are being tested <strong>for</strong> their<br />

pluripotent status by a teratoma <strong>for</strong>mation assay.<br />

In the advent of applying patient-derived induced<br />

pluripotent stem cells in regenerative medicine,<br />

this approach serves as a paradigm <strong>for</strong> knocking<br />

out a dominant mutant allele with minimal<br />

genomic intervention and without the need of<br />

introducing an antibiotic selection marker.<br />

Session: Tumor Biology and (Cancer) Stem Cells<br />

P 53<br />

Optimizing Virus-Magnetic Nanoparticle<br />

Complexes <strong>for</strong> <strong>Gene</strong> Transfer in Cell Lines<br />

and Stem Cells<br />

Olga M Mykhaylyk 1 , Yolanda Sanchez-<br />

Antequera 1 , Markus Döblinger 2 , Stefan<br />

Thalhammer 3 , Per Sonne Holm 1 , Christian Plank 1<br />

1Institute of Experimental Oncology and <strong>Therapy</strong><br />

Research, Klinikum rechts der Isar, TU <strong>Munich</strong>,<br />

<strong>Munich</strong>, <strong>German</strong>y; 2 Department of Chemistry and<br />

Biochemistry, Ludwig-Maximilians-Universität<br />

München, <strong>Munich</strong>, <strong>German</strong>y; 3 Helmholtz Zentrum<br />

München, Institute of Radiation Protection,<br />

NanoAnalytics, <strong>Munich</strong>, <strong>German</strong>y<br />

The goal of this work was providing magnetic<br />

nanoparticles (MNPs) and their <strong>for</strong>mulations <strong>for</strong><br />

efficient viral genetic modification of cell lines and<br />

primary cells in vitro and ex vivo. MNPs of the<br />

core-shell type with magnetite cores of about 10<br />

nm stabilized and decorated with surfactants and<br />

charged polymers were selected from our library<br />

of in-house synthesized MNPs <strong>for</strong> self-assembly<br />

with viral particles. To achieve maximal vector<br />

binding magnetic vectors were optimized with<br />

account <strong>for</strong> the vector association and magnetic<br />

sedimentation with MNPs. Measuring the time<br />

course of the turbidity of suspensions of the<br />

virus-MNP complexes in defined magnetic fields<br />

was used <strong>for</strong> the evaluation of the<br />

magnetophoretic mobility. We have learned that<br />

<strong>for</strong> magnetic viral vectors, it is reasonable to<br />

express the composition in terms of iron weight<br />

per PHYSICAL virus particle, and NOT per<br />

infectious virus particle, taking into account that<br />

both infectious and non-infectious virus particles<br />

are associated with appropriate MNPs. We<br />

suggest a “rule” to <strong>for</strong>mulate virus magnetic<br />

complexes with suitable MNPs, based on the<br />

association and magnetic sedimentation of the<br />

virus with MNPs as well as with account <strong>for</strong> the<br />

functionality of gene delivery. The optimal<br />

complex composition of 2.5-20 fg iron per<br />

physical virus particle, depending on the MNPs<br />

used, is applicable to both adenoviral and<br />

lentiviral vectors. Optimized magnetic virus<br />

complexes were stable in 50% FCS. Electron and<br />

atomic <strong>for</strong>ce microscopy data showed structurally<br />

intact viruses surrounded by multiple MNPs.<br />

<strong>Gene</strong>tic modification of cells on a cell separation<br />

column modified with <strong>for</strong>mulated magnetic viral<br />

vectors (magselectofection) is highly efficient in<br />

hematopoietic stem cells and mesenchymal stem<br />

cells from human umbilical cord, hUC-HSCs and<br />

hUC-MSCs, respectively. Under optimized<br />

transduction conditions, viral magselectofection<br />

of hUC-MSCs with SO-Mag2 lentivirus<br />

complexes at MOI as low as 0.5 pfu/cell resulted<br />

in 60-100% transduced cells. We have also found<br />

a two-fold increase in the percentage of the<br />

reporter gene expressing cells postmagselectofection<br />

of the hUC-MSCs prelabelled<br />

with 20 pg Fe/cell of MNPs.<br />

Session: Tumor Biology and (Cancer) Stem Cells<br />

P 54<br />

T Cell-Based Immunotherapy <strong>for</strong> Renal Cell<br />

Carcinoma<br />

Matthias Leisegang 1 , Adriana Turqueti-Neves 2 ,<br />

Boris Engels 1 , Thomas Blankenstein 1 , Dolores J<br />

Schendel 2 , Elfriede Nößner 2 , Wolfgang Uckert 1<br />

1 Max-Delbrück-Center <strong>for</strong> Molecular Medicine,<br />

Berlin, <strong>German</strong>y; 2 Helmholtz Zentrum München,<br />

<strong>Munich</strong>, <strong>German</strong>y<br />

Adoptive therapy with genetically engineered T<br />

cells carrying redirected antigen specificity (TCR<br />

gene therapy) is a new option to treat cancer,<br />

especially melanoma. However, this treatment is<br />

not yet available <strong>for</strong> metastatic renal cell<br />

carcinoma (RCC), due to the scarcity of<br />

therapeutically useful reagents. We analyzed<br />

tumor-infiltrating lymphocytes (TIL) from RCC to<br />

identify T cells with shared tumor-specific<br />

recognition <strong>for</strong> the generation of T cell receptor<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong>


(TCR)-engineered T lymphocytes <strong>for</strong> TCR gene<br />

therapy of RCC. We established a T cell clone<br />

from TIL that recognized an HLA-A2-restricted<br />

tumor antigen. The TCRalpha- and beta-chain<br />

genes (TCR53) were isolated, modified by codon<br />

optimization and murinization, and retrovirally<br />

transduced into peripheral blood lymphocytes<br />

(PBL). TCR53-engineered PBL recapitulated the<br />

specificity of the TIL and demonstrated tumorspecific,<br />

HLA-A2-restricted effector activites (IFNγ,<br />

TNF-α, IL-2, MIP-1β, cytotoxicity). TCR53engineered<br />

PBL of healthy donors and RCCpatients<br />

exhibited similar TCR expression levels,<br />

expansion, and polyfunctional profile. Based on<br />

murine B3Z cells, a TCR53-expressing indicator<br />

line (B3Z-TCR53) was established <strong>for</strong> the<br />

screening of the antigen prevalence in RCC, in<br />

other malignancies, and in normal cell<br />

counterparts. Using B3Z-TCR53 cells, 130 tumor<br />

and normal cells were analyzed and shared<br />

TCR53 peptide:MHC expression was found in<br />

more than 60% of RCC and 25% of tumor cell<br />

lines of other histology, while normal tissue cells<br />

were not recognized. To date, TCR53 is the only<br />

TCR with shared HLA-A2-restricted recognition of<br />

RCC. It fulfills important criteria <strong>for</strong> utilization in<br />

TCR gene therapy and could advance T cellbased<br />

immunotherapy to patients with RCC and<br />

other malignancies expressing the TCR-ligand.<br />

Session: Cancer Immune <strong>Therapy</strong> and T-Cell<br />

<strong>Therapy</strong><br />

P 55<br />

Novel Immunostimulatory <strong>Therapy</strong> by<br />

Adjuvant Magnetofection Prolongs Relapse-<br />

Free Survival of Fibrosarcoma Bearing Cats:<br />

A Veterinary Clinical Study<br />

Ulrike Schillinger 1 , Miriam Rutz 2 , Cornelia<br />

Fischer 2 , Anika Jahnke Jahnke 2 , Florian Walsch 2 ,<br />

Mehrije Ferizi 1 , Veronika Benda 1 , Johannes<br />

Hirschberger 2 , Roberto Köstlin 3 , Bernd<br />

Gänsbacher 1 , Thomas Brill 1 , Christian Plank 1<br />

1 Institute of Experimental Oncology and <strong>Therapy</strong><br />

Research, Klinikum rechts der Isar, TU <strong>Munich</strong>,<br />

<strong>Munich</strong>, <strong>German</strong>y; 2 Department of Small Animal<br />

Medicine, Ludwig-Maximilians-University <strong>Munich</strong>,<br />

<strong>Munich</strong>, <strong>German</strong>y; 3 Department of Small Animal<br />

Surgery and Reproduction, Ludwig-Maximilians-<br />

University <strong>Munich</strong>, <strong>Munich</strong>, <strong>German</strong>y<br />

Feline fibrosarcoma is an everyday challenge in<br />

veterinary practice. Despite aggressive pre or<br />

post-operative treatment it has a high relapse<br />

rate of aprox. 75 % within 6 months after surgical<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong><br />

| 70<br />

resection. To obtain a better therapeutic<br />

outcome, novel strategies are necessary. Hence,<br />

we established immunostimulatory therapy by<br />

magnetofection. Here we report preliminary<br />

results from a comparative clinical study where<br />

the genes <strong>for</strong> feline GM-CSF, IFN-γ and IL-2,<br />

feline GM-CSF alone or human GM-CSF were<br />

administered. The study design is prospective,<br />

randomized, placebo-controlled ( = standard<br />

therapy) and includes four arms: (1) standard<br />

therapy, i.e., surgery alone; (2) nonviral<br />

magnetofection of the triple-combination of feline<br />

GM-CSF, IFN-γ and IL-2 genes into the tumor<br />

be<strong>for</strong>e surgery or nonviral administration by<br />

magnetofection of feline (3) or human GM-CSF<br />

(4) gene alone. Preliminary clinical endpoints of<br />

the studies are relapse-free survival. Nonviral<br />

magnetofection, a procedure developed in our<br />

laboratory, is gene therapy by plasmids<br />

associated with magnetic nanoparticles under the<br />

influence of a magnetic field. The magnetic field<br />

was applied to achieve improved retention of the<br />

injected vector dose in the tumor. All genetherapeutic<br />

treatments were well tolerated and<br />

led to significantly prolonged relapse-free<br />

survival. Recent results from FACS-analysis of<br />

the primary tumor cells from the treated groups<br />

showed increased MHC-II expression compared<br />

to those of control cats. This is encouraging<br />

concerning future use in veterinary practice as<br />

this treatment can be easily administered. The<br />

results are promising with respect to their<br />

potential in human medicine, as they have been<br />

obtained in real patients instead of experimental<br />

tumor models.<br />

Session: Cancer Immune <strong>Therapy</strong> and T-Cell<br />

<strong>Therapy</strong>


71 |<br />

P 56<br />

Targeting the Epidermal Growth Factor<br />

Receptor (HER) Family by T Cell Receptor<br />

<strong>Gene</strong>-Modified T Lymphocytes<br />

Peter Meyerhuber 1 , Heinke Conrad 2 , Lilian<br />

Stärck 1 , Matthias Leisegang 1 , Dirk H. Busch 3 ,<br />

Helga Bernhard 4 , Wolfgang Uckert 1<br />

1Max-Delbrück-Center <strong>for</strong> Molecular Medicine,<br />

Berlin, <strong>German</strong>y; 2 Department of<br />

Hematology/Oncology, Technical University of<br />

<strong>Munich</strong>, Klinikum rechts der Isar, <strong>Munich</strong>,<br />

<strong>German</strong>y; 3 Institute of Microbiology, Immunology<br />

and Hygiene, Technical University of <strong>Munich</strong>,<br />

<strong>Munich</strong>, <strong>German</strong>y; 4 Department of<br />

Hematology/Oncology, Klinikum Darmstadt,<br />

Darmstadt, <strong>German</strong>y<br />

HER2 is over-expressed in 25–40% of all breast<br />

cancers and in a variety of other tumors such as<br />

ovarian cancer and hematological malignancies.<br />

Due to the selective over-expression in malignant<br />

tissue, HER2 is considered one of the most<br />

attractive targets <strong>for</strong> therapeutic interventions,<br />

such as monoclonal antibodies, kinase inhibitors,<br />

and cancer vaccines. However, tumor regression<br />

following immunization with vaccines is rare,<br />

likely a consequence of HER2 being a self<br />

antigen and there<strong>for</strong>e inducing tolerance. To<br />

overcome tolerance, adoptive T cell transfer<br />

(ATT) strategies have been developed. However,<br />

clinical application of ATT is limited, because<br />

isolation and characterization of HER2-reactive T<br />

cells is laborious. Yet, a promising approach is<br />

the transfer of TCRαβ genes, which have been<br />

isolated from non-tolerant settings, into T cells. In<br />

this study, we isolated the TCR genes of a<br />

HER2-reactive, allo-HLA-A2-restricted CTL clone<br />

and introduced the genes into a retroviral vector.<br />

Efficient cell surface expression of the TCR and<br />

improved functional avidity of the gene-modified<br />

T cells were achieved after murinization<br />

(replacement of the human TCR constant regions<br />

by mouse counterparts), codon-optimization and<br />

application of the P2A gene linker (HER2-TCRopt).<br />

Thus, the TCR expression in transduced T<br />

cells increased from 1.5% to 41% measured by<br />

A2/HER2-multimer staining. The ability to secrete<br />

IFN-γ of HER2-TCR-opt transduced T cells was<br />

comparable to that of the CTL clone, which<br />

showed a half maximum IFN-γ secretion at 10-7<br />

M towards HER2 peptide-loaded T2 cells.<br />

Furthermore, HER2-TCR-opt-transduced T cells<br />

lysed HER2-expressing tumor cell lines as<br />

efficient as the parental clone (lysis of 58% at a<br />

E:T of 30:1). The TCR showed a cross-reactivity<br />

to HER3 and HER4 that was similar to the<br />

parental CTL clone. Extensive testing with<br />

HER2/3/4 negative cell lines did not reveal any<br />

further epitopes that were recognized by the<br />

TCR. Our results contribute to the development<br />

of a TCR-based approach <strong>for</strong> the treatment of<br />

HER2-positive breast cancer, as well as of other<br />

malignancies expressing HER2, HER3 and/or<br />

HER4.<br />

Session: Cancer Immune <strong>Therapy</strong> and T-Cell<br />

<strong>Therapy</strong><br />

P 57<br />

Humanized Mouse Models <strong>for</strong> the Pathogenic<br />

Analysis of T-Cell Leukemia/Lymphoma<br />

Development<br />

Benjamin Rengstl 1 , Sebastian Newrzela 1 , Sylvia<br />

Hartmann 1 , Alexander Benz 1 , Tim Heinrich 1 ,<br />

Martin-Leo Hansmann 1 , Dorothee von Laer 2<br />

1<br />

Department of Pathology, University of<br />

Frankfurt, Frankfurt am Main, <strong>German</strong>y;<br />

2<br />

Department of Virology, University of Innsbruck,<br />

Innsbruck, Austria<br />

The oncogenic potential of human hematopoietic<br />

stem cells (HSCs) in the context of retroviral<br />

gene therapy, as reported in patients treated <strong>for</strong><br />

X-linked severe combined immunodeficiency (X-<br />

SCID) or X-linked chronic granulomatous disease<br />

(X-CGD), is now well described. Syngeneic<br />

mouse models <strong>for</strong> the development of<br />

leukemia/lymphoma are established as well as<br />

the reconstitution of immunodeficient mice with<br />

human stem cells. The combination of both<br />

systems, a mouse model <strong>for</strong> the development of<br />

human leukemia/lymphoma is still missing. To<br />

address this issue, we aim to test different<br />

humanized mouse models. Currently, the most<br />

promising mouse strains available <strong>for</strong> the<br />

engraftment of human cells are NOD-SCID γc −/−<br />

(NOG) and Rag2 −/− γc −/− mice. These strains will<br />

be repopulated with human HSCs or mature T<br />

cells expressing potent (proto-)oncogenes after<br />

retroviral/lentiviral gene transfer. To further<br />

enhance the repopulation dynamics of mature T<br />

cells, human HSCs will be transduced with<br />

mouse strain-specific murine T-cell receptors<br />

(TCRs). After successful repopulation the strainadapted<br />

human, mature T cells will be isolated, in<br />

vitro transduced with potent (proto-)oncogenes<br />

and transplanted into secondary recipients. A<br />

second strategy to overcome the mismatch<br />

between the human TCR and the murine major<br />

histocompatibility complex (MHC) is to utilize<br />

huMHC-transgene mice. Due to the lack of<br />

functional lymph nodes in the immunodeficient<br />

mouse strains in a third strategy we want to use<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong>


the so-called trimera mouse model: lethally<br />

irradiated and cyclophosphamid treated<br />

immunocompetent mice will be reconstituted with<br />

bone marrow of highly immunodeficient mice to<br />

generate an optimal environment <strong>for</strong> the<br />

repopulation with retrovirally/lentivirally modified<br />

human, mature lymphocytes. Moreover, all<br />

described mouse models will be used to<br />

investigate established human<br />

leukemia/lymphoma cell lines in vivo. The future<br />

goals of the project are on the one hand to<br />

compare expression profiles of healthy and<br />

neoplastic lymphocytes and on the other hand<br />

the identification of leukemic stem cells (LSCs) of<br />

leukemia/lymphoma cell lines.<br />

Session: Cancer Immune <strong>Therapy</strong> and T-Cell<br />

<strong>Therapy</strong><br />

P 58<br />

Clonal Dynamics Within Mature T-Cell<br />

Populations in the Preleukemic Phase of T-<br />

Cell Leukemia/Lymphoma<br />

Tim Heinrich 1 , Dorothee von Laer 2 , Sebastian<br />

Newrzela 1 , Benjamin Rengstl 1 , Sylvia Hartmann 1 ,<br />

Martin-Leo Hansmann 1<br />

1 Department of Pathology, University of<br />

Frankfurt, Frankfurt am Main, <strong>German</strong>y; 2 Medical<br />

University of Innsbruck, Department of Virology,<br />

Innsbruck, Austria<br />

Retroviral insertional mutagenesis in<br />

hematopoietic progenitor cells can activate<br />

neighboring proto-oncogenes and thus contribute<br />

to leukemia development. This genotoxic activity<br />

of integrating viruses is highly relevant <strong>for</strong> the risk<br />

assessment of stem cell gene therapy and also<br />

has been the basis <strong>for</strong> the identification of several<br />

proto-oncogenes. In previous studies, we<br />

analyzed the susceptibility of mature T cells to<br />

trans<strong>for</strong>mation (Newrzela et al., Blood, 2008).<br />

Surprisingly, T-cell transplanted animals showed<br />

no sign of leukemia/lymphoma development<br />

during a follow-up of more than 500 days. These<br />

results show that polyclonal mature T cells are<br />

less susceptible to trans<strong>for</strong>mation in vivo by<br />

known T-cell oncogenes than progenitor cells. In<br />

contrast, TCR-monoclonal T lymphocytes<br />

expressing oncogenes developed T-cell<br />

leukemia/lymphoma in recipient mice. We thus<br />

postulate that clonal competition may control<br />

leukemogenesis in mature T-cell populations. To<br />

analyze the mechanisms that control the<br />

outgrowth of malignant clones in TCR polyclonal,<br />

but not in TCR monoclonal mice, clonal<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong><br />

| 72<br />

fluctuation within the T-cell population will be<br />

analyzed during the manifestation of mature Tcell<br />

leukemia/lymphoma. DNA samples from<br />

peripheral blood T cells will be frozen in regular<br />

intervals from mice transplanted with polyclonal<br />

or monoclonal T cells expressing specific (proto-<br />

)oncogenes or a control gene. As the<br />

gammaretroviral vector used <strong>for</strong> gene transfer<br />

integrates randomly into the host genome,<br />

individual T-cell clones are marked by their<br />

specific integration sites. Integration site analysis<br />

by LM-PCR will reveal dominant clones within the<br />

T-cell population. In order to visualize the clonal<br />

fluctuation and progressing dominance of certain<br />

clones with a growth advantage due to insertional<br />

mutagenesis, these T-cell clones will be<br />

investigated retrospectively by integration-site<br />

specific PCR. In addition, the TCR diversity in the<br />

polyclonal setting will be analyzed on a genomic<br />

level during leukemia/lymphoma<br />

development/suppression to correlate the<br />

homeostatic clonal competition and the outgrowth<br />

of malignant T-cell clones.<br />

Session: Cancer Immune <strong>Therapy</strong> and T-cell<br />

<strong>Therapy</strong><br />

P 59<br />

A Hexon Modification in hAd5-Based Vectors<br />

Results in an Increased Transduction Efficacy<br />

of Pancreatic Cancer and Stellate Cells<br />

Tanja Lucas 1 , Karim Benihoud 2 , Andreas<br />

Wortmann 3 , Max G Bachem 4 , Stefan Kochanek 1<br />

1Department of <strong>Gene</strong> <strong>Therapy</strong>, University of Ulm,<br />

Ulm, <strong>German</strong>y; 2 CNRS UMR 8121, Vectorology<br />

and <strong>Gene</strong> Transfer, Gustave Roussy Institute,<br />

Villejuif, France; 3 Tier<strong>for</strong>schungszentrum,<br />

University of Ulm, Ulm, <strong>German</strong>y; 4 Department of<br />

Clinical Chemistry, University of Ulm, Ulm,<br />

<strong>German</strong>y<br />

Pancreatic cancer is the fifth leading cause of<br />

cancer-related death in <strong>German</strong>y. Pancreatic<br />

adenocarcinomas (PDACs) are characterised by<br />

rapid progression, early metastasis, diagnosis at<br />

an advanced stage, and high resistance to<br />

standard therapy. Thus, patients suffering from<br />

PDACs have a very poor prognosis. PDACs are<br />

composed of infiltrating tumor cells surrounded<br />

by extracellular components and other nonneoplastic<br />

cell types. Very frequently, tumor cells<br />

can be acounted <strong>for</strong> significantly less than half of<br />

the cellular mass within the tumor. The complex<br />

mutual interaction between tumor cells and nonneoplastic<br />

human pancreatic stellate cells


73 |<br />

(hPSCs – the major stromal cell type present in<br />

pancreatic cancer) is highly regulated through<br />

secretion of different growth factors (e.g., TGF-β),<br />

strongly influencing growth and malignancy of<br />

PDACs. In mouse experiments coinjection of<br />

tumor cells and hPSCs resulted in an advanced<br />

tumor progression. For treatment of pancreatic<br />

cancer with adenoviral (Ad) vectors, it would be<br />

there<strong>for</strong>e of advantage to target these vectors to<br />

both cell types. To date the systemic<br />

admininstration of Ad vectors, however, is<br />

characterised by two major drawbacks: the low<br />

transduction of tumor cells and the severe<br />

damage of healthy tissues (e.g., liver). To<br />

address these obstacles a hexon-modified Ad<br />

vector has been developed displaying the TGF-β<br />

receptor binding peptide CSK17 on the<br />

hypervariable region 5 of hexon resulting in<br />

AdhCKS17. Subsequent in vitro studies have<br />

shown a significantly enhanced transduction of<br />

early tumor cells and primary hPSCs after<br />

infection with AdhCKS17. As a consequence the<br />

hexon-modification resulted in an increased burst<br />

size and cytotoxicity in both cell types. A<br />

preliminary biodistribution study indicates a<br />

decreased liver uptake of this hexon-modified<br />

vector compared to the control vector indicating<br />

detargeting from liver, thereby potentially<br />

reducing liver damage. Subsequent in vivo<br />

investigations are directed at analysing this<br />

vector within the human tumor in an ex vivo<br />

culture system and in murine xenotransplantation<br />

models bearing composite tumors consisting of<br />

human pancreatic cancer cells and stroma cells.<br />

Session: Pharmacology and Toxicology<br />

P 60<br />

Controlled Removal of a Nonviral Episome<br />

Claudia Hagedorn 1 , Sina Rupprecht 2 , Hans J.<br />

Lipps 2<br />

1 Universität Witten/Herdecke, Institut für<br />

Zellbiologie, Witten/Herdecke, <strong>German</strong>y; 2 Institut<br />

für Zellbiologie ZBAF – Zentrum für<br />

biomedizinische Ausbildung und Forschung<br />

Universität Witten/Herdecke, Witten/Herdecke,<br />

<strong>German</strong>y<br />

The vector pEPI is the first non-viral autonomous<br />

replicon that was constructed <strong>for</strong> mammalian<br />

cells. It represents a minimal model system to<br />

study the epigenetic regulation of replication and<br />

transcription. Its function relies on a transcription<br />

unit linked to an S/MAR sequence. In gene<br />

therapy, situations exist in which transgenes<br />

have to be expressed <strong>for</strong> a limited time and thus<br />

it would be of considerable interest not only to<br />

silence the expression of the transgene but to<br />

construct a vector which can be removed from<br />

the cell at any time. There<strong>for</strong>e we followed two<br />

approaches: (I) We constructed the inducible<br />

episomal vector pEPI-Tet in which transcription<br />

running into the S/MAR is regulable with<br />

doxycycline. We found that <strong>for</strong> vector replication<br />

and long-term maintenance an ongoing<br />

transcription running into the S/MAR element is<br />

required. Once established, the vector is lost<br />

from the cell upon switching off transcription from<br />

the gene linked to the S/MAR. (II) Secondly, we<br />

used the siRNA approach: Cells in which pEPIeGFP<br />

has been stably established were<br />

transfected with a vector expressing siRNA<br />

directed against eGFP. As a consequence, due<br />

to the abrogated transcription of the transgene<br />

running in to the S/MAR, reduced plasmid copies<br />

per cell were detected using quantitative PCR.<br />

Furthermore, when comparing cells transfected<br />

with an eGFP-siRNA with those transfected with<br />

a nonsense-siRNA, we found that active<br />

chromatin markers, e.g., H3K36me or H3K4me3,<br />

got lost.<br />

Session: Pharmacology and Toxicology<br />

P 61<br />

Transposition from a High-Capacity<br />

Adenoviral Vector: Optimization of the<br />

System and Mapping of Transposon<br />

Integration Sites in Murine Liver<br />

Wenli Zhang 1 , Martin Hausl 1 , Anja Ehrhardt 1<br />

1 Department of Virology, Max von Pettenkofer-<br />

Institute, LMU, <strong>Munich</strong>, <strong>German</strong>y<br />

High-capacity adenoviral vectors (HC-AdVs)<br />

devoid of all viral coding sequences offer high<br />

transduction efficiencies, large packaging<br />

capacities (up to 35 kb) and reduced toxicity. To<br />

further improve the persistence of transgene<br />

expression and the maintenance of the vector<br />

genome in cycling cells, we recently developed<br />

an adeno/transposase hybrid-vector-system<br />

utilizing a hyperactive Sleeping Beauty<br />

transposase HSB5 <strong>for</strong> somatic integration (Hausl,<br />

Zhang et al., Molecular <strong>Therapy</strong>, accepted).<br />

However, with respect to preclinical evaluation<br />

this hybrid-vector system needs to be<br />

characterized in more detail. Herein, we<br />

optimized the virus dose required <strong>for</strong> long-term<br />

effects without toxicity and we per<strong>for</strong>med a vector<br />

fate analysis. Based on our established hybrid-<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong>


vector system, female and male C57Bl/6 mice<br />

were co-injected at three dosages [1.6 × 10 8 8 ×<br />

10 8 and 4 × 10 9 transducing units (TU) total per<br />

mouse] with a canine factor IX (cFIX) transposon<br />

donor vector and a second vector encoding the<br />

hyperactive SB transposase HSB5 and Flp<br />

recombinase (n = 5 per group). In groups which<br />

received the highest dose, toxicity was observed<br />

in mice of both genders. At the lowest dose, only<br />

low levels of serum cFIX were detected ( ≈ 100<br />

ng/ml). This was in sharp contrast to the middledose<br />

group in which we measured ≈ 1000 ng/ml<br />

serum cFIX levels without toxicity. We concluded<br />

that an efficient non-toxic dose <strong>for</strong> our hybridvector<br />

lies between 8 × 10 8 to 4 × 10 9 TUs per<br />

mouse. Notably, at the middle dose we observed<br />

a gender influence on transgene expression<br />

levels. In order to address issues related to<br />

genotoxicity after HSB5-mediated somatic<br />

integration, transposition events were analyzed<br />

by a PCR-based high-throughput sequencing<br />

method. For each individual we generated 4<br />

genomic DNA libraries using different restriction<br />

enzymes. Of the 73 transposition events<br />

identified in liver of HSB treated mice, 74%<br />

(54/73) were found in non-gene areas and 26%<br />

(19/73) in genes indicating a random integration<br />

pattern. Of the transposition events detected in<br />

genes, 17 insertion sites were in introns and only<br />

2 in exons. From all animals, no integration site<br />

was found in or near putative oncogenes.<br />

Session: Pharmacology and Toxicology<br />

P 62<br />

Insertional Mutagenesis in Mature T Cells – A<br />

Mathematical Modelling Approach<br />

Sebastian Gerdes 1 , Sebastian Newrzela 2,3 , Tim<br />

Heinrich 2,3 , Dorothee von Laer 2,4 , Ingmar<br />

Glauche 1 , Ingo Roeder 1<br />

1 Institute <strong>for</strong> Medical In<strong>for</strong>matics and Biometry,<br />

Faculty of Medicine “Carl Gustav Carus”,<br />

Dresden University of Technology, Dresden,<br />

<strong>German</strong>y; 2 Frankfurter Stiftung für krebskranke<br />

Kinder, Frankfurt am Main, <strong>German</strong>y; 3 Pathology<br />

of the Goethe-University Hospital Frankfurt,<br />

Frankfurt am Main, <strong>German</strong>y; 4 Innsbruck Medical<br />

University, Innsbruck, Austria<br />

Leukemia caused by insertional mutagenesis is a<br />

major risk of gene therapy with HSCs. In<br />

contrast, polyclonal mature T cells have turned<br />

out to be surprisingly resistant to leukemogenic<br />

insertional mutagenesis. Newrzela et al. showed<br />

that leukemia/lymphoma did not occur upon<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong><br />

| 74<br />

transplantation of wild-type mature T cells with<br />

polyclonal T-cell receptors (TCR) being<br />

transduced with high copy numbers of<br />

gammaretroviral vectors encoding potent T- cell<br />

oncogenes into RAG1-1-deficient recipients [1].<br />

However, further studies demonstrated that the<br />

transplantation of T cells from TCR-monoclonal<br />

OT1 mice that were transduced with the same<br />

protocol resulted in leukemia/lymphoma. The<br />

underlying mechanisms that prevent oncogenesis<br />

in the polyclonal situation and permit the<br />

outbreak of leukemia in the monoclonal situation<br />

are currently unclear. Using a mathematical<br />

modelling approach, we challenge the arising<br />

hypothesis that polyclonality induces competition<br />

within the T-cell repertoire, which in turn<br />

suppresses the emergence of a leukemic clone.<br />

As a first step, we developed a mathematical<br />

model of T-cell homeostasis that is derived from<br />

a similar niche-based based model of<br />

hematopoiesis [2]. The key assumption of the<br />

novel model is that T-cell survival is critically<br />

dependant on the interaction of the clone-specific<br />

TCR with self-peptide-MHC-complexes and<br />

there<strong>for</strong>e subject to competition between different<br />

T-cell clones. Our model consistently reproduces<br />

the polyclonal pattern observed in T-cell<br />

homeostasis, and responds in an adequate<br />

manner to perturbations of the system such as<br />

infection. Furthermore, based on our modelling<br />

results, we speculate how the deregulation of Tcell<br />

receptor affinities supports the <strong>for</strong>mation of<br />

dominant clones. The modelling results<br />

underscore the possibility that clonal competition<br />

can prevent the outbreak of overt mature T-cell<br />

leukemia/lymphoma. References: [1] Newrzela S<br />

et al. Resistance of mature T cells to oncogene<br />

trans<strong>for</strong>mation. Blood. 2008;112(6):2278–2286.<br />

[2] Roeder I., Loeffler M. A novel dynamic model<br />

of hematopoietic stem cell organization based on<br />

the concept of within-tissue plasticity. Exp.<br />

Hematol. 2002;30(8):853-861.<br />

Session: Pharmacology and Toxicology


75 |<br />

P 63<br />

Zinc-Finger Nucleases <strong>for</strong> Targeted <strong>Gene</strong><br />

Repair: Studying Off-Target Activities<br />

Frank Radecke 1 , Sarah Radecke 2 , Toni<br />

Cathomen 3 , Klaus Schwarz 1,4<br />

1 Institute <strong>for</strong> Transfusion Medicine, University of<br />

Ulm, Ulm, <strong>German</strong>y; 2 Institute <strong>for</strong> Clinical<br />

Transfusion Medicine and Immunogenetics Ulm,<br />

Ulm, <strong>German</strong>y; 3 Department of Experimental<br />

Hematology, Hannover Medical School,<br />

Hannover, <strong>German</strong>y; 4 Institute <strong>for</strong> Clinical<br />

Transfusion Medicine and Immunogenetics,<br />

University of Ulm, Ulm, <strong>German</strong>y<br />

Targeted gene repair with single-stranded<br />

oligodeoxynucleotides as repair matrix often<br />

yields low rates of corrected cells, but these rates<br />

can be enhanced by the concomitant induction of<br />

a DNA double-strand break (DSB) close to the<br />

mutation. As a tool to achieve defined DSBs,<br />

customisable zinc-finger nucleases (ZFNs) are<br />

studied. They consist of a specific DNA-binding<br />

domain (designed zinc-finger motifs) linked to a<br />

nonspecific FokI-derived dimer-<strong>for</strong>ming nuclease<br />

domain. Here, off-target activities of two ZFNs<br />

(GZF1 and GZF3) were investigated using three<br />

complementing approaches. To reduce toxicity,<br />

both ZFNs already harbour a variant dimer<br />

interface to favour heterodimerisation. For in vitro<br />

cleavage assays, the ZFN proteins were<br />

incubated with target locus DNA. Off-target<br />

activity was detected at two positions due to<br />

unwanted GZF1 homodimerisation, where one<br />

ZFN bound its perfect 9-mer site while the<br />

partner functionally attached to a mismatched<br />

sequence. To examine whether this might<br />

happen in a 293 cell line with a transgenic singlecopy<br />

target locus, one expression vector (<strong>for</strong><br />

GZF1 or GZF3) was transfected to then analyse<br />

target locus DNAs <strong>for</strong> signatures of DSB repair<br />

(e.g., nonhomologous end joining (NHEJ)). At the<br />

regular GZF1/GZF3 site, one NHEJ-derived<br />

modification per ≈ 15,000 alleles was observed<br />

when only GZF1 was present. Since similar<br />

binding site variants were found also <strong>for</strong> GZF3<br />

homodimers, related off-target activity—not yet<br />

detected—might also occur. In silico analyses<br />

(Ensembl BLASTN on haploid genome) predicted<br />

28,100 and 12,695 additional perfect sites <strong>for</strong><br />

GZF1 and GZF3, respectively. Based on<br />

sequence analyses from the in vitro and in cellula<br />

assays, ≈ 6% of these sites were classified as<br />

potential off-targets with canonical 5–7 base-pair<br />

spacers and second binding sites (9-mers) with<br />

6–8 matches. At half of these sites, the favoured<br />

heterodimers might <strong>for</strong>m. In conclusion, our<br />

multifaceted plat<strong>for</strong>m consisting of genome-wide<br />

in silico predictions combined with in vitro and in<br />

cellula assays represents a potent tool to<br />

evaluate ZFN specificities and can be used to<br />

estimate biological activities of any ZFN with<br />

known DNA binding parameters.<br />

Session: Pharmacology and Toxicology<br />

P 64<br />

Control Mechanisms in Mature T Cell<br />

Leukemia/Lymphoma<br />

Sebastian Newrzela 1 , Nabil Al-Ghaili 2 , Tim<br />

Heinrich 1 , Mina Petkova 2 , Kerstin Cornils 3 , Sylvia<br />

Hartmann 1 , Martin-Leo Hansmann 1 , Boris<br />

Fehse 3 , Sebastian Gerdes 4 , Ingo Roeder 4 ,<br />

Dorothee von Laer 5<br />

1Pathology of the Goethe-University Frankfurt,<br />

Frankfurt am Main, <strong>German</strong>y; 2 Frankfurter<br />

Stiftung für krebskranke Kinder, Frankfurt am<br />

Main, <strong>German</strong>y; 3 University Medical Center<br />

Hamburg-Eppendorf, Hamburg, <strong>German</strong>y;<br />

4 Institute <strong>for</strong> Medical In<strong>for</strong>matics and Biometry,<br />

Faculty of Medicine “Carl Gustav Carus”,<br />

Dresden University of Technology, Dresden,<br />

<strong>German</strong>y; 5 Innsbruck Medical University,<br />

Innsbruck, Austria<br />

In safety studies in a mouse model, we found that<br />

in hematopoietic stem cells high copy numbers of<br />

gammaretroviral vectors expressing T-cell<br />

oncogenes readily induced T-cell<br />

leukemia/lymphoma in recipient mice. However,<br />

mature polyclonal T cells could not be<br />

trans<strong>for</strong>med (Newrzela et al., Blood 2008). Yet,<br />

and quite surprisingly, T-cell receptor (TCR)<br />

monoclonal T lymphocytes derived from OT-I<br />

mice were trans<strong>for</strong>med with the same kinetics<br />

and efficiency as stem cells. This propensity of<br />

monoclonal T-cell populations to develop<br />

malignancies is highly relevant <strong>for</strong> evaluating the<br />

safety of therapeutic gene transfer into mature T<br />

cells, especially the adoptive transfer of<br />

therapeutic TCRs. On the other hand, these<br />

observations imply that polyclonal T-cell<br />

populations can control the outgrowth of<br />

malignant T-cell clones. The population-dynamics<br />

of T cells will be analyzed with respect to the<br />

diversity of the T-cell repertoire. The generated<br />

data on T-cell dynamics will furthermore help to<br />

validate mathematical models on the normal and<br />

malignant T-cell homeostasis, which will be<br />

developed in the group of Ingo Roeder, Dresden.<br />

In addition, the molecular mechanisms that<br />

control T-cell malignancy in the polyclonal, but<br />

enhance leukemogenesis in the monoclonal<br />

setting will be identified. The mechanisms<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong>


involved in the pathogenesis of mature T-cell<br />

malignancies are addressed in this study.<br />

Session: Pharmacology and Toxicology<br />

P 65<br />

Characterization of rAAV Genomes After<br />

Delivery of ssAAV or scAAV in Rats by LAM-<br />

PCR and Deep Sequencing<br />

Christine Kaeppel 1 , Ali Nowrouzi 1 , Uwe Appelt 1 ,<br />

Anne Arens 1 , Christian Weber 1 , Maude Flageul 2 ,<br />

Paula Miranda 3 , Nicolas Ferry 2 , Piter Bosma 3 ,<br />

Christof von Kalle 1 , Manfred Schmidt 1<br />

1National Center <strong>for</strong> Tumor Diseases Heidelberg,<br />

Department of Translational Oncology, <strong>German</strong><br />

Cancer Research Center, Heidelberg, <strong>German</strong>y;<br />

2 INSERM UMR 948, Nantes, France; 3 Academic<br />

Medical Center Liver Center, Amsterdam, The<br />

Netherlands<br />

Long-term gene expression can be achieved<br />

following recombinant adeno-associated virus<br />

(rAAV) vector delivery in vivo. Previously, we<br />

have shown that rAAV8 delivery to newborn rat<br />

liver results in low frequent integrated rAAV<br />

genomes in a minority of cells. However, even<br />

the rare integration frequency of the rAAV vector<br />

raises concerns about its clinical safety. To<br />

evaluate the safety of different AAV-serotypes<br />

and vector doses in preclinical settings we<br />

estimated rAAV integration characteristics in rat<br />

liver after single-stranded (ss)AAV or selfcomplementary<br />

(sc)AAV injection of different<br />

vector doses. The persistence of ssAAV and<br />

scAAV was analysed in transduced rat liver 14<br />

months and 13 months post-rAAV injection. In<br />

rats transduced with scAAV partial hepatectomy<br />

(HP) was per<strong>for</strong>med 13 months post-injection<br />

and the integration frequency was evaluated 3<br />

months post HP. LAM-PCR was per<strong>for</strong>med on<br />

250-500 ng of total DNA derived from 12 ssAAV1,<br />

6 scAAV1 and 6 scAAV8 transduced rats and<br />

combined with deep sequencing allowing<br />

simultaneous and efficient identification of<br />

complex rAAV concatemeric <strong>for</strong>ms and<br />

proviruses. Sequencing of > 68.000 LAMamplicons<br />

revealed 67 unique insertion sites (IS),<br />

in ssAAV transduced rats up to seven per sample<br />

and in scAAV transduced ones up to eight per<br />

sample. The averaged number of IS was higher<br />

in rat liver transduced with 1.2 × 10 12 TU/kg<br />

ssAAV1 compared to a vector dose of 8 × 10 12<br />

TU/kg (4.6 IS vs. 0.5 IS). Semi-quantitative<br />

measurements of rAAV concatemers showed<br />

that in rat liver transduced with 8 × 10 12 TU/kg<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong><br />

| 76<br />

ssAAV1 up to 100% of the analysed sequences<br />

were due to concatemers while in rat liver<br />

transduced with 1.2 × 10 12 TU/kg up to 3/4 of all<br />

analysed sequences could be referred to IS. In<br />

addition, sequencing of rAAV LAM-amplicons<br />

derived from ssAAV and scAAV transduced rat<br />

liver was used to show the feasibility of complete<br />

ITR sequencing in vivo. In partial ITR no<br />

preferential ITR breakpoints were detected in<br />

ssAAV transduced rat liver while in scAAV<br />

transduced ones preferential ITR breakpoints<br />

were detected. These insights are of clinical<br />

relevance <strong>for</strong> potential vector genotoxicity of<br />

different rAAV serotypes and doses.<br />

Session: Pharmacology and Toxicology<br />

P 66<br />

Improved Drug-Selectable Fluorescent<br />

Proteins as Bifunctional Marker <strong>Gene</strong>s<br />

Kristoffer Weber 1 , Ellen Preuß 1,2 , Lisa<br />

Weingarten 1 , Boris Fehse 1<br />

1 Universitätsklinikum Hamburg-Eppendorf,<br />

Hamburg, <strong>German</strong>y; 2 Frankfurter Stiftung für<br />

krebskranke Kinder, Hamburg, <strong>German</strong>y<br />

The use of fluorescent proteins (FPs) as markers<br />

expressed by gene transfer vectors facilitates fast<br />

and unambiguous identification of gene-modified<br />

cells. However, to select cells based on their<br />

expression of FPs, fluorescence-activated cell<br />

sorting would have to be used. This complex and<br />

expensive technique is not available in all<br />

laboratories, bears a certain contamination risk<br />

and might be too stressful <strong>for</strong> some delicate cell<br />

types. To overcome these limitations we have<br />

recently developed ten fusion proteins as<br />

bifunctional markers composed of a variety of<br />

fluorescent proteins (Cerulean, eGFP, Venus,<br />

dTomato, mCherry) fused to drug resistance<br />

proteins (NeomycinR, ZeocinR, PuromycinR,<br />

HygromycinR, BlasticidinR). Now we present the<br />

second generation of bifunctional markers which<br />

are greatly improved in brightness by inserting a<br />

2A peptide in between the fluorescent protein<br />

and the resistance protein. In fact, the brightness<br />

of the so far dimmest marker (eGFP fused to<br />

PuromycinR) was increased by a factor of seven,<br />

now reaching 90% of the brightness of unfused<br />

eGFP. All bifunctional markers have been tested<br />

in lentiviral vectors (www.LentiGO-Vectors.de)<br />

and were cloned as “building blocks” to easily<br />

interchange the resistances in a single cloning<br />

step. Importantly, working with the new<br />

bifunctional markers revealed that the efficacy of


77 |<br />

cell selection could be overestimated with<br />

standard antibiotic-resistance genes. In fact, in<br />

different adherent cell lines we found by flow<br />

cytometry that antibiotic-mediated selection of<br />

cells was by far not complete, even though all<br />

control cells without the respective antibiotic<br />

resistance were dead. This data not only has<br />

important implications <strong>for</strong> assays with antibioticsselected,<br />

apparently pure cell populations, but<br />

also indicates some so far unknown bystander<br />

protection of nontransduced cells in adherent<br />

cultures. In conclusion we suppose the novel<br />

bifunctional markers are well suited <strong>for</strong> numerous<br />

applications and different vector systems.<br />

Session: Pharmacology and Toxicology<br />

P 67<br />

From Insertion Site Profiles to Clonal<br />

Contributions<br />

J. Uwe Appelt, Frank A. Giordano, Anne Arens,<br />

Cynthia C Bartholomae, Manfred Schmidt,<br />

Christof von Kalle, Stephanie Laufs<br />

Department of Translational Oncology, National<br />

Center <strong>for</strong> Tumor Diseases and <strong>German</strong> Cancer<br />

Research Center, Heidelberg, <strong>German</strong>y<br />

Initially, our goals were to test if viral insertion is<br />

nonrandom and, specifically, to clarify whether<br />

host and disease-specific integration patterns<br />

exist. We developed the bioin<strong>for</strong>matic<br />

applications IntegrationSeq, IntegrationMap and<br />

QuickMap, and others (manuscript in revision)<br />

which allow high throughput analyses (next<br />

generation sequencing) of insertion sites of viral<br />

vectors (QuickMap available at<br />

http://www.gtsg.org). Furthermore, we designed a<br />

first version of a database, which to date contains<br />

more than 77,334 different vector insertion sites<br />

derived from previous analyses on different<br />

vectors (ASLV, FIV, MMTV, HTLV, HIV, EIAV,<br />

FV, MLV, and SIV) and different host cells. We<br />

subjected all insertion sites stored in this<br />

database to our analysis pipeline and<br />

characterized insertions with regard to<br />

localization on chromosomes, in or next to a<br />

genes, cancer genes, fragile sites, transcription<br />

factor binding sites, CpGs, or repetitive elements<br />

(SINE, LINE, LTR). A random insertion site set<br />

(10 6 sequences) reliably reflects random<br />

distribution and serves as our reference <strong>for</strong> all<br />

analyses of experimental data. We saw that the<br />

distinct insertion profiles of the different<br />

retroviruses resemble genomic finger prints,<br />

which allow distinguishing between different<br />

retroviruses. Further, a high-resolution metaanalysis<br />

HIV vector insertions revealed that HIV<br />

significantly spare a region of 1 kb+ / − to<br />

transcription start sites (Giordano et al., AIDS<br />

2009). Using the insertion site profiles and the<br />

in<strong>for</strong>mation on redundancy of recovered<br />

sequences (read counts), both provided by<br />

QuickMap and our other integration site<br />

sequence data mining tools, we could reliably<br />

estimate clonal compositions of complex samples<br />

(preclinical and clinical studies), thus allowing<br />

monitoring of transduced cells and assessment<br />

<strong>for</strong> the safety of gene transfer. We are now<br />

building up a database that will allow the dynamic<br />

storage of integration site sequences in the Terra<br />

range and comprehensively fulfill the needs <strong>for</strong> (i)<br />

more specialized features of vectors (e.g., IDLV,<br />

AAV), (ii) genomic and transcriptomic features<br />

near the integration site and (iii) new powerful<br />

sequencing technologies.<br />

Session: Pharmacology and Toxicology<br />

P 68<br />

Next <strong>Gene</strong>ration Sequencing Data<br />

Management <strong>for</strong> Integration Site Analyses<br />

Anne Arens, Derek Gustafson, Cynthia C<br />

Bartholomae, Uwe Appelt, Frank Giordano,<br />

Annette Deichmann, Hanno Glimm, Stephanie<br />

Laufs, Christof von Kalle, Manfred Schmidt<br />

Department of Translational Oncology, National<br />

Center <strong>for</strong> Tumor Diseases (NCT) and <strong>German</strong><br />

Cancer Research Center (DKFZ), Heidelberg,<br />

<strong>German</strong>y<br />

Integration site analyses have become a valuable<br />

and powerful tool to detect clonal repopulation<br />

kinetics in clinical gene therapy, to address the<br />

biosafety of a specific gene transfer approach or<br />

to uncover vector-induced side effects. With a<br />

combinatorial approach of standard or<br />

nonrestrictive (nr) linear amplification-mediated<br />

(LAM) PCR followed by next generation<br />

sequencing, time- and cost-efficiency of<br />

integration site sequence retrieval can be<br />

improved by several logs compared to standard<br />

Sanger sequencing technologies. Using our<br />

implemented analyses tools, such as Quickmap<br />

(www.gtsg.org) and others (Paruzynski A et al.,<br />

Nature Protocols 2010), sequencing data output<br />

provides relevant integration site in<strong>for</strong>mation<br />

such as chromosome, chromosomal locus,<br />

distance and orientation to the next RefSeq<br />

genes and retrieval frequency of individual<br />

integration sites. However, integration site data<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong>


mining nowadays not only requires setting up<br />

certain software that allow an automated and<br />

standardized readout which can be handled by<br />

non-bioin<strong>for</strong>maticians. The vast amount of<br />

different vector types used in various areas of<br />

gene transfer approaches (e.g., high frequent<br />

integrating oncoretro- and lentiviral vectors, lowfrequent<br />

integrating integrase deficient lentiviral<br />

and AAV vectors as well as transposons such as<br />

Sleeping Beauties) <strong>for</strong> different cell types and<br />

species also requires a hitherto unknown<br />

flexibility of relevant downstream programs to<br />

answer the specific scientific questions beyond.<br />

Here, we will present some aspects of our<br />

‘downstream’ bioin<strong>for</strong>matical data analysis<br />

system that meet these requirements, including<br />

among others (i) therapeutically relevant<br />

in<strong>for</strong>mations on clonal constitution and dynamics<br />

of gene-modified cell populations in preclinical<br />

and clinical gene transfer settings<br />

(‘pharmacokinetics'), (ii) precision of vector<br />

integration and (iii) sequence homology<br />

comparison and others.<br />

Session: Pharmacology and Toxicology<br />

P 69<br />

Investigation of the Localisation and<br />

Interactions of Adenoviral Vectors with<br />

Immune Cell Populations in the Spleen<br />

Following Intravascular Delivery<br />

Lynda Coughlan 1 , Angela C. Bradshaw 1 , Raoul<br />

Alba 1 , Robert A. McDonald 1 , Nico van Rooijen 2 ,<br />

James M. Brewer 3 , Paul Garside 3 , Stuart A.<br />

Nicklin 1 , Andrew H. Baker 1<br />

1 BHF GCRC, University of Glasgow, Glasgow,<br />

UK; 2 Department of Molecular Cell Biology, Vrije<br />

Universiteit Medical Centre (VUMC), Amsterdam,<br />

The Netherlands; 3 Division of Immunology,<br />

Infection and Inflammation, GBRC, University of<br />

Glasgow, Glasgow, UK<br />

The spleen is an important site <strong>for</strong> the induction<br />

of immune responses to adenoviral (Ads)<br />

vectors, especially following intravascular<br />

delivery. Ads have been shown to interact with<br />

macrophage populations and dendritic cells in the<br />

spleen, and high level splenic uptake has been<br />

associated with vector-induced toxicity in vivo.<br />

However, the precise immune effectors which<br />

determine the release of characteristic<br />

inflammatory cytokines in response to Ad remain<br />

to be fully identified. Using a panel of viruses,<br />

including Ad5, Ad48 and hexon-modified Ad5, we<br />

assessed their interactions with cell markers in<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong><br />

| 78<br />

the spleen (CD11b, CD11c, SIGNR1, MARCO,<br />

CD169 and F4/80) at low and high doses of Ad,<br />

and at early and late time-points post-injection. In<br />

order to fully analyse these interactions and their<br />

contribution to inflammatory responses, we<br />

administered a cohort of mice with clodronateencapsulated<br />

liposomes and assessed the<br />

effects on macrophage and other immune cell<br />

populations in the spleen. We quantified serum<br />

cytokines 6 hr post-injection using a 20-plex<br />

luminex assay, <strong>for</strong> clodronate treated and<br />

untreated animals. Fluorescently labeled Ads<br />

(Ad5, Ad48 and hexon-modified Ad5) were<br />

detected by immunohistochemistry in colocalisation<br />

with MARCO + and Moma-1 + cells,<br />

within the marginal zone surrounding the white<br />

pulp of the spleen, and with F4/80 + red pulp<br />

macrophages (1 h). These macrophage<br />

populations were almost completely absent in<br />

animals treated with clodronate liposomes and<br />

subsequently the spatial distribution of virus<br />

within the spleen was altered. The defining<br />

splenic cell markers <strong>for</strong> virus transgeneexpressing<br />

cell types at later time-points (48 h)<br />

were not the same as those detected at early<br />

time-points. These currently remain to be<br />

identified. Further studies will be required to fully<br />

characterise the time-course of adenoviral uptake<br />

in the spleen and the contribution of the various<br />

cellular effectors to the induction of inflammation.<br />

Session: Pharmacology and Toxicology


79 |<br />

DGGT 2010 Abstract author index<br />

Abken, Hinrich, Inv 22<br />

Acosta-Sanchez, Abel, Inv 12<br />

Al-Ghaili, Nabil, P 64<br />

Alba, Raoul, P 38, P 69<br />

Albarenque, Stella, P 48<br />

Ali, Robin R, Inv 14, P 30<br />

Almarza, David Gomez, Or 2<br />

Aloysius, Mark, Inv 23<br />

Altomonte, Jennifer, P 14, P 44, P 45<br />

Anton, Martina, Inv 2, Or 18, P 41, P 46, P 50<br />

Antoniou, Michael, Or 9<br />

Apfel, Sibylle, P 14<br />

Appelt, Uwe, P 65, P 67, P 68<br />

Arens, Anne, Or 7, P 65, P 67, P 68<br />

Argyros, Orestis, P 23, P 24<br />

Armeanu, Sorin, P 42<br />

Arzumanov, Andrey A, Inv 11<br />

Bachem, Max G, P 59<br />

Bahadur, Dhirendra, P 15<br />

Bai, Lin, Or 6<br />

Baier, Ruth, P 18, P 19<br />

Baiker, Armin, P 21<br />

Bainbridge, JWB, Inv 14<br />

Baker, Andrew H, Inv 10, Or 21, P 38, P 69<br />

Bakshi, Rakesh, P 26<br />

Ball, Claudia R, Or 7, P 37<br />

Ballmaier, Matthias, Or 10<br />

Banerjee, Rinti, P 15<br />

Banfi, Andrea, P 28<br />

Bartholomae, Cynthia C, P 67, P 68<br />

Bauer, Ralf, P 29<br />

Baum, Christopher, Or 10, Or 15, Or 19<br />

Becirovic, Elvir, Or 6<br />

Beck, Susanne C, Or 6<br />

Bednarski, Christien, P 17, P 22<br />

Belay, Eyayu, Inv 12<br />

Bell, John C, Inv 18, Inv 19<br />

Benda, Veronika, P 55<br />

Bendle, Gavin M, Or 17<br />

Benihoud, Karim, P 59<br />

Benz, Alexander, P 57<br />

Bergemann, Christian, P 8<br />

Bernhard, Helga, P 56<br />

Bhalla, Joti, Inv 23<br />

Bhattacharya, SS, Inv 14<br />

Biel, Martin, Or 6<br />

Bies, Laura, Or 17<br />

Bitzer, Michael, P 42<br />

Blaesen, Markus, P 18, P 19<br />

Blankenstein, Thomas, P 54<br />

Blasczyk, Rainer, Or 7<br />

Bock-Marquette, Ildiko, Or 8, P 27, P 36<br />

Bode, Juergen, P 12<br />

Boekstegers, Peter, P 27, P 28<br />

Böhm, Marie, Or 7<br />

Böning, Guido, Or 14<br />

Bosma, Piter, P 65<br />

Boztug, Kaan, Or 7<br />

Bradke, Frank, P 14<br />

Bradshaw, Angela C, Or 21, P 69<br />

Braren, Rickmer, P 45<br />

Bräuchle, Christoph, Or 5, P 3<br />

Breitbach, Caroline, Inv 18<br />

Brewer, James M., P 69<br />

Brill, Thomas, P 55<br />

Brugman, Martijn, Or 10, Or 15<br />

Buch, Prateek, P 30<br />

Buchholz, Christian J, P 42<br />

Buck, Andreas K, P 45<br />

Buckley, Suzanne MK, Inv 13<br />

Büning, Hildegard, Or 2, Or 6, P 32<br />

Busch, Dirk H, P 56<br />

Büsche, Guntram, Or 10<br />

Caizergues, Didier, Inv 24<br />

Cantz, Tobias, Or 15, P 52, P 52<br />

Carpentier, A, Inv 15<br />

Cathomen, Toni, Or 3, P 11, P 17, P 22, P 52, P 52, P<br />

63<br />

Cattaneo, Roberto, Inv 17, P 42<br />

Cengizeroglu, Arzu, Or 18, P 41<br />

CGD Consortium, Inv 16<br />

Chan, Jerry, Inv 13<br />

Chan, Lucas, Inv 23<br />

Charrier, Sabine, Inv 24<br />

Chick, Helen, P 38<br />

Chuah, Marinee KL, Inv 12<br />

Cichutek, Klaus, Inv 25, P 42<br />

Cim, Abdullah, Or 9<br />

Collins, Louise, Or 9<br />

Conrad, Heinke, P 56<br />

Cooper, Jonathan D, Inv 13<br />

Corjon, Stephanie, Or 1<br />

Cornils, Kerstin, P 64<br />

Cornu, Tatjana I, P 17<br />

Coughlan, Lynda, P 69<br />

Coutelle, Charles, P 23, P 24<br />

Cowled, Christopher, Inv 23<br />

Daenthanasanmak, Anusara, P 26<br />

Dannemann, Nadine, P 22<br />

de Bruin, Karla, P 3<br />

Deedigan, Laura, P 48<br />

Deichmann, Annette, P 68<br />

Deindl, Elisabeth, Or 8<br />

Deutsch, Daniela, P 41<br />

Diaz, Margarita, Or 20<br />

Diepolder, Helmut, P 26<br />

DiMaio, Michael, P 27<br />

Döblinger, Markus, P 53<br />

Dohmen, Christian, P 5<br />

Dorer, Dominik E, P 16, P 39<br />

Draganovici, Dan, Or 14<br />

Dreyer, Anne-Kathrin, P 22<br />

Duffy, Margaret R, Or 21<br />

Dumler, Katja, P 46, P 50<br />

Ebert, Oliver, P 14, P 44, P 45<br />

Edinger, Daniel, P 7<br />

Egana, Tomas J, P 34<br />

Egerer, Lisa M, P 31, P 32<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong>


Ehrhardt, Anja, Or 3, P 9, P 10, P 20, P 21, P 35, P 61<br />

El-Aouni, Chiraz, P 28<br />

Ellwart, Joachim, Or 18<br />

Emmrich, Stephan, P 43<br />

Engels, Boris, P 54<br />

Engler, Tatjana, Or 1<br />

Eremin, Oleg, Inv 23<br />

Espenlaub, Sigrid, Or 1, P 13<br />

Fabre, John, Or 9<br />

Fahl, Edda, Or 6<br />

Farkasova, Katarina, Or 18, P 7, P 41<br />

Farzaneh, Farzin, Inv 21, Inv 23<br />

Fedonidis, Constantinos, P 23<br />

Fehse, Boris, P 64, P 66<br />

Fella, Carolin, Or 1<br />

Ferizi, Mehrije, P 55<br />

Ferna´ndez Ulibarri, Ine´s, P 16<br />

Ferry, Nicolas, P 65<br />

Filipczyk, Adam, Or 15<br />

Fischer, Cornelia, P 55<br />

Fischer, M Dominik, Or 6<br />

Fitzke, FW, Inv 14<br />

Flageul, Maude, P 65<br />

Foralosso, Ruggero, P 5<br />

Frankenberger, Bernhard, Or 16<br />

Frenz, Jessica, P 25<br />

Frieb, Wolfgang, P 1<br />

Fröhlich, Thomas, P 5, P 6<br />

Fulda, Simone, P 48<br />

Gait, Michael J, Inv 11<br />

Galla, Melanie, Or 15<br />

Galy, Anne, Inv 24<br />

Gänsbacher, Bernd, P 33, P 46, P 50, P 55<br />

Garritsen, Henk, Inv 21<br />

Garside, Paul, P 69<br />

Gaudet, D, Inv 15<br />

Gellhaus, Katharina, P 11, P 17<br />

Geraerts, Martine, Inv 12<br />

Gerdes, Sebastian, P 51, P 62, P 64<br />

Gildehaus, Franz Josef, Or 14, P 47<br />

Giordano, Frank A, P 67, P 68<br />

Giri, Jyotsnendu, P 15<br />

Giroglou, Tsanan, Or 11<br />

Glauche, Ingmar, P 51, P 62<br />

Glimm, Hanno, Or 7, Or 17, Or 20, P 37, P 68<br />

Globisch, Franziska, P 28, P 36<br />

Göke, Burkhard, Or 13, Or 14, P 47<br />

Gollisch, Tim, Or 6<br />

Gottlieb, Elena, P 36<br />

Greentree, S, Inv 15<br />

Grez, Manuel, Inv 16, Or 20<br />

Grünwald, Geoffrey K, Or 13, P 47<br />

Guhl, Eva, P 52, P 52<br />

Gustafson, Derek, P 68<br />

Gutzmer, Ralf, Inv 21<br />

Haase, Rudolf, Or 18, P 4, P 21, P 41<br />

Habib, Nagy, Inv 23<br />

Hagedorn, Claudia, P 60<br />

Hallek, Michael, Or 2<br />

Händel, Eva M, P 17<br />

Hanschmann, Kay-Martin O, P 42<br />

Hansmann, Martin-Leo, P 57, P 58, P 64<br />

Harbottle, Richard, P 23, P 24<br />

Hardwick, Nicola, Inv 23<br />

Harms, Nina, Or 3, P 35<br />

Hartmann, Marianne, P 31<br />

Hartmann, Sylvia, P 57, P 58, P 64<br />

Hartog, Christoph, P 25<br />

Hatzopoulos, Antonis K, P 27<br />

Hausl, Martin, P 10, P 20, P 21, P 35, P 61<br />

Heckl, Dirk, Or 10<br />

Heilbronn, Regine, P 11, P 17, P 52, P 52<br />

Heinrich, Tim, P 57, P 58, P 62, P 64<br />

Heinz, Niels, Or 19<br />

Hensel, Karin, P 8<br />

Herbst, Friederike, P 37<br />

Hermann, Felix G, P 31<br />

Hinkel, Rabea, Inv 3,Or 8, P 27, P 28, P 36<br />

Hirschberger, Johannes, P 55<br />

Hoeffer, Klemens, Inv 13<br />

Holder, GE, Inv 14<br />

Holm, Per Sonne, Or 13, P 13, P 46, P 49, P 53<br />

Hopfner, Ursula, P 34<br />

Horstkotte, Jan C, P 27<br />

Howe, Steven, P 23, P 24<br />

Huber, Gesine, Or 6<br />

Imhoff, Andreas, P 50<br />

Ingram, Wendy, Inv 23<br />

Itaka, Keiji, Or 4<br />

Ivics, Zoltán, Inv 12, P 35<br />

Izsvák, Zsuzsanna, Inv 12, P 35<br />

Jahnke, Anika, P 55<br />

Janicki, Hanna, P 32<br />

John, Katja, P 43<br />

Jones, Peter, Or 9<br />

Jun, Tye Gee, Inv 23<br />

Kaeppel, Christine, P 65<br />

Kahle, Joerg, P 31<br />

Kampik, Daniel, P 30<br />

Kane, NM, P 38<br />

Kasper, Julia C, P 1<br />

Kataoka, Kazunori, Or 4<br />

Kathöfer, Astrid, P 25<br />

Katus, Hugo A, P 29<br />

Kay, Mark A, Inv 8<br />

Khan, Kafaitullah, P 17, P 52, P 52<br />

Kimpel, Janine, P 31, P 32<br />

Kirn, David H, Inv 18<br />

Klein, Christoph, Or 7<br />

Klibanov, Alexander L, P 8<br />

Kloz, Ulrich, P 37<br />

Klump, Hannes, Or 15<br />

Klutz, Kathrin, Or 13, Or 14, P 47<br />

Kneiske, Inna, Or 11<br />

Knoop, Kerstin, Or 14<br />

Koch, Christian, P 15, P 33, P 34<br />

Koch, Susanne, Or 6<br />

Kochanek, Stefan, Or 1, P 59<br />

Koenig, Frauke M, Or 5<br />

Kolk, Andreas, P 33<br />

Kolokythas, Marie, Or 14<br />

Kondratenko, Irina, Or 7<br />

Kontermann, Roland, P 39<br />

Köstlin, Roberto, P 55<br />

Kostova, Youlia, P 46<br />

Kreppel, Florian, Or 1, P 13<br />

Krishnamoorthy, Vidhyasankar, Or 6<br />

Kritz, AB, P 38<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong><br />

| 80


81 |<br />

Kuehle, Johannes, P 12<br />

Kühlcke, Klaus, Or 7<br />

Kühle, Johannes, Or 15<br />

Kuhs, Sandra, Inv 21, P 26<br />

Kupatt, Christian, Inv 3, Or 8, P 27, P 28, P 36<br />

Larcher, Fernando Laguzzi, Or 2<br />

Larkin, Daniel F.P., P 30<br />

Lauer, Ullrich, P 42<br />

Laufs, Stephanie, P 67, P 68<br />

Lebherz, Corinna, Or 8, P 27, P 28, P 36<br />

Leisegang, Matthias, Or 16, P 54, P 56<br />

Li, Wenzhong, P 43<br />

Linnemann, Carsten, Or 17<br />

Lipps, Hans J., Or 9, P 60<br />

Loew, Rainer, Or 19<br />

Lucas, Tanja, P 59<br />

Luhmann, Ulrich FO, P 30<br />

Lulay, Christina, Or 7<br />

Lütge, Fabienne, P 52, P 52<br />

Lyko, Frank, P 37<br />

Ma, Ling, Inv 12<br />

Machens, Hans G, P 25, P 34<br />

Magnusson, Terese, P 4<br />

Mailänder, Peter, P 25<br />

Maitland, Norman J, Inv 20<br />

Mantwill, Klaus, P 46, P 49<br />

Maródi, Laszló, Or 7<br />

Marozin, Sabrina, P 44<br />

Mátés, Lajos, Inv 12, P 35<br />

Ma´ trai, Janka, Inv 12<br />

Mattar, Citra, Inv 13<br />

Mätzig, Tobias, Or 15<br />

Mayr, Juliane, P 2<br />

McDonald, Robert A, P 38, P 69<br />

McVey, JH, Or 21<br />

Merten, Otto-Wilhelm, Inv 24<br />

Meyer, Johann, Or 10<br />

Meyerhuber, Peter, P 56<br />

Michalakis, Stylianos, Or 6<br />

Milosevic, Slavoljub, Or 16<br />

Miranda, Paula, P 65<br />

Modlich, Ute, Or 10<br />

Mohr, Andrea, P 48<br />

Momma, Stefan, Or 11<br />

Montazami-Astaneh, Kaweh, P 11<br />

Moore, AT, Inv 14<br />

Muehlebach, Michael D, P 42<br />

Mufti, Ghulam, Inv 23<br />

Mühlfriedel, Regine, Or 6<br />

Muik, Alexander, Or 11<br />

Müller, Oliver J, P 29<br />

Müller-Lerch, Felix, P 17<br />

Müther, Nadine, P 35<br />

Mykhaylyk, Olga M, P 8, P 15, P 53<br />

Naumann, Ulrike, P 49<br />

Naundorf, Sonja, Or 7<br />

Nelson, Peter J, Or 14<br />

Nettelbeck, Dirk M, P 16, P 39<br />

Newrzela, Sebastian, P 31, P 57, P 58,<br />

P 62, P 64<br />

Nicklin, Stuart A, Or 21, P 69<br />

Noble, Alistair, Inv 23<br />

Noske, Nadja, Or 3<br />

Nowrouzi, Ali, Or 7, P 65<br />

Nößner, Elfriede, P 54<br />

Ogris, Manfred, Or 1, Or 5, Or 13, Or 18, P 1, P 4, P<br />

13, P 40, P 41, P 47<br />

Oppenheim, David, Inv 23<br />

Osiak, Anna, P 52<br />

Ott, Michael, P 26<br />

Pachler, Karin, P 2<br />

Paquet-Durand, Franc¸ois, Or 6<br />

Parker, A.L., Or 21<br />

Paruzynski, Anna, Or 7<br />

Perabo, Luca, Or 2<br />

Petkova, Mina, P 64<br />

Petry, Harald, Inv 15<br />

Pfosser, Achim, Or 8, P 28, P 36<br />

Philipp, Alexander, P 6, P 7<br />

Plank, Christian, P 8, P 15, P 33, P 34, P 53, P 55<br />

Pradhan, Pallab, P 8, P 15<br />

Preuß, Ellen, P 66<br />

Pützer, Brigitte M, P 43<br />

Quattrocelli, Mattia, Inv 12<br />

Radecke, Frank, P 52, P 63<br />

Radecke, Sarah, P 52, P 63<br />

Rahim, Ahad A, Inv 13<br />

Rahman, Shamim H, P 11<br />

Rauschhuber, Christina, Inv 1, P 9, P 10, P 20<br />

Reckhenrich, Ann K, P 34<br />

Reidy, Mairead, P 48<br />

Rengstl, Benjamin, P 57, P 58<br />

Robins, Adrian, Inv 23<br />

Rödl, Wolfgang, P 13, P 47<br />

Roeder, Ingo, P 51, P 62, P 64<br />

Rosenecker, Josef, Inv 7<br />

Rubin, GS, Inv 14<br />

Rudolph, Carsten, Inv 4<br />

Ruggiero, Eliana, Or 17<br />

Rupprecht, Sina, P 48, P 60<br />

Ruthardt, Nadia, Or 5, P 3<br />

Rutz, Miriam, P 55<br />

Ruzsics, Zsolt, P 20<br />

Rub, Verena, P 6<br />

Saleh, Amer F, Inv 11<br />

Salguero, Gustavo, P 26<br />

Sallach, Jessica, Or 2<br />

Samara-Kuko, Ermira, Inv 12<br />

Sampaolesi, Maurillio, Inv 12<br />

Sanchez-Antequera, Yolanda, P 53<br />

Sancho-Bru, Pau, Inv 12<br />

Sawyer, Greta, Or 9<br />

Schaffert, David, P 1, P 47<br />

Schambach, Axel, Or 10, Or 15, Or 20, P 12<br />

Schaser, Thomas, P 42<br />

Schendel, Dolores J, Or 16, P 54<br />

Scheu, Christina, P 13<br />

Schiedlmeier, Bernhard, Or 19<br />

Schillinger, Ulrike, P 55<br />

Schinkel, Stefanie, P 29<br />

Schleef, Martin, P 18, P 19<br />

Schmeer, Marco, P 18, P 19<br />

Schmidt, Manfred, Or 7, Or 17, Or 20, P 37, P 65, P<br />

67, P 68<br />

Schmitz, Georg, P 8<br />

Schöler, Hans, Or 15<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong>


Scholz, Simone J, Or 20<br />

Schroeder, Timm, Or 15<br />

Schumacher, Ton NM, Or 17<br />

Schwarz, Klaus, P 52, P 52, P 63<br />

Seeliger, Mathias W, Or 6<br />

Senekowitsch-Schmidtke, Reingard, Or 13, P 47<br />

Sessa, WC, P 38<br />

Seymour, Len W, Inv 6, Inv 9<br />

Seznec, Janina, P 49<br />

Slobodianski, Alex, P 25<br />

Smith, Alexander J, Inv 14, P 30<br />

Spitzweg, Christine, Inv 5, Or 13, Or 14, P 13, P 47<br />

Spranger, Stefani, Or 16<br />

Stachel, Georg, Or 8, P 36<br />

Stärck, Lilian, P 56<br />

Stein, Stefan, Or 20<br />

Steingoetter, Andreas, P 15<br />

Stiess, Michael, P 14<br />

Stockman, A, Inv 14<br />

Stripecke, Renata, Inv 21, P 26<br />

Stroes, E, Inv 15<br />

Su, Baowei, P 40<br />

Su, Haibin, Or 9<br />

Sultana, Shahana, Or 8, P 36<br />

Sundarasetty, Bala Sai, P 26<br />

Tanimoto, Naoyuki, Or 6<br />

Thalhammer, Stefan, P 53<br />

Thielecke, Lars, P 51<br />

Thormann, Michael, P 28<br />

Thrasher, AJ, P 38<br />

Tolmachov, Oleg, P 23<br />

Trenkwalder, Teresa, Or 8, P 36<br />

Tuorto, Francesca, P 37<br />

Turan, Sören, P 12<br />

Turqueti-Neves, Adriana, P 54<br />

Twisk, J, Inv 15<br />

Uckert, Wolfgang, Or 16, P 54, P 56<br />

van den Born, I, Inv 14<br />

van der Hoeven, Franciscus, P 37<br />

van Rooijen, Nico, P 69<br />

VandenDriessche, Thierry, Inv 12<br />

Verfaillie, Catherine, Inv 12<br />

Vermeesch, Joris, Inv 12<br />

Vetter, Alexandra, P 13<br />

Vigh, Balasz, P 33<br />

Virdi, Kulpreet Singh, P 13<br />

Vlasak, Reinhard, P 2<br />

Vlaskou, Dialechti, P 8<br />

Vogt, Stephan, P 50<br />

Voigtlander, Richard, P 20, P 21<br />

Volk, Andreas, P 31, P 32<br />

Volz, Christian, P 29<br />

von Kalle, Christof, Or 7, Or 17, Or 20, P 37, P 65, P<br />

67, P 68<br />

von Laer, Dorothee, Or 11, P 31, P 32, P 57, P 58, P<br />

62, P 64<br />

Waddington, Simon N, Inv 13, P 23<br />

Wagner, Ernst, Or 1, Or 5, Or 14, Or 18, P 1, P 3, P 4,<br />

P 5, P 6, P 7, P 13, P 40, P 41, P 47<br />

Walsch, Florian, P 55<br />

Wang, Wei, P 37<br />

Wang, Weiwei, P 43<br />

Warlich, Eva, Or 15<br />

Weber, Christian, P 65<br />

Weber, Kristoffer, P 66<br />

Wedemeyer, Heiner, P 26<br />

Weingarten, Lisa, P 66<br />

Wells, James, Inv 23<br />

Wester, Hans-Ju¨ rgen, P 46<br />

Wexel, Gabriele, P 50<br />

Wicke, Daniel, Or 10<br />

Wilde, Susanne, Or 16<br />

Willhauck, Michael J, Or 13, Or 14, P 47<br />

Wolf, Anja, P 46<br />

Wolf, Nicola M, P 35<br />

Wong, Andrew MS, Inv 13<br />

Wong, Suet-Ping, P 23, P 24<br />

Wood, Matthew, Inv 11<br />

Wortmann, Andreas, P 59<br />

Wübbenhorst, Daniela, P 50<br />

Wuchrer, Alexander, P 27, P 28<br />

Wunderlich, Nathalie, Or 13, Or 14, P 47<br />

Yan, Bing, Inv 12<br />

Yin, Haifang, Inv 11<br />

Yu, Rui, P 48<br />

Yzer, S, Inv 14<br />

Zach, Christian, Or 14, P 47<br />

Zhang, Wenli, P 35, P 61<br />

Zhang, Xiahong, Or 9<br />

Zhang, Ziyang, P 25<br />

Zimmer, Gert, Or 11<br />

Zimmermann, Martina, P 42<br />

Zong, Xiangang, Or 6<br />

Zwacka, Ralf, P 48<br />

17 th <strong>Annual</strong> <strong>Meeting</strong> of the <strong>German</strong> <strong>Society</strong> <strong>for</strong> <strong>Gene</strong> <strong>Therapy</strong> (DG-GT e.V.), October 7-9, 2010,<br />

<strong>Munich</strong><br />

| 82


Schnellbahnnetz<br />

S8<br />

Flughafen München<br />

<strong>Munich</strong> Airport<br />

Flughafen Besucherpark<br />

S1 S1<br />

Freising<br />

Pulling<br />

S2<br />

Petershausen<br />

R<br />

R<br />

Neufahrn<br />

Eching<br />

Lohhof<br />

Unterschleißheim<br />

Partner im<br />

Vierkirchen-<br />

Esterhofen<br />

Hallbergmoos<br />

Oberschleißheim<br />

Röhrmoos<br />

Altomünster<br />

Kleinberghofen<br />

S2<br />

Hebertshausen<br />

R<br />

Feldmoching<br />

Erdweg<br />

Erding<br />

Ismaning<br />

Garching-<br />

Forschungszentrum<br />

Garching<br />

Garching-Hochbrück<br />

Fröttmaning<br />

Arnbach<br />

S3<br />

Mammendorf<br />

R<br />

Altenerding<br />

Markt Indersdorf<br />

Unterföhring<br />

Aufhausen<br />

Fasanerie<br />

Niederroth<br />

Malching<br />

St. Koloman<br />

R Moosach<br />

Dachau<br />

Karlsfeld<br />

Allach<br />

Schwabhausen<br />

Ottenhofen<br />

Johanneskirchen<br />

Kieferngarten<br />

Freimann<br />

Studentenstadt<br />

Alte Heide<br />

Nordfriedhof<br />

Dietlindenstr.<br />

Münchner Freiheit<br />

Hasenbergl Dülferstr. Harthof Am Hart<br />

Frankfurter Ring<br />

Milbertshofen<br />

Bonner Platz<br />

OberwiesenfeldOlympiazentrumPetuelring<br />

OEZ<br />

Moosacher<br />

St.-Martins-<br />

Platz<br />

Arabellapark<br />

Scheidplatz<br />

Olympiapark<br />

in Bau bis<br />

vsl. Ende 2010<br />

Bachern Dachau Stadt<br />

R<br />

Maisach<br />

Hohenzollernplatz<br />

Markt Schwaben<br />

R<br />

Richard-Strauss-Str.<br />

Georg-Brauchle-<br />

Ring<br />

Westfriedhof<br />

Gernlinden<br />

Poing<br />

Josephsplatz<br />

Böhmerwaldplatz<br />

Chinesischer<br />

Turm<br />

Giselastr.<br />

Englschalking<br />

Untermenzing<br />

Obermenzing<br />

Esting<br />

Grub<br />

Prinzregentenplatz<br />

Olching<br />

Heimstetten<br />

Universität<br />

Odeonsplatz Lehel Max-Weber-Pl.<br />

Theresienstr.<br />

MaillingerStiglmaierstr.platz Gern<br />

Rotkreuzplatz<br />

Gröbenzell<br />

Feldkirchen<br />

Riem<br />

Daglfing<br />

Königsplatz<br />

Messestadt<br />

Ost<br />

Rosenheimer S1<br />

Platz Ostbahnhof<br />

Marienplatz<br />

City Center Isartor<br />

Karlsplatz<br />

(Stachus)<br />

S27<br />

Hackerbrücke<br />

Hauptbahnhof<br />

Central Station<br />

Donnersbergerbrücke<br />

Messestadt<br />

West<br />

Hirschgarten<br />

Laim<br />

Lochhausen<br />

Langwied Pasing<br />

Berg am Laim<br />

Moosfeld<br />

R R<br />

R<br />

R<br />

Messe München<br />

International / ICM<br />

Leuchtenbergring<br />

Trudering<br />

Kreillerstr.<br />

St.-Martin-Str. Josephsburg<br />

Karl-Preis-Platz<br />

S20<br />

Westkreuz<br />

Leienfelsstr.<br />

Aubing<br />

Puchheim<br />

Deutsches<br />

Museum<br />

Gronsdorf<br />

Rathaus<br />

Theresienwiese<br />

Haar<br />

Heimeranplatz<br />

Neuaubing<br />

Harthaus<br />

Eichenau<br />

Untersbergstr.<br />

Silberhornstr.<br />

Kolumbusplatz<br />

Sendlinger<br />

Tor Fraunhoferstr.<br />

Schwanthalerhöhe<br />

Laimer Platz Friedenheimer<br />

Str.<br />

Giesing<br />

Lochham<br />

Germering-<br />

Unterpfaffenhofen<br />

Fürstenfeldbruck<br />

Vaterstetten<br />

Innsbrucker Ring<br />

Michaelibad<br />

Candidplatz<br />

Goetheplatz<br />

I s a r<br />

R<br />

Harras<br />

Westendstr.<br />

Partnachplatz<br />

Gräfelfing<br />

Geisenbrunn<br />

Buchenau<br />

Quiddestraße<br />

Baldham<br />

S6<br />

Zorneding<br />

Poccistr.<br />

Implerstr.<br />

Brudermühlstr.<br />

Perlach<br />

Mittersendling<br />

Obersendling<br />

Westpark<br />

Holzapfelkreuth<br />

Haderner<br />

Planegg<br />

Gilching-<br />

Argelsried<br />

Stern<br />

Schöngeising<br />

Stockdorf<br />

Neugilching<br />

R<br />

Siemenswerke<br />

Grafrath<br />

Eglharting<br />

Neuperlach Zentrum<br />

Therese-Giehse-Allee<br />

Neuperlach Süd<br />

Neubiberg<br />

Ottobrunn<br />

Hohenbrunn<br />

Wettersteinplatz<br />

Aidenbachstr.<br />

Machtlfinger<br />

St.-Quirin-Platz<br />

Thalkirchen (Tierpark)<br />

Fasangarten<br />

Mangfallplatz<br />

Groß-<br />

Str.<br />

hadern Klinikum<br />

Großhadern<br />

Solln<br />

Gauting<br />

Weßling<br />

Fasanenpark<br />

Türkenfeld<br />

Forstenrieder<br />

Allee<br />

Kirchseeon<br />

Geltendorf<br />

R<br />

R<br />

Starnberg<br />

R<br />

Possenhofen<br />

S4<br />

Grafing<br />

Bahnhof<br />

Wächterhof<br />

Höhenkirchen-<br />

Siegertsbrunn<br />

Dürrnhaar<br />

Grafing<br />

Stadt<br />

Aying<br />

Herrsching<br />

S8<br />

Ammersee<br />

Ebersberg<br />

Peiß<br />

R<br />

S4<br />

Großhelfendorf<br />

Kreuzstraße<br />

Starnberger<br />

See<br />

Tutzing<br />

S6<br />

R<br />

R<br />

Großhesselohe Isartalbf.<br />

Unterhaching<br />

Pullach<br />

Taufkirchen<br />

Höllriegelskreuth<br />

Furth<br />

Buchenhain<br />

S20 S27<br />

Baierbrunn<br />

Deisenhofen<br />

Hohenschäftlarn<br />

Innenraum<br />

Sauerlach<br />

Ebenhausen-Schäftlarn<br />

München XXL<br />

Icking<br />

Otterfing<br />

Wolfratshausen<br />

Außenraum<br />

Holzkirchen<br />

S7 S3 R<br />

S7<br />

Starnberg<br />

Nord<br />

Steinebach<br />

Basler Str.<br />

Fürstenried<br />

West<br />

Seefeld-<br />

Hechendorf<br />

Feldafing<br />

Tarifzonen<br />

Regionalzughalt<br />

Fernzughalt<br />

R<br />

MVV/Stand: Dezember 2009

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!