11.01.2013 Views

5th EuropEan MolEcular IMagIng MEEtIng - ESMI

5th EuropEan MolEcular IMagIng MEEtIng - ESMI

5th EuropEan MolEcular IMagIng MEEtIng - ESMI

SHOW MORE
SHOW LESS

Create successful ePaper yourself

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

of the European network of excellence DiMI - Diagnostic Molecular Imaging, and is organised in collaboration with the<br />

ESR - European Society of Radiology, the European network of excellence CliniGene – Clinical Gene Transfer and Therapy,<br />

and is also supported through COST actions D38 and BM0607 – the European cooperation in science and technology.<br />

DAY<br />

0EMIM2010 is the <strong>5th</strong>annual meeting of the European Society for Molecular Imaging - <strong>ESMI</strong>. The EMIM2010 is also the final meeting


WarSaW, poland May 26 – 29, 2010<br />

committees<br />

5 th EMIM EXECUTIVE COMMITTEE<br />

Andreas H. Jacobs, <strong>ESMI</strong> President<br />

Renata Mikołajczak, Local Organiser<br />

Clemens W.G.M. Löwik, <strong>ESMI</strong> Vice President<br />

Bertrand Tavitian, <strong>ESMI</strong> Past President<br />

Silvio Aime, <strong>ESMI</strong> Secretary<br />

Simone Mergui, <strong>ESMI</strong> Treasurer<br />

5 th EMIM SCIENTIFIC COMMITTEE<br />

Ludwig Aigner, Salzburg (A)<br />

Silvio Aime, Torino (I)<br />

Veerle Baekelandt, Leuven (B)<br />

Hervé Boutin, Manchester (Uk)<br />

Harald Carlsen, Oslo (N)<br />

John Clark, Edinburgh (Uk)<br />

Marion Dejong, Rotterdam (Nl)<br />

Silvana Del Vecchio, Naples (I)<br />

Frédéric Dollé, Orsay (F)<br />

Eugeniusz Dzuik, Warsaw (Pl)<br />

Cornel Fraefel, Zuerich (Ch)<br />

Jorgen Frokiaer, Aarhus (Dk)<br />

Nicolas Grenier, Bordeaux (F)<br />

Denis Guilloteau, Tours (F)<br />

Uwe Haberkorn, Heidelberg (D)<br />

Mathias Höhn, Cologne (D)<br />

Andreas H. Jacobs, Münster (D)<br />

Fabian Kiessling, Aachen (D)<br />

Izabela Kozlowicz-Gudzinska, Warsaw (P)<br />

Leszek Krolicki, Warsaw (Pl)<br />

Tony Lahoutte, Jette (B)<br />

Adriaan Lammertsma, Amsterdam (Nl)<br />

5 th EMIM Young Investigator Award SELECTION COMMITTEE<br />

Veerle Baekelandt<br />

Hervé Boutin<br />

Peter Brader<br />

John Clark (Chair)<br />

Frédéric Dollé<br />

Nicolas Grenier<br />

Fabian Kiessling<br />

Adriaan Lammertsma<br />

LOCAL ORGANIZING COMMITTEE<br />

Iliana Chwalinska, Warsaw (P)<br />

Eugeniusz Dzuik, Warsaw (P)<br />

Izabela Kozlowicz-Gudzinska, Warsaw (P)<br />

Leszek Krolicki, Warsaw (P)<br />

Renata Mikolajczak, Warsaw (P)<br />

Bengt Långstrom, (S)<br />

Clemens W.G.M. Löwik, Leiden (Nl)<br />

Helmut R. Maecke, Freiburg (Ch)<br />

Adriana Maggi, Milano (I)<br />

Serge Maitrejean, Paris (F)<br />

Renata Mikolajczak, Warsaw (P)<br />

Chrit Moonen, Bordeaux (F)<br />

Klaas Nicolay, Eindhoven (Nl)<br />

Vasilis Nziachristos,Munich (D)<br />

Sabina Pappata, Naples (I)<br />

David Parker, Durham (Uk)<br />

Bernd PIchler, Tübingen (D)<br />

Anna Planas, Barcelona (E)<br />

Juha Rinne, Turku (Fin)<br />

Jorge Ripoll, Heraklion (Gr)<br />

Markus Rudin, Zürich (Ch)<br />

Michael Schäfers, Münster (D)<br />

Markus Schwaiger, Munich (D)<br />

Bertrand Tavitian, Orsay (F)<br />

Eva Tóth, Orleans (F)<br />

Annemie Van Der Linden, Antwerp (B)<br />

David Wyper, Glasgow (Uk)<br />

Renata Mikolajczak<br />

Chrit Moonen<br />

Klaas Nicolay<br />

Markus Schwaiger<br />

Bertrand Tavitian (Chair)<br />

Eva Tóth<br />

David Wyper


content – per topic<br />

<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

Day 0 - Wednesday 26,2010 6<br />

Overview oral presentations 6<br />

Welcome 8<br />

Venue 10<br />

Thank you! 11<br />

Programme at a Glance 12<br />

Programme Datails 14<br />

Inaugural Lecture by Christopher H. Contag 20<br />

Day 1 - Thursday May 27, 2010 22<br />

<strong>ESMI</strong> Plenary Lecture 1 by Violaine Sée 24<br />

Parallel Session 1: CANCER I - together with the ESR 26<br />

Parallel Session 2: NEUROSCIENCE I 32<br />

Parallel Session 3: TECHNOLOGY 38<br />

Parallel Session 4: PROBES - supported by COST actions 44<br />

<strong>ESMI</strong> Plenary Lecture 2 by Francesca Odoardi 50<br />

Plenary Session on Excellent and Late Breaking Abstracts 52<br />

Day 2 - Friday May 28, 2010 58<br />

<strong>ESMI</strong> Plenary Lecture 3 by Theodorus W.J. Gadella Jr. 60<br />

Parallel Session 5: NEUROSCIENCE II 62<br />

Parallel Session 6: CARDIOVASCULAR I - together with the ESR 68<br />

Parallel Session 7: PROBES II - together with the EANM 74<br />

Parallel Session 8: Gene and cell based therapies - together with CliniGene 80<br />

<strong>ESMI</strong> Plenary Lecture 4 by Hans-Jürgen Wester 86<br />

Plenary Session on Current Contribution of Imaging Technologies to<br />

Drug Development 88<br />

Day 3 - Friday May 29, 2010 92<br />

<strong>ESMI</strong> Plenary Lecture by Jagat Narula 94<br />

Parallel Session 9: CARDIOVASCULAR II 96<br />

Parallel Session 10: CANCER II - supported by COST actions 102<br />

Poster Session, guided poster walks, topics, and titles 108<br />

Index 224<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong>


6<br />

WarSaW, poland May 26 – 29, 2010<br />

overview oral presentations – per title<br />

Inaugural Lecture by Christopher H. Contag<br />

imaging life<br />

Day 0 - Wednesday May 26, 2010<br />

Point-of-Care Microscopy: Molecular Imaging with Cellular Resolution 20<br />

Day 1 - Thursday May 27, 2010<br />

<strong>ESMI</strong> Plenary Lecture 1 by Violaine Sée<br />

Transcription factors dynamics: from discrete pulses to oscillatory pattern to control gene expression 24<br />

Parallel Session 1: CANCER I - together with the ESR<br />

Multimodal imaging approaches for cell-cell interaction 26<br />

Molecular imaging for monitoring treatment with protein kinase inhibitors 27<br />

Potentials of imaging biomarkers in patients 28<br />

Pre-clinical screening of anti - HER2 nanobodies for molecular imaging of breast cancer 29<br />

[18F]-FDG/[18F]-FLT-PET and bioluminescence imaging of therapy response in B-cell lymphoma mice treated with<br />

cytotoxic and antiproliferative agents 30<br />

TF-Pimonidazole: an hypoxia marker suitable for in vivo 19 F MRS imaging 31<br />

Parallel Session 2: NEUROSCIENCE I<br />

[11C](R)-PK11195- a tricky tracer. Do we really need it to image microglial activation? 32<br />

Preclinical imaging of the type 1 cannabinoid receptor in neurodegenerative diseases<br />

Longitudinal non-invasive detection of amyloid plaques by combined molecular, functional and morphological imaging<br />

33<br />

in transgenic mouse models of Alzheimer‘s disease 34<br />

Serotonergic neurotransmission in early Alzheimer’s disease<br />

Qualitative and quantitative assessment of florbetapir F-18 PET (<br />

35<br />

18F-AV45) amyloid deposition PET imaging - validation<br />

by pathology. Preliminary report 36<br />

In-vivo imaging of a mouse traumatic brain injury model using dual-isotope quantitative spect/ct 37<br />

Parallel Session 3: TECHNOLOGY<br />

Molecular analysis of adaptive immune function through microscopic and mesoscopic imaging 38<br />

Magnetic resonance imaging – from structure to molecular interactions 39<br />

Imaging modalities: PET and SPECT 40<br />

In vivo fluorescence kinetic imaging for improved contrast and studies of temporal and quantitative biodistribution 41<br />

Cerenkov radiation imaging of a xenograft murine model of mammary carcinoma 42<br />

A system for 4D (3D+Kinetics) molecular imaging in bioluminescence and fluorescence 43<br />

Parallel Session 4: PROBES - supported by COST actions<br />

The alignment of target-specific lipoCEST MRI contrast agents 44<br />

Lanthanide-based in vivo luminescence imaging 45<br />

New perspectives for imaging molecules and metabolism in vivo 46<br />

In vivo biodistribution of radiolabeled matrix metalloproteinase-2 activatable cell penetrating peptides 47<br />

Bioresponsive MRI contrast agents based on self-assembling β-cyclodextrin nanocapsules 48<br />

Photochemical activation of endosomal escape of MRI-Gd-agents in tumor cells 49<br />

<strong>ESMI</strong> Plenary Lecture 2 by Francesca Odoardi<br />

Immune surveillance and autoimmunity: how encephalitogenic T cells enther their target organ 50<br />

Plenary Session on Excellent and Late Breaking Abstracts<br />

Exendin-4 derivatives labeled with radiometals for the detection of insulinoma: a “from bench to bed approach” 52<br />

Assessment of HIF transcriptional activity in a mouse tumor model using GPI anchored avidin– a novel protein<br />

reporter for in vivo imaging 53<br />

A leukocyte ligand of vascular adhesion protein-1 as an imaging tool in PET 54<br />

Clinical translation of ex vivo sentinel lymph node mapping for colorectal cancer using invisible near-infrared<br />

fluorescence light 55<br />

Development of strategies for in vivo MRI and Optical Imaging of neural stem cells distribution for the treatment of<br />

traumatic spinal cord injury 56<br />

Validation of bone marrow-derived stromal cell graft position by colocalisation of histology, bioluminescence and<br />

magnetic resonance imaging 57<br />

Day 2 - Friday May 28, 2010<br />

<strong>ESMI</strong> Plenary Lecture 3 by Theodorus W.J. Gadella<br />

New probe-based strategies for quantitative microscopy of signaling dynamics in single cells 60


Parallel Session 5: NEUROSCIENCE II<br />

<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

Potential to use transgenic animals for imaging of neurological diseases 62<br />

Specific cell labeling with imaging reporters in vivo by viral vectors: the challenges 63<br />

Optimization of an MRI method to measure microvessel density: application to monitor angiogenesis after stroke 64<br />

MRI study of intra-arterial bone marrow-derived macrophage administration after acute focal ischemic stroke in rats 65<br />

Inactivated paramagnetic tissue plasminogen activator predicts thrombolysis outcome following stroke 66<br />

Matrix metalloproteinase-9 – a possible therapeutic target in intractable epilepsy 67<br />

Parallel Session 6: CARDIOVASCULAR I - together with the ESR<br />

Potentials of new contrast agents for vascular molecular imaging in patients 68<br />

Protease specific nanosensors in atherosclerosis 69<br />

Imaging of inflamed carotid artery atherosclerotic plaques with the use of 99mTc-HYNIC-IL-2 scintigraphy in the<br />

end-stage renal disease patients 70<br />

Uptake of 68 Ga-Chloride in Atherosclerotic Plaques of LDLR -/- ApoB 100/100 Mice 71<br />

Hybrid imaging using dual energy µCT and FMT for characterization of atherosclerotic plaques in ApoE -/- mice 72<br />

Long circulating emulsion based ct contrast agents 73<br />

Parallel Session 7: PROBES II - together with the EANM<br />

Application of microfluidics to the ultra-rapid preparation of fluorine-18 and carbon-11 labelled compounds 74<br />

Radioligand for in vivo measuring neurotransmitters release 75<br />

18f-tracers for amyloid plaques 76<br />

Chelators for radiocopper: biological/pharmacokinetic differences of [Tyr3 ,Thr 8 ]octreotide conjugates 77<br />

[ 11C]SOMADAM: potential sert ligand for pet studies and comparison with [ 11C]MADAM 78<br />

MR imaging of extracellular redox by a thiolsensitive gd(iii)-do3a derivative 79<br />

Parallel Session 8: Gene and cell based therapies - together with CliniGene<br />

Everything but not cell replacement with somatic neural stem cell transplantation 80<br />

Mesenchymal stem cell transplantation: an option to promote remyelination? 81<br />

Viral vector-mediated transcriptional targeting of dendritic cells for antigen-specific tolerance induction in EAE/MS. 82<br />

Delivery of a bioluminescent transgene to a tumor via bone marrow engraftment and local control of gene expression<br />

by non invasive local hyperthermia<br />

Dendritic cell labelling with paramagnetic nanoparticles and<br />

83<br />

111In-oxine for in vivo magnetic resonance imaging and<br />

scintigraphic imaging 84<br />

The type 2 cannabinoid receptor as a new PET reporter gene for the brain 85<br />

<strong>ESMI</strong> Plenary Lecture 4 by Hans-Jürgen Wester<br />

Molecular imaging of CXCR4 receptors 86<br />

Plenary Session on Current Contribution of Imaging Technologies to Drug Development<br />

Evaluation of the temporal window for drug delivery following ultrasound mediated membrane<br />

permeability enhancement 88<br />

Integrisense: a novel near-infrared fluorescent probe for α β integrin and its applications in drug discovery v 3 88<br />

Harnessing the power of bioluminescence to cross the in vitro–in vivo divide 90<br />

In vivo imaging in drug discovery: the example of application in the development of novel estrogenic compounds 91<br />

Day 3 - Saturday May 29, 2010<br />

<strong>ESMI</strong> Plenary Lecture by Jagat Narula<br />

Molecular imaging of unstable coronary plaques 94<br />

Parallel Session 9: CARDIOVASCULAR II<br />

Advances in contrast-enhanced MRI of the mouse heart 96<br />

Molecular imaging of α β integrin expression with v 3 18F-galacto-RGD after experimental myocardial infarction:<br />

comparison with left ventricular remodeling and function 97<br />

Existing and emerging animal models mimicking cardiovascular disease and their relevance for molecular imaging<br />

Imaging of Matrix Metalloproteinase Activity in Vulnerable Human Carotid Plaques with Multispectral Optoacoustic<br />

98<br />

Tomography 99<br />

c-Jun N-terminal kinase promotes inflammation at atherosclerosis-prone sites by enhancing expression and activity<br />

of NF-κB transcription factors 100<br />

Absolute Quantification in Small Animal Pinhole Gated Myocardial Perfusion SPECT 101<br />

Parallel Session 10: CANCER II - supported by COST actions<br />

Cancer imaging 102<br />

Controlled drug delivery under image guidance 103<br />

Comparative biodistribution of twelve gastrin/CCK2 receptor targeting peptides 104<br />

Targeting cancer stem cells using radiolabeled Sonic Hedgehog 105<br />

Evaluation of the photosensitizer Bremachlorin for photodynamic treatment of breast cancer bone metastasis. 106<br />

In vivo targeting of HEK-hsst Xenografts by 2/3/5 111in-labeled [(DOTA)Ser 1 ,Leu 8 ,trp22 ,Tyr25 ]-SS-28 in SCID mice 107<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

overview oral presentations


8<br />

WarSaW, poland May 26 – 29, 2010<br />

Welcome to the 5 th EMIM in Warsaw, Poland<br />

Dear Colleagues,<br />

on behalf of the <strong>ESMI</strong> as well as the other organising and contributing societies and networks it is our great pleasure<br />

to welcome you to Warsaw, Poland, for the 5 th European Molecular Imaging Meeting. EMIM 2010 brings together top<br />

European scientists from 3 societies, 2 European Networks of Excellence (NoEs) and 2 COST actions. With great honour<br />

we may state that your fellow attendees represent not only colleagues from all over Europe, but also Asia and the US.<br />

The societies and networks with primary responsibility for organising the meeting are:<br />

• European Society for Molecular Imaging – <strong>ESMI</strong><br />

• NoE Diagnostic Molecular Imaging – DiMI<br />

• European Cooperation in Science and Technology – COST Actions D38 and BM0607<br />

• European Society of Radiology – ESR<br />

• We also gratefully acknowledge the contribution from the following participating organisations:<br />

• European Association of Nuclear Medicine – EANM<br />

• NoE Clinical Gene Transfer and Therapy – Clinigene<br />

All these organisations were working together, with input from a scientifically complementary scientific steering committee,<br />

to develop a strong scientific programme which integrates developments in imaging technologies and molecular<br />

imaging agents with applications for drug development, basic science investigations, and clinical translation with special<br />

emphasis in the main disease burdens of our society such as cancer, cardiovascular disease and neurodegeneration.<br />

We developed the 5 th EMIM scientific programme primarily on the outstanding strength of submitted abstracts.<br />

About 150 abstracts were submitted, reviewed and scored. Then, each of the concurrent session co-chairs reviewed<br />

the scored abstracts which were considered relevant to their particular sessions. Session co-chairs selected abstracts<br />

for oral presentation based on average abstract score, session content and structure. Attention was also paid to diversity<br />

of disciplines and geographical distribution. From this effort 39 abstracts were integrated into the Parallel as<br />

well as Plenary Sessions that bring attendees from different disciplines together for a comprehensive examination of<br />

the role of molecular imaging in particular biomedical problems. 21 of these oral abstract presentations will be held<br />

by Young Investigator Award (YIA) applicants. The YIA selection committee is going to select the best 3 YIA presentations<br />

to be held again during the final Plenary Session on Saturday where then THE Young Investigator Awardee<br />

will be announced. Furthermore, special attention has been put on seven guided poster walks which have been<br />

scheduled on Thursday and Friday afternoon with no other competing sessions in order to enhance the knowledge<br />

exchange of experienced and young researchers. Each of the guided poster walks will be chaired by two chairs and will<br />

result in one poster award. The seven Poster Awards will also be offered during the Closing Ceremony on Saturday.<br />

We are especially proud and thankful that Prof. Dr. Christopher Contag will give the inaugural lecture,<br />

entitled, “Point-of-care microscopy: molecular imaging with cellular resolution”. In addition, we have planned 5 exciting<br />

plenary lectures, covering topics on probe design, systems biology, cancer, neuroinflammation, and atherosclerosis.<br />

Furthermore, we have dedicated one Plenary Session each to Excellent, Late Breaking, and YIA Abstracts as<br />

well as to Current Contribution of Imaging Technologies to Drug Development.<br />

We would like to take this opportunity to acknowledge the support of this year’s sponsors of the conference, especially<br />

recognizing the continued interest and support from DG Research of the European Commission. Moreover<br />

we would like to thank YOU for participating in this year’s EMIM and making it possible by your attendance. We<br />

hope you will enjoy the conference. If this is not your first time attending an EMIM conference then welcome back<br />

and we hope you will establish new contacts and continue to expand your network of scientific excellence. Please<br />

help first-time attendees to meet others and generally make people feel at home. Regardless whether this is your<br />

first time with us or not we will endeavour to have you come back again in the future.<br />

As in previous years our intention is that this high-level meeting will foster the coherence of a sustainable European<br />

Molecular Imaging Community with the common goal to translate fundamental research discoveries into medical<br />

application and health benefit for the European Society.<br />

Sincerely,<br />

The <strong>5th</strong> Andreas H. Jacobs, <strong>ESMI</strong> President<br />

Renata Mikołajczak, Local Organiser<br />

Clemens W.G.M. Löwik, <strong>ESMI</strong> Vice President<br />

Bertrand Tavitian, <strong>ESMI</strong> Past President<br />

Silvio Aime, <strong>ESMI</strong> Secretary<br />

European Molecular Imaging Meeting Executive Committee<br />

Simone Mergui, <strong>ESMI</strong> Treasurer<br />

imaging life


Dear Colleagues,<br />

<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

it is a pleasure for me to welcome you to the 5 th European Molecular Imaging Meeting in Warszawa. With an impressive<br />

history of previous successful meetings, this time the European Society of Molecular Imaging decided<br />

in favour for Poland, the country belonging to East as well as to Central Europe.<br />

In some way this in-between-situation is also characteristic for the field of MI: the discipline MI is not defined by<br />

a profession, no one is automatically a MI professional. Molecular Imaging is multidisciplinary. This makes it on<br />

the one hand difficult: we have to define ourselves and the direction of our research; it forces us to look beyond<br />

our bench and own disciplines. On the other hand this is also the advantage of MI: it means at any time to be in<br />

process, be in cross-communication and stay open-minded.<br />

Me and my colleagues involved in the organisation of this meeting believe that it will strengthen Molecular Imaging<br />

research activities within Eastern and Central Europe, as well as between the various disciplines working<br />

in the broad field of MI.<br />

The efforts of the <strong>ESMI</strong> to provide a platform for exchange have started to lead into the formation of a European<br />

MI community. This is just the beginning of our common efforts and with our activities and willingness for<br />

cross-interaction we can further establish this development.<br />

We hope that the familiar arrangement of the meeting venue will create a friendly atmosphere and provide a<br />

maximum of opportunities for you to meet friends and exchange views. Enjoy the interesting scientific programme<br />

of invited talks, oral and poster presentations as well as the technical exhibition. I also want to say<br />

thanks to the companies who are supporting this meeting and the MI community; especially to those who come<br />

to Warszawa.<br />

Do not forget that Warszawa is a lively city, with a number of historical sites, museums and parks. We are also in<br />

the year of Chopin; you can find a lot of information about his music and places he used to live. Take some time<br />

to immerse yourself in the history of the city.<br />

On behalf of local organizing committee I wish your visit to Warszawa will be informative, interesting and enjoyable.<br />

Sincerely, Renata Mikołajczak, Local Organiser<br />

Dear Ladies and Gentlemen,<br />

It is my great pleasure to welcome you to the 5 th annual meeting of the European Society for Molecular Imaging.<br />

In my capacity as president of the European Society of Radiology I would like to thank the organisers of this important<br />

event that has been organised in close cooperation with the ESR for their kind invitation.<br />

It is an honour to be here today as the ESR has supported the creation of the European Society for Molecular Imaging<br />

from the very beginning and many ESR representatives have been and are still represented in this organisation.<br />

We are looking forward to enhancing the successful cooperation between the European Society for Molecular Imaging<br />

and the European Society of Radiology in the future.<br />

Realising the importance of molecular imaging, latest research discoveries in this field and possible translations<br />

into medical practice, I will reinforce the topic of molecular imaging within the European Society of Radiology during<br />

my presidency and make sure that the work of all ESR bodies is coordinated with a special focus on this issue.<br />

I would also like to take this opportunity to invite you all to attend ECR 2011 from March 3 to 7 in Vienna, Austria.<br />

The ESR would be most happy to welcome you at its annual meeting.<br />

I thank you for having taken the time to be with us in Warsaw this week and I wish you a most successful meeting.<br />

Sincerely, Professor Maximilian F. Reiser, ESR President<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

Welcome


10<br />

WarSaW, poland May 26 – 29, 2010<br />

Why Warsaw?<br />

Warsaw, the capital of Poland, a captivating city with<br />

a distinctive atmosphere and definitely worth visiting.<br />

Warsaw is a perfect embodiment of changes that<br />

have taken place in Poland in the past 20 years. It is a<br />

city of many faces - a contrasting blend of historical<br />

and socrealist buildings neighbouring post-modern<br />

skyscrapers, of cosy cafes and vibrant clubs, of historical<br />

Żoliborz and artistic Praga district, but most of all<br />

it is a thriving European capital. Whether you come<br />

to Warsaw on a business trip, for a conference, or as a<br />

tourist, Warsaw will definitely exceed your expectations.<br />

Novotel Warsawa Centrum<br />

Marszałkowska 94/98, 00-510 Warszawa<br />

Tel: +48 22 596 25 48 | Fax: +48 22 596 01 22<br />

The Novotel Warsawa Centrum hotel provides excellent<br />

conference facilities. Close to the iconic Palace of Culture<br />

and Science and 500 m from the railway station with a<br />

convenient connection to the airport. The plenary lectures<br />

will take place in ROZA and IRYS, the parallel sessions will<br />

take place in ROZA or IRYS. Visit the exhibition in LILIA,<br />

FREZJA as well as in the FOYER; and get in communication!<br />

The poster sessions will take place in PROMENADA. You are<br />

mostly welcome to view the posters also besides of the<br />

“official” poster sessions.<br />

Floor Plan / Exhibition<br />

1<br />

2<br />

3<br />

coffee break<br />

exhibition and coffee<br />

7 8 9<br />

exhibition and coffee<br />

14 16<br />

15<br />

13<br />

coffee<br />

12<br />

4<br />

5<br />

6<br />

internal meetings<br />

internal meetings<br />

imaging life<br />

11<br />

19<br />

10<br />

lectures<br />

exhibition space 3x2m<br />

20<br />

office<br />

17<br />

How to get there<br />

From OKECIE Airport, take bus route No.175 to Centrum. From<br />

Warszawa Centralna railway station take tram No.7, 8, 9, 22, 24<br />

or 25 to Centrum (one stop). Via the E30, E77 or E67 highways,<br />

head in the direction of Centrum. By metro, alight at Centrum.<br />

Airport : WARSZAWA OKECIE AIRPORT<br />

Railway Station : WARSZAWA CENTRALNA STATION<br />

The Gala dinner will take place on Friday May 28, 2010 at<br />

Zamojski Palace Warsaw 2, Foksal street. Zamojski Palace is<br />

within walking distance. Looking forward to meeting you there.<br />

Floor plan<br />

poster session<br />

lectures<br />

18<br />

1. GEHC<br />

2. ART<br />

3. BIOSCAN/PHILIPS<br />

4. ZINSSER ANALYTIC<br />

5. CaliperLS<br />

6. MR solutions<br />

7. Carestream<br />

8. BRUKER<br />

9. VISUALSONICS<br />

10. SIEMENS<br />

11. BIOSPACE LAB<br />

12. LOT-Oriel<br />

13. CT Imaging<br />

14. SEDECAL<br />

15. VisEn<br />

16. LI-COR<br />

17. SKYSCAN/MILabs<br />

18. Polatom<br />

19. information


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

This meeting would not have been possible without your support.<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

THANK YOU!


WMIC 2010 Speakers<br />

Keynote Address by Shizuo Akira, MD, PhD<br />

Professor of Department of Host Defense, Research Institute for Microbial<br />

Diseases and Director of the WPI Immunology Frontier Research Center<br />

(WPI-IFReC), Osaka University<br />

Featured Plenary Speakers<br />

Kevin Brindle, PhD<br />

Professor, Dept of Biochemistry<br />

University of Cambridge<br />

Robert J. Gropler, MD<br />

Chief, Cardiovascular Imaging Center,<br />

Mallinckrodt Institute of Radiology<br />

Gregory Lanza, MD, PhD<br />

Professor of Medicine<br />

Div. of Cardiovascular Diseases<br />

Washington University School of Medicine<br />

Herman P. Spaink, PhD<br />

Professor of Molecular Cell Biology<br />

Leiden University<br />

Jun Takahashi, MD, PhD<br />

Associate Professor<br />

Center for iPS Cell Research and Application (iCeMS)<br />

Kyoto University, Japan<br />

September 8-11, 2010<br />

Kyoto, Japan<br />

Scientific Sessions<br />

Preliminary List. For full list, visit our website.<br />

In Vivo Studies<br />

* Clinical Studies<br />

* Translational Studies<br />

* Animal Models<br />

Imaging Instrumentation and Methodology<br />

* Hybrid & Multimodality Imaging<br />

* Magnetic Resonance Imaging<br />

* Optical and Opto-acoustic Imaging<br />

* PET/SPECT/CT Imaging<br />

* Ultrasonic Imaging & Drug Delivery<br />

Development/Novel Use of Imaging Probes<br />

* CT<br />

* MRI<br />

* Multi-modality<br />

* Optical and Photoacoustic<br />

* PET/SPECT<br />

* Ultrasound<br />

Imaging Disease/Organ Processes<br />

* Cancer<br />

* Cardiovascular System<br />

* Central Nervous System<br />

* Immune System<br />

Drug and/or Radiation Therapy<br />

* Image Guided Therapy<br />

* Monitoring Therapy<br />

* Novel Therapy Development<br />

REGISTER NOW<br />

September 8-11, 2010<br />

Kyoto, Japan<br />

www.WMICmeeting.org


<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

Wednesday - 26 May 2010 Thursday - 27 May 2010 Friday - 28 May 2010 Saturday - 29 May 2010<br />

room ROZA IRYS ROZA IRYS ROZA IRYS ROZA IRYS<br />

08:00-09:00 Registration Registration<br />

09:00-10:00<br />

<strong>ESMI</strong> Plenary Lecture 1 by Violaine Sée<br />

(Liverpool, UK)<br />

<strong>ESMI</strong> Plenary Lecture 3 by Theodorus W.J.Gadella<br />

(Amsterdam, The Netherlands)<br />

10:00-10:30 Coffee break & Exhibition Coffee break & Exhibition<br />

10:30-12:00<br />

PS 1: Cancer I -<br />

together with ESR<br />

PS 2: Neuroscience I PS 5: Neuroscience II<br />

PS 6: Cardiovascular I -<br />

together with ESR<br />

<strong>ESMI</strong> Plenary Lecture 5 by Klaas Nicolay<br />

(Eindhoven, The Netherlands)<br />

PS 9: Cardiovascular II<br />

12:00-13:00 Lunch break & Exhibition Break<br />

13:00-14:30 PS 3: Technology<br />

14:30-16:00<br />

16:00-17:00<br />

17:00-18:00<br />

Registration<br />

18:00-19:30<br />

Open Ceremony and Inaugural Lecture by<br />

19:00<br />

Christopher H. Contag (Stanford, USA)<br />

19:30 Opening Reception and Exhibition<br />

PS 4: Probes I -<br />

together with COST<br />

PS 7: Probes II<br />

PS 8: Gene and Cell<br />

Based Therapies -<br />

together with CliniGene<br />

Guided Poster Session 1 with Coffee break Guided Poster Session 2 with Coffee break<br />

<strong>ESMI</strong> Plenary Lecture 2 by Francesca Odoardi<br />

(Goettingen, Germany)<br />

<strong>ESMI</strong> Plenary Session on Excellent, Late Breaking<br />

and YIA Abstracts<br />

Sightseeing Tour<br />

<strong>ESMI</strong> Plenary Lecture 4 by Hans-Jürgen Wester<br />

(Munich, Germany)<br />

<strong>ESMI</strong> Plenary Session on Current Contribution of<br />

Imaging Technologies to Drug Development<br />

<strong>ESMI</strong> Executive Committee meets Industry<br />

Gala Dinner at Zamojski Palace<br />

Warsaw 2, Foksal street<br />

Programme at a Glance<br />

PS 10: Cancer II -<br />

together with COST<br />

12:30-13:30<br />

<strong>ESMI</strong> Plenary Session and Closing Ceremony<br />

<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010


WarSaW, poland May 26 – 29, 2010<br />

ROZA IRYS GERBERA<br />

09:00-12:00 Exhibitor’s set up<br />

14:00-18:00 Registration<br />

13:00-15:00<br />

15:00-17:00<br />

imaging life<br />

Posters set up<br />

Speaker’s documents set up<br />

18:00-19:30 Opening Ceremony and Inaugural Lecture<br />

Mariusz Klubczuk plays Chopin:<br />

Nocturne B flat Minor Op.9 No 1; Mazurka C Major Op.24 No 2<br />

Welcome – Andreas H. Jacobs (<strong>ESMI</strong> President)<br />

Welcome – Renata Mikolajczak (EMIM Local Organiser)<br />

Welcome – Andrzey Siemaszko (NCP in Poland for EU research)<br />

Welcome – Maximilian Reiser (ESR President)<br />

Announcement of <strong>ESMI</strong> Award 2010, John Clark (<strong>ESMI</strong> Award Winner 2009)<br />

Mariusz Klubczuk plays Chopin:<br />

Etude C sharp Minor Op. 25 No 7; Etude A Minor Op. 25 No 12<br />

Inaugural lecture by Christopher H. Contag (Stanford, USA)<br />

Point-of-Care Microscopy: Molecular Imaging with Cellular Resolution<br />

19:30-21:00 Opening Reception and Exhibition<br />

DiMI WP Meetings<br />

(Closed Meetings)<br />

<strong>ESMI</strong> Executive Committee<br />

(Closed Meeting)<br />

<strong>ESMI</strong> Council Meeting<br />

(Closed Meeting)<br />

ROZA IRYS GERBERA<br />

08:00-08:45 DiMI SMB (Closed Meeting)<br />

08:00-09:00 Registration<br />

09:00-10:00<br />

<strong>ESMI</strong> Plenary Lecture 1 by Violaine Sée (Liverpool, UK)<br />

Transcription factors dynamics: from discrete pulses to oscillatory pattern to control gene expression<br />

Co-Chairs: Clemens WGM Löwik (Leiden, The Netherlands), Leszek Krolicki (Warsaw, Poland)<br />

10:00-10:30 Coffee break & Exhibition<br />

10:30-12:00<br />

Parallel Session 1: Cancer I (together with ESR)<br />

Co-Chairs: Markus Schwaiger (Munich, Germany),<br />

Peter Brader (Vienna, Austria)<br />

10:30-10:45 Multimodal imaging approaches for cell-cell interaction<br />

Steffen Maßberg, (Munich, Germany)<br />

10:45-11:00 Molecular imaging for monitoring treatment with<br />

protein kinase inhibitors<br />

Wolfgang Weber (Freiburg, Germany)<br />

11:00-11:15 Potentials of imaging biomarkers inpatients<br />

Bernard Van Beers (Paris, France)<br />

11:15-11:30 YIA Applicant’s Presentation: Pre-clinical screening of anti - her2<br />

nanobodies for molecular imaging of breast cancer<br />

Ilse Vaneycken (Brussels, Belgium)<br />

11:30-11:45 YIA Applicant’s Presentation: [18f]-fdg/[18f]-flt-pet and<br />

bioluminescence imaging of therapy response in b-cell lymphoma<br />

mice treated with cytotoxic and antiproliferative agents,<br />

Marijke De Saint-Hubert (Leuven, Belgium)<br />

11:45-12:00 tf-pimonidazole: an hypoxia marker suitable for in vivo 19f mrs<br />

imaging<br />

Arend Heerschap (Nijmegen, The Netherlands)<br />

12:00-13:00 Lunch break<br />

Day 0 - Wednesday May 26, 2010<br />

Day 1 - Thursday May 27, 2010<br />

YIA Selection Committee<br />

(Closed Meeting)<br />

Parallel Session 2: Neuroscience I<br />

Co-Chairs: Annemie van der Linden (Antwerp, Belgium),<br />

David Wyper (Glasgow, UK), Hervé Boutin (Manchester, UK)<br />

[11C](R)-PK11195- a tricky tracer. Do we really need it to image<br />

microglial activation?<br />

Alex Gerhard (Manchester, UK)<br />

YIA Applicant’s Presentation: Preclinical imaging of the type 1<br />

cannabinoid receptor in neurodegenerative diseases<br />

Cindy Casteels (Leuven, Belgium)<br />

YIA Applicant’s Presentation: Longitudinal non-invasive detection of<br />

amyloid plaques by combined molecular, functional and morphological<br />

imaging in transgenic mouse models of alzheimers disease,<br />

Florian C. Maier (Tübingen, Germany)<br />

Serotonergic neurotransmission in early Alzheimer’s disease,<br />

Steen G. Hasselbalch (Copenhagen, Denmark)<br />

Qualitative and quantitative assessment of florbetapir f-18 pet (18fav45)<br />

amyloid deposition PET imaging - validation by pathology.<br />

preliminary report<br />

Grzegorz Romanowicz (Gdańsk, Poland)<br />

YIA Applicant’s Presentation: In-vivo imaging of a mouse traumatic<br />

brain injury model using dual-isotope quantitative SPECT/CT<br />

Domokos Máthé (Budapest, Hungary)<br />

TOPIM2011 committee<br />

(Closed meeting)


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

Parallel Session 3: Technology<br />

13:00-14:30 Co-Chairs: Jorge Ripoll (Heraklion, Greece),<br />

Markus Rudin (Zürich, Switzerland)<br />

13:00-13:15 Molecular analysis of adaptive immune function through<br />

microscopic and mesoscopic imaging<br />

Jens Stein (Bern, Switzerland)<br />

13:15-13:30 Magnetic resonance imaging – from structure to molecular interactions<br />

Markus Rudin (Zürich, Switzerland)<br />

13:30-13:45 Imaging modalities: PET/SPECT<br />

Sibylle Ziegler (Munich, Germany)<br />

13:45-14:00 In vivo fluorescence kinetic imaging for improved contrast and<br />

studies of temporal and quantitative biodistribution<br />

James Mansfield (Cambridge, UK)<br />

14:00-14:15 Cerenkov radiation imaging of a xenograft murine model of<br />

mammary carcinoma<br />

Federico Boschi (Verona, Italy)<br />

14:15-14:30 A system for 4d (3d+kinetics) molecular imaging in<br />

bioluminescence and fluorescence<br />

Serge Maitrejean (Paris, France)<br />

14:30-16:00<br />

16:00-17:00<br />

17:00-18:00<br />

ROZA IRYS GERBERA<br />

Parallel Session 4: Probes I (together with COST action D38)<br />

Co-Chairs: Eva Jakab-Tóth (Orléans, France),<br />

Silvio Aime (Torino, Italy)<br />

The alignment of target-specific lipoCEST MRI contrast agents<br />

Sander Langereis (Eindhoven, The Netherlands)<br />

Lanthanide-based in vivo luminescence imaging<br />

Stephane Petoud (Orléans, France and Pittsburgh, US)<br />

New perspectives for imaging molecules and metabolism in vivo<br />

Rolf Gruetter (Lausanne, Switzerland)<br />

YIA Applicant’s Presentation: In vivo biodistribution of radiolabeled<br />

matrix metalloproteinase-2 activatable cell penetrating peptides<br />

Sander Van Duijnhoven (Eindhoven, The Netherlands)<br />

YIA Applicant’s Presentation: Bioresponsive mri contrast agents<br />

based on self-assembling beta-cyclodextrin nanocapsules<br />

Jonathan Martinelli (Alessandria, Italy)<br />

Photochemical activation of endosomal escape of MRI-gd-agents in<br />

tumor cells<br />

Eliana Gianolio (Torino, Italy)<br />

Guided Poster Session 1 with Coffee break (see page 110)<br />

Poster Walk 1: Imaging Cancer Biology (P001-P012) & (P105-P106)<br />

Co-Chairs: Fabian Kiessling (Aachen, Germany), Markus Rudin (Zürich, Switzerland)<br />

Poster Walk 2: Technology & Data Analysis Methods (P070-P078) & (P097-P104)<br />

Co-Chairs: Serge Maitrejean (Paris, France), Adriaan Lammertsma (Amsterdam, The Netherlands)<br />

Poster Walk 3: Molecular Neuroimaging (P034-P047) & (P107)<br />

Co-Chairs: Sabina Pappata (Naples, Italy), Andreas H. Jacobs (Münster, Germany)<br />

Poster Walk 4: Probe Design (P058-P069)<br />

Co-Chairs: Frédéric Dollé (Orsay, France), Helmut Maecke (Freiburg, Germany)<br />

<strong>ESMI</strong> Plenary Lecture 2 by Francesca Odoardi (Göttingen, Germany)<br />

Immune surveillance and autoimmunity: how encephalitogenic T cells enter their target organ<br />

Co-Chairs: Renata Mikolajczak (Warsaw, Poland), Chrit Moonen (Bordeaux, France)<br />

Plenary Session on Excellent, Late Breaking, and Young Investigator Award Abstracts<br />

Co-Chairs: Renata Mikolajczak (Warsaw, Poland), Chrit Moonen (Bordeaux, France)<br />

17:00-17:10 Exendin-4 derivatives labeled with radiometals for the detection of insulinoma: a “from bench to bed approach”<br />

Damian Wild (Freiburg, Germany)<br />

17:10-17:20 YIA Applicant’s Presentation:<br />

Assessment of hif transcriptional activity in a mouse tumor model using gpi anchored avidin-<br />

a novel protein reporter for in vivo imaging<br />

Steffi Lehmann (Zürich, Switzerland)<br />

17:20-17:30 YIA Applicant’s Presentation:<br />

A leukocyte ligand of vascular adhesion protein-1 as an imaging tool in PET<br />

Anu Autio (Turku, Finland)<br />

17:30-17:40 YIA Applicant’s Presentation:<br />

Clinical translation of ex vivo sentinel lymph node mapping for colorectal cancer using invisible near-infrared fluorescence light<br />

Merlijn Huttemann (Leiden, The Netherlands)<br />

17:40-17:50 YIA Applicant’s Presentation:<br />

Development of strategies for in vivo MRI and optical imaging of neural stem cells distribution for the treatment of traumatic<br />

spinal cord injury<br />

Ramona Lui (Milano, Italy)<br />

17:50-18:00 Validation of bone marrow-derived stromal cell graft position by colocalisation of histology,<br />

bioluminescence and magnetic resonance imaging<br />

Nathalie de Vocht (Antwerp, Belgium)<br />

18:15-18:45<br />

19:00-20:00<br />

DiMI General Assembly<br />

(Closed Meeting)<br />

DiMI WP Meeting<br />

(Closed Meeting)<br />

DiMI WP Meeting<br />

(Closed Meeting)<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong>


08:00-08:45<br />

WarSaW, poland May 26 – 29, 2010<br />

imaging life<br />

ROZA IRYS GERBERA<br />

DiMI WP Meeting<br />

(Closed Meeting)<br />

08:00-09:00 Registration<br />

09:00-10:00<br />

<strong>ESMI</strong> Plenary Lecture 3 by Theodorus W.J. Gadella Jr. (Amsterdam, Netherlands)<br />

New probe-based strategies for quantitative microscopy of signaling dynamics in single cells<br />

Co-Chairs: Vasilis Nziachristos (Munich, Germany), Bengt Långström (Uppsala, Sweden)<br />

10:00-10:30 Coffee break & Exhibition<br />

10:30-12:00<br />

Parallel Session 5: Neuroscience II<br />

Co-Chairs: Markus Rudin (Zürich, Switzerland),<br />

Veerle Baekelandt (Leuven, Belgium),<br />

Mathias Hoehn (Cologne, Germany)<br />

10:30-10:45 Potential to use transgenic animals for imaging of neurological<br />

diseases<br />

Ludwig Aigner (Salzburg, Austria)<br />

10:45-11:00 Specific cell labeling with imaging reporters in vivo by viral vectors:<br />

the challenges<br />

Veerle Baekelandt (Leuven, Belgium)<br />

11:00-11:15 YIA Applicant’s Presentation: Optimization of an MRI method to<br />

measure microvessel density: application to monitor<br />

angiogenesis after stroke<br />

Philipp Boehm-Sturm (Cologne, Germany)<br />

11:15-11:30 MRI study of intra-arterial bone marrow-derived macrophage<br />

administration after acute focal ischemic stroke in rats<br />

Adrien Riou (Lyon, France)<br />

11:30-11:45 YIA Applicant’s Presentation: Inactivated paramagnetic tissue<br />

plasminogen activator predicts thrombolysis<br />

outcome following stroke<br />

Maxime Gauberti (Caen, France)<br />

11:45-12:00 Matrix metalloproteinase-9 – a possible therapeutic target<br />

in intractable epilepsy<br />

Grzegorz Wilczynski (Warsaw, Poland)<br />

12:00-13:00 Lunch Break<br />

13:00-14:30<br />

Parallel Session 7: Probes II<br />

(together with EANM)<br />

Co-Chairs: Frédéric Dollé (Orsay, France) ,<br />

Denis Guilloteau (Tours, France)<br />

13:00-13:15 Application of microfluidics to the ultra-rapid preparation of<br />

fluorine-18 and carbon-11 labelled compounds<br />

Philip Miller (London, UK)<br />

13:15-13:30 Radioligand for in vivo measuring neurotransmitters release,<br />

Christer Halldin (Stockholm, Sweden)<br />

13:30-13:45 18F-tracers for Amyloid plaques<br />

Denis Guilloteau (Tours, France)<br />

13:45-14:00 YIA Applicant’s Presentation: Chelators for radiocopper: biological/<br />

pharmacokinetic differences of [tyr3,thr8]octreotide conjugates,<br />

Abiraj Keelara (Basel, Switzerland)<br />

14:00-14:15 [11c]SOMADAM: potential sert ligand for PET studies<br />

and comparison with [11c]MADAM<br />

Fabienne Gourand (Caen, France)<br />

14:15-14:30 MR imaging of extracellular redox by a thiolsensitive<br />

gd(iii)-do3a derivative<br />

Giuseppe Digilio (Torino, Italy)<br />

Day 2 - Friday May 28, 2010<br />

<strong>ESMI</strong> Exec. Com. & <strong>ESMI</strong> Council<br />

Meeting (Closed Meeting)<br />

together with Treasurer<br />

Parallel Session 6: Cardiovascular I (together with ESR)<br />

Co-Chairs: Tony Lahoutte (Mons, Belgium),<br />

Nicolas Grenier (Bordeaux, France)<br />

Potentials of new contrast agents for vascular molecular imaging in<br />

patients<br />

Philippe Douek (Lyon, France)<br />

Protease specific nanosensors in atherosclerosis<br />

Eyk Schellenberger (Berlin, Germany)<br />

YIA Applicant’s Presentation: Imaging of inflamed carotid artery<br />

atherosclerotic plaques with the use of 99mtc-hynic-il-2 scintigraphy<br />

in the end-stage renal disease patients<br />

Marta Opalinska (Krakow, Poland)<br />

Uptake of 68ga-chloride in atherosclerotic plaques of ldlr-/apob100/100mice<br />

Johanna Silvola (Turku, Finland)<br />

YIA Applicant’s Presentation: Hybrid imaging using dual energy µct<br />

and fmt for characterization of atherosclerotic<br />

plaques in apoe -/- mice<br />

Felix Gremse (Aachen, Germany)<br />

YIA Applicant’s Presentation: Long circulating emulsion<br />

based CT contrast agents<br />

Anke De Vries (Eindhoven, The Netherlands)<br />

<strong>ESMI</strong> Training Committee<br />

(Closed Meeting)<br />

Parallel Session 8: Gene and Cell Based Therapies<br />

(together with Clinigene)<br />

Co-Chairs: Ludwig Aigner (Salzburg, Austria),<br />

Cornel Fraefel (Zürich, Switzerland)<br />

Everything but not cell replacement with neural<br />

stem cell transplants<br />

Stefano Pluchino (Milano, Italy)<br />

Mesenchymal Stem Cell Transplantation: An Option to Promote<br />

Remyelination? Ludwig Aigner (Salzburg, Austria)<br />

Viral vector-mediated transcriptional targeting of dendritic cells for<br />

antigen-specific tolerance induction in EAE/MS<br />

Christiane Dresch (Zürich, Switzerland)<br />

Delivery of a bioluminescent transgene to a tumor via bone marrow<br />

engraftment and local control of gene<br />

expression by non invasive local hyperthermia<br />

Pierre-Yves Fortin (Bordeaux, France)<br />

YIA Applicant’s Presentation: Dendritic cell labelling with<br />

paramagnetic nanoparticles and 111in-oxine for in vivo magnetic<br />

resonance imagingand scintigraphic imaging<br />

Cristina Martelli (Milan, Italy)<br />

YIA Applicant’s Presentation: The type 2 cannabinoid receptor<br />

as a new PET reporter gene for the brain<br />

Caroline Vandeputte (Leuven, Belgium)


14:30-16:00<br />

16:00-17:00<br />

17:00-18:00<br />

<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

ROZA IRYS GERBERA<br />

Guided Poster Session 2 with Coffee break (see page 110)<br />

Poster Walk 5: Imaging in Cancer & Drug Development (P013-P030)<br />

Co-Chairs: Peter Brader (Graz, Austria), Nicolas Grenier (Bordeaux, France)<br />

Poster Walk 6: Imaging in Other Diseases (P031-P033) & (P079-P091)<br />

Co-Chairs: Bertrand Tavitian (Orsay, France), John Clark (Edingburgh, UK)<br />

Poster Walk 7: Imaging-guided Gene and Cell based and Targeted Therapies (P048-P057) & (P092-P096)<br />

Co-Chairs: Ludwig Aigner (Salzburg, Austria), Adriana Maggi (Milano, Italy)<br />

<strong>ESMI</strong> Plenary Lecture 4 by Hans-Jürgen Wester (Munich, Germany)<br />

Molecular Imaging of CXCR4 Receptors<br />

Co-Chairs: Adriana Maggi (Milano, Italy), Adriaan Lammertsma (Amsterdam, The Netherlands)<br />

Plenary Session on Current Contribution of Imaging Technologies to Drug Development<br />

Co-Chairs: Adriana Maggi (Milano, Italy), Adriaan Lammertsma (Amsterdam, The Netherlands)<br />

17:00-17:15 Evaluation of the temporal window for drug delivery following ultrasound mediated membrane permeability enhancement<br />

Matthieu Lepetit-Coiffe (Bordeaux, France)<br />

17:15-17:30 Integrisense: a novel near-infrared fluorescent probe for a v b 3 integrin and its applications in drug discovery<br />

Cyrille Sur, Merck Research Laboratories (Westpoint, USA)<br />

17:30-17:45 Harnessing the power of bioluminescence to cross the in vitro–in vivo divide<br />

John Watson, PROMEGA (Madison, Wisconsin, USA)<br />

17:45-18:00 In vivo imaging in drug discovery: the example of application in the development of novel estrogenic compounds<br />

Andrea Biserni, TOP Srl (Milano, Italy)<br />

18:00-19:00<br />

19:30 Gala Dinner at Zamojski Palace<br />

08:00-08:45 <strong>ESMI</strong> General Assembly (Closed Meeting)<br />

09:00-10:00<br />

<strong>ESMI</strong> Executive Committee<br />

meets Industry<br />

(Closed Meeting)<br />

ROZA IRYS GERBERA<br />

<strong>ESMI</strong> Plenary Lecture 5 by Klaas Nicolay (Eindhoven, The Netherlands)<br />

Multi-modality molecular imaging of atherosclerosis, usingtargeted contrast agents<br />

Co-Chairs: Uwe Haberkorn (Heidelberg, Germany), Helmut Maecke (Freiburg, Germany)<br />

10:00-10:30 Coffee break & Exhibition YIA Selection Committee<br />

10:30-12:00<br />

Parallel Session 9: Cardiovascular II<br />

Co-Chairs: Klaas Nicolay (Eindhoven, The Netherlands),<br />

Michael Schäfers (Münster, Germany)<br />

10:30-10:45 Advances in contrast-enhanced MRI of the mouse heart<br />

Gustav Strijkers (Eindhoven, The Netherlands)<br />

10:45-11:00 Molecular imaging of α v β 3 integrin expression with 18F-galacto-<br />

RGD after experimental myocardial infarction: comparison with left<br />

ventricular remodeling and function<br />

Antti Saraste (Turku, Finland/Munich, Germany)<br />

11:00-11:15 Existing and emerging animal models mimicking cardiovascular<br />

disease and their relevance for molecular imaging<br />

Michael Schäfers (Münster, Germany)<br />

11:15-11:30 Imaging of matrix metalloproteinase activity in vulnerable human<br />

carotid plaques with multispectral optoacoustic tomography<br />

Daniel Razansky (Munich, Germany)<br />

11:30-11:45 C-jun n-terminal kinase promotes inflammation at atherosclerosisprone<br />

sites by enhancing expression and activity<br />

of nf-κb transcription factor<br />

Paul Evans (London, UK)<br />

11:45-12:00 YIA Applicant’s Presentation: Absolute quantification in small<br />

animal pinhole gated myocardial perfusion SPECT<br />

Lode Goethals (Brussels, Belgium)<br />

Parallel Session 10: Cancer II(with COST action BM0607)<br />

Co-Chairs: Marion de Jong (Rotterdam, The Netherlands),<br />

Fabian Kiessling (Aachen, Germany)<br />

Cancer Imaging<br />

Uwe Haberkorn (Heidelberg, Germany)<br />

Controlled Drug Delivery under Image guidance<br />

Holger Grüll (Eindhoven, The Netherlands)<br />

Targeted radionuclide imaging and therapy (COST Action BM0607),<br />

Marion de Jong (Rotterdam, The Netherlands)<br />

Targeting cancer stem cells using radiolabeled sonic hedgehog,<br />

Izabela Tworowska (Houston, USA)<br />

YIA Applicant’s Presentation: Evaluation of the photosensitizer<br />

bremachlorin for photodynamic treatment of<br />

breast cancer bone metastasis<br />

Pieter Van Driel (Leiden, The Netherlands)<br />

In vivo targeting of hek-hsst2/3/5 xenografts by 111in-labeled<br />

[(dota)ser1,leu8,trp22,tyr25]-ss-28 in scid mice<br />

Theodosia Maina (Athens, Greece)<br />

12:00-12:30 Break YIA Selection Committee<br />

12:30-13:30<br />

<strong>ESMI</strong> Plenary Session and Closing Ceremony and Young Investigator Award Presentations<br />

Young Investigator Awards & Poster Awards Ceremony<br />

Co-Chairs: John Clark (Edinburgh, UK), Bertrand Tavitian (Orsay, France)<br />

Day 3 - Saturday May 29, 2010<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong>


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

Do not forget:<br />

Join the next <strong>ESMI</strong> Winter Conference – TOPIM2011:<br />

further information/pre-registration<br />

send mail to office@ e-smi.eu<br />

WWW.E-SMI.Eu<br />

HOT TOPIC 2011: Emerging image-guided methods in Medicine<br />

Jan. 16-21, 2011 in Les Houches, France &<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

MY NOTES fl


20<br />

WarSaW, poland May 26 – 29, 2010<br />

Dr. Contag is an Associate Professor of Pediatrics in the Division of Neonatal and Developmental Medicine,<br />

and a member of the BioX faculty at Stanford University. He is the Director of the Stanford Infrared Optical<br />

Science and Photomedicine Program, director of Stanford’s Center for Innovation in In Vivo Imaging (SCI3)<br />

and co-director of the Molecular Imaging Program at Stanford (MIPS). Dr. Contag received his B.S. in<br />

Biology from the University of Minnesota, St. Paul in 1982; and earned his Ph.D. in Microbiology from the<br />

University of Minnesota, Minneapolis in 1988. He was a postdoctoral fellow at Stanford University from<br />

1990-1994, and joined the faculty in Pediatrics at Stanford in 1995 with a joint appointment in Microbiology<br />

and Immunology and a courtesy appointment in Radiology.<br />

Dr. Contag is a pioneer in the emerging field of molecular imaging and is developing imaging approaches<br />

aimed at revealing molecular processes in living subjects and advancing therapeutic strategies through<br />

imaging. His laboratory develops macroscopic and microscopic optical imaging tools and uses imaging to<br />

assess tissue responses to stress, reveal immune cell migration patterns, understand stem cell biology and<br />

advance biological therapies. He is a founding member, and a past president, of the Society for Molecular<br />

Imaging, and for his fundamental contributions in imaging, is a recipient of the Achievement Award from<br />

the Society for Molecular Imaging. Dr. Contag is a scientific founder of Xenogen Corp. – now Caliper<br />

LifeSciences. He is also a founder of ConcentRx Corp.<br />

Selected References<br />

• Cao, Y-A, Stevenson, DKS, Weissman, I, Contag, CH. 2008. Heme Oxygenase-1 Deficiency leads to disrupted response<br />

to acute stress in stem cells and progenitors. Blood. 112: 4494-4502<br />

• Banaszynski, LA, Sellmyer, MA, Thorne, SH, Contag, CH, Wandless, TJ. 2008. Chemical control of protein stability and<br />

function in living mice. Nature Med. 14 (10):1123-7.<br />

• Wender, PA, Goun, EA, Jones, LR, Pillow, TH, Rothbard, JB, Shinde, R, Contag, CH. 2007. Real-time analysis of uptake<br />

and bioactivatable cleavage of luciferin-transporter conjugates in transgenic reporter mice. Proc Nat Acad Sci, USA.<br />

104(25):10340-5.<br />

• Thorne SH, Negrin, RS, Contag, CH. 2006. Synergistic antitumor effects of immune cell-viral biotherapy. Science. 311:1780-1784.<br />

• Cao, Y-A, Wagers, A, Beilhack, A, Dusich, J, Bachmann, MH, Negrin, RS, Weissman, IL, Contag, CH. 2004. Shifting foci of<br />

hematopoeisis during reconstitution from single stem cells. Proc. Natl. Acad. Sci. USA. 101(1):221-226.<br />

• Hardy, JK, Chu, P, Gibbs, K, Contag, CH. 2004. Extracellular replication of Listeria monocytogenes in the gall bladder.<br />

Science. 303(5659): 851-853<br />

• Contag, CH and Bachmann, MH. 2004. The writing is on the vessel wall. Nature. 429:618-620.<br />

• Shachaf, CM, Kopelman, A, Arvanitis, C, Beer, S, Mandl, S, Bachmann, MH, Borowsky, AD, Ruebner, B, Cardiff, RD, Yang,<br />

Q, Bishop, JM, Contag, CH and Felsher, DW. 2004. MYC inactivation uncovers stem cell properties and induces the state<br />

of tumor dormancy in hepatocellular cancer. Nature, 431:1112-1117.<br />

• Contag, PR, Olomu, IN, Stevenson, DK, Contag, CH. 1998. Bioluminescent indicators in living mammals. Nature Med.<br />

4(2):245-247.<br />

imaging life<br />

Christopher H. Contag


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

Point-of-Care Microscopy: Molecular Imaging with Cellular Resolution<br />

Contag C.H. .<br />

Division of Neonatal and Developmental Medicine, and a member of the BioX faculty at Stanford University.<br />

ccontag@stanford.edu<br />

Micro-optical designs are enabling the development<br />

of miniaturized microscopes that can reach inside<br />

the body to interrogate disease states microscopically.<br />

This is leading to an emerging field of in vivo<br />

pathology that is changing the diagnostic paradigm<br />

from biopsy<br />

and conventional<br />

histopathology to<br />

one of point-ofcare<br />

histopathology<br />

coupled with telepathology.<br />

These<br />

advances are closing<br />

the gap between<br />

micro–mirror<br />

the patient and the<br />

pathologist and<br />

have the potential of accelerating diagnosis and<br />

guiding therapy. While recent advances in this<br />

field have been significant, many issues must be<br />

resolved before this clinical transformation may be<br />

fully realized. There are technological and translational<br />

advances that are driving this field and are<br />

leading towards in vivo microscopy becoming a standard<br />

clinical tool. By removing the spatiotemporal<br />

separation between the pathologist and patient, we<br />

can accelerate clinical diagnosis and advance clinical<br />

care for patients with a wide variety of diseases.<br />

References<br />

1. Liu, JTC, Mandella, MJ, Loewke, NO, Haeberle, H, Ra, H,<br />

Piyawattanametha, W, Solgaard, O, Kino, GS, Contag,<br />

CH (2010) Micromirror-scanned dual-axis confocal<br />

microscope utilizing a gradient-index relay lens for<br />

image guidance during brain surgery. JBO. In Press.<br />

2. Piyawattabanetha, W, Ra, H, Mandella, MJ, Loewke,<br />

Wang, TD, Kino, GS, Solgaard, O, Contag, CH (2009) 3-D<br />

near-infrared fluorescence imaging using a MEMSbased<br />

miniatuire dual axis confocal microscope. IEEE J.<br />

Sel. Topics Quantum Electronics. 15(5): 1344-1350.<br />

3. Mackanos, MA, Hargrove, J, Wolters, R, Du, CB,<br />

Friedland, S, Soetikno, RM, Contag, CH, Arroyo, MR,<br />

Crawford, JM, Wang, TD (2009) Use of an endoscopecompatible<br />

probe to detect colonic dysplasia with<br />

Fourier transform infrared spectroscopy. J Biomed<br />

Optics 14, 044006. PMID: 19725718<br />

4. Liu, JT, Mandella, MJ, Crawford, JM, Contag, CH, Wang,<br />

TD, Kino, GS. (2008) Efficient rejection of scattered light<br />

enables deep optical sectioning in turbid media with<br />

low-numerical-aperture optics in a dual-axis confocal<br />

architecture. J Biomed Opt. 13(3):034020.<br />

5. Hsiung, P-L, Hardy, JW, Friedland, S, Soetikno, R, Du, CB,<br />

Wu, APW, Sahbaie, P, Crawford, JM, Lowe, AW, Contag,<br />

CH, Wang, TD. (2008) Detection of colonic dysplasia in<br />

vivo using a targeted fluorescent septapeptide and<br />

confocal microendoscopy. Nat. Med. 14(4): 454-8.<br />

6. Wang, TD, Triadafilopoulos, G, Crawford, JM, Dixon, LR,<br />

Bhandari, T, Sahbaie, P, Friedland, S, Soetikno, R, Contag,<br />

CH. (2007) Detection of Endogenous Biomolecules<br />

in Barrett’s Esophagus by Fourier Transform Infrared<br />

Spectroscopy. Proc. Natl. Sci, USA. 104(40): 15864-9.<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

day0<br />

Inaugural Lecture by Christopher H. Contag


DAY<br />

1


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

• <strong>ESMI</strong> Plenary Lecture 1: Violaine Sée (Liverpool, UK)<br />

Transcription factors dynamics: from discrete pulses to oscillatory pattern to control gene expression<br />

Chairs: Clemens W.G.M. Löwik (Leiden, The Netherlands) , Leszek Krolicki (Warsaw, Poland)<br />

• Parallel Session 1: Cancer I – together with ESR<br />

Chairs: Markus Schwaiger (Munich, Germany), Peter Brader (Vienna, Austria)<br />

• Parallel Session 2: Neuroscience I<br />

Chairs: Annemie van der Linden (Antwerp, Belgium), David Wyper (Glasgow, UK),<br />

Hervé Boutin (Manchester, UK)<br />

• Parallel Session 3: Technology<br />

Chairs: Jorge Ripoll (Heraklion, Greece) , Bernd Pichler (Tuebingen, Germany)<br />

• Parallel Session 4: Probes I – together with COST<br />

Chairs: Eva Jakab-Tóth (Orléans, France) , Silvio Aime (Torino, Italy)<br />

• <strong>ESMI</strong> Plenary Lecture 2: Francesca Odoardi (Göttingen, Germany)<br />

Immune surveillance and autoimmunity: how encephalitogenic T cells enter their target organ<br />

Chairs: Renata Mikolajczak (Warsaw, Poland), Chrit Moonen (Bordeaux, France)<br />

• Plenary Session on Excellent and Late Breaking Abstracts<br />

Chairs: Renata Mikolajczak (Warsaw, Poland), Chrit Moonen (Bordeaux, France)<br />

Day 1 - Thursday May 27, 2010<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

day1<br />

Day 1 - Thursday May 27, 2010


24<br />

WarSaW, poland May 26 – 29, 2010<br />

Violaine Sée is a BBSRC David Phillips Research Fellow at the University of Liverpool. She graduated in<br />

Chemistry and Molecular and Cellular Biology at the University Louis Pasteur in Strasbourg (France). After a<br />

Master in Pharmacology, in 2001 she obtained her PhD in Pharmacology and Neurobiology at the University<br />

Louis Pasteur. She was then assistant lecturer and subsequently moved to the University of Liverpool as a<br />

Post-doctoral Research Fellow. In 2005, she obtained a prestigious David Phillips Fellowship, to develop<br />

her work on intracellular signalling dynamics. She is focusing on the imaging of single living cells in order<br />

to understand regulation of gene transcription and cell fate. She has recently been interested in using new<br />

techniques for single molecule imaging in live cells based on the use of gold nanoparticles.<br />

Selected References<br />

• Nelson, D. E., Ihekwaba, A. E., Elliott, M., Johnson, J. R., Gibney, C. A., Foreman, B. E., Nelson, G., See, V., Horton, C. A.,<br />

Spiller, D. G., Edwards, S. W., McDowell, H. P., Unitt, J. F., Sullivan, E., Grimley, R., Benson, N., Broomhead, D., Kell, D. B.,<br />

and White, M. R. (2004) Oscillations in NF-kappaB signaling control the dynamics of gene expression. Science 306,<br />

704-708<br />

• See, V., Rajala, N. K., Spiller, D. G., and White, M. R. (2004) Calcium-dependent regulation of the cell cycle via a novel<br />

MAPK--NF-kappaB pathway in Swiss 3T3 cells. The Journal of cell biology 166, 661-672<br />

• Nelson, D. E., See, V., Nelson, G., and White, M. R. (2004) Oscillations in transcription factor dynamics: a new way to<br />

control gene expression. Biochemical Society transactions 32, 1090-1092<br />

• Ashall, L., Horton, C. A., Nelson, D. E., Paszek, P., Harper, C. V., Sillitoe, K., Ryan, S., Spiller, D. G., Unitt, J. F., Broomhead, D.<br />

S., Kell, D. B., Rand, D. A., See, V., and White, M. R. (2009) Pulsatile stimulation determines timing and specificity of NFkappaB-dependent<br />

transcription. Science 324, 242-246<br />

• See, V., Free, P., Cesbron, Y., Nativo, P., Shaheen, U., Rigden, D. J., Spiller, D. G., Fernig, D. G., White, M. R., Prior, I. A., Brust,<br />

M., Lounis, B., and Levy, R. (2009) Cathepsin L digestion of nanobioconjugates upon endocytosis. ACS nano 3, 2461-<br />

2468<br />

• Meley, D., Spiller, D.G., White, M.R., McDowell, H., Pizer, B.L., and See, V. (2010) p53-mediated delayed NF-kB activity<br />

enhances etoposide-induced cell death in medulloblastoma. Cell Death and Disease 1, In press<br />

imaging life<br />

Violaine Sée


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

Transcription factors dynamics: from discrete pulses to oscillatory pattern to control gene<br />

expression<br />

Sée V.<br />

Centre for Cell Imaging, University of Liverpool<br />

violaine@liverpool.ac.uk<br />

At a given time-point, cells in a population are<br />

heterogeneous in their functions and fate and it is<br />

therefore vital to develop and apply methods that<br />

allow the measurement of dynamic molecular processes<br />

in single cells. We have shown, using single<br />

cell imaging, the critical role of nucleo-cytoplasmic<br />

localisation oscillations of the NF-κB transcription<br />

factor to control downstream pattern of gene transcription<br />

(Nelson et al, Science 2004; Ashall et al,<br />

Science 2009). NF-κB regulates cellular stress and<br />

immune responses to infection. In most cases, oscillations<br />

had previously been masked in population<br />

level studies by cellular heterogeneity. We developed<br />

a protocol based on cell treatment by repeated short<br />

pulses of TNFα at various intervals to mimic pulsatile<br />

inflammatory signals. This allowed obtaining<br />

synchronous cycles of NF-κB nuclear translocation.<br />

We have also observed cell to cell heterogeneity in<br />

other signalling systems such as in cellular response<br />

to low oxygen environment (hypoxia). In both inflammatory<br />

and hypoxic signalling systems one<br />

source of heterogeneity is due to the presence of extrinsic<br />

dynamic processes that are functionally coupled<br />

and that are occurring over different time scales.<br />

We identified the cell cycle as one source of variability<br />

as cells must coordinate and prioritise their<br />

response to the environment depending on their<br />

cell cycle status. We apply mathematical modelling<br />

using the quantitative data generated by imaging<br />

experiments to predict the role of the negative feedback,<br />

to unravel new network motifs and to characterise<br />

the cross-talk between the signalling systems.<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

day1<br />

<strong>ESMI</strong> Plenary Lecture 1 by Violaine Sée


26 Cell-cell<br />

WarSaW, poland May 26 – 29, 2010<br />

Multimodal imaging approaches for cell-cell interaction<br />

Massberg S.<br />

Deutsches Herzzentrum München, Technische Universität München<br />

massberg@idi.harvard.edu<br />

interactions play a pivotal role in the control<br />

of critical cellular functions, such as cell adhesion,<br />

cell migration as well as cell differentiation and<br />

proliferation. The scientific focus of our lab is on<br />

the mechanisms that regulate cell-cell interactions<br />

during scenarios, such as arterial or venous thrombosis,<br />

stem cell homing and differentiation, during<br />

inflammation as well as in cancer development and<br />

progression. To evaluate the interaction between<br />

different cell subsets in their physiological (micro-)<br />

environment, we have established novel imaging approaches<br />

that allow dissecting all the steps involved<br />

both in reductionist in vitro assays and in vivo.<br />

imaging life<br />

This includes the used of gene-targeted mice, in<br />

which distinct cellular lineages are genetically<br />

marked in combination with innovative imaging<br />

techniques, including intravital multi-photon microscopy.<br />

The in vivo approaches are complemented<br />

by sophisticated in vitro assays allowing investigation<br />

of cell-cell interactions on a subcellular and<br />

molecular level. The multimodal imaging approaches<br />

will contribute to a better understanding of the<br />

molecular mechanisms and the kinetic aspects of<br />

the dynamic process of cell-cell interactions under<br />

physiological conditions and in the disease states.


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

Molecular imaging for monitoring treatment with protein kinase inhibitors<br />

Weber W.<br />

University Freiburg, Germany<br />

wolfgang.weber@uniklinik-freiburg.de<br />

Targeted drugs that modulate the function of specific<br />

molecules in diseased tissues hold great promise for<br />

the treatment of many diseases including malignant<br />

tumors. However, there are several challenges for<br />

the efficient evaluation of these drugs in clinical<br />

trials as well as for the use in clinical practice.<br />

These include<br />

(i) the selection of patients likely to benefit from<br />

treatment with a specific targeted drug,<br />

(ii) finding the right dose and dose schedule,<br />

(iii) monitoring target inhibition, and<br />

(iv) assessing tumor response to therapy.<br />

Standard anatomic imaging continues to play an<br />

important role for addressing these challenges,<br />

but molecular imaging provides several new<br />

opportunities to make the use of targeted drugs more<br />

efficient. Using molecular imaging the expression<br />

of drug targets can be assessed non-invasively, the<br />

concentration of drugs can be measured in the<br />

tumor tissue, target inhibition can be monitored and<br />

tumor response to therapy can be evaluated earlier<br />

than with anatomic imaging techniques.<br />

Therefore it is expected that molecular imaging<br />

will play an increasing role for guiding molecularly<br />

defined therapeutic interventions.<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

day1<br />

Parallel Session 1: CANCER I - together with the ESR


28 Introduction:<br />

WarSaW, poland May 26 – 29, 2010<br />

Potentials of imaging biomarkers in patients<br />

Van Beers B.E.<br />

Department of Radiology and INSERM U 773, Beaujon University Hospital, University Paris Diderot, France.<br />

bernard.van-beers@bjn.aphp.fr<br />

There is an increasing need for imaging<br />

biomarkers to objectively assess the prognosis and<br />

response to treatment in cancer.<br />

Methods: Quantitative anatomical, molecular, and<br />

functional characteristics can be used as imaging<br />

biomarkers.<br />

Results: The RECIST criteria are anatomical imaging<br />

biomarkers that are accepted surrogate endpoints<br />

in cancer treatment. However, they are not early<br />

biomarkers and are limited in the assessment of<br />

some tumors and targeted treatments.<br />

The use of molecular imaging biomarkers is<br />

restricted by their narrow target specificity and<br />

by the complexity of the biology in tumors. In a<br />

given tumor type, several different subgroups often<br />

exist with overexpression of different proteins and<br />

signaling pathways.<br />

Therefore, it is often preferred to assess downstream<br />

changes of molecular pathways with functional<br />

imaging life<br />

imaging. The rationale of this approach is that<br />

most cancers have acquired the same functional<br />

capabilities including upregulated glycolysis, limitless<br />

proliferation, extensive angiogenesis, resistance to<br />

apoptosis and metastasis formation. These capabilities<br />

are assessed with functional biomarkers using PET,<br />

dynamic contrast-enhanced CT and MRI, diffusionweighted<br />

MRI, MR elastography and spectroscopy.<br />

Conclusions: Functional biomarkers have an<br />

important potential to help in selecting the patients<br />

and assessing the response to new treatments,<br />

especially in phase 1 and 2 clinical trials.<br />

However, important efforts of validation and<br />

standardization remain to be done before the wide<br />

use of functional biomarkers and their acceptance<br />

as surrogate endpoints that can replace clinical<br />

endpoints such as survival or time to progression.<br />

References:<br />

1. Hanahan D, Weinberg RA. The hallmarks of cancer. Cell.<br />

2000;100(1):57-70.<br />

2. Rudin M. Imaging readouts as biomarkers or surrogate<br />

parameters for the assessment of therapeutic<br />

interventions. Eur Radiol. 2007 (10):2441-57.<br />

3. Padhani AR, Liu G, Koh DM, Chenevert TL, et al.<br />

Diffusion-weighted magnetic resonance imaging as a<br />

cancer biomarker. Neoplasia. 2009;11(2):102-25.<br />

4. Van Beers BE, Vilgrain V. Biomarkers in abdominal<br />

imaging. Abdom Imaging. 2009;34(6):663-7.


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

Pre-clinical screening of anti - HER2 nanobodies for molecular imaging of breast cancer<br />

Vaneycken I. (1) , Devoogdt N. (1) , Peleman C. (1) , Xavier C. (1) , Lahoutte T. (1) , Caveliers V. (1) .<br />

Vrije Universiteit Brussel, Belgium<br />

ilse.vaneycken@gmail.com<br />

Introduction: Nanobodies are small antigen-binding<br />

fragments of camelid heavy-chain antibodies. Besides<br />

therapeutic agents, they are also effective as a<br />

diagnostic tool for targeted imaging when labeled<br />

with a suitable radionuclide[1]. We produced Nanobodies<br />

for molecular imaging of Human Epidermal<br />

Growth Factor Receptor 2 (HER2) expression in<br />

breast cancer patients[2] for a phase I clinical trial.<br />

After production, a number of candidate binders<br />

are obtained. we designed a standardized selection<br />

procedure based on production yield, in vitro and<br />

in vivo criteria to identify the lead anti-HER2 Nanobody<br />

with the highest potential as a tracer for clinical<br />

translation.<br />

Methods: A camel was immunized with HER2 recombinant<br />

protein and serum IgG2 and IgG3 subclasses<br />

with heavy-chain-only antibodies were<br />

separated. Eleven different anti-HER2 Nanobodies<br />

were produced in E. coli, purified and labeled with<br />

99mTc(I)-tricarbonyl. In vitro saturation binding<br />

studies were performed on recombinant HER2 and<br />

HER2 positive SKOV3 cells with the native and radiolabeled<br />

anti-HER2 Nanobodies. Competition<br />

studies allowed assessing whether the anti-HER2<br />

99mTc-Nanobodies competed with the therapeutic<br />

anti-HER2 antibodies Trastuzumab and Pertuzumab.<br />

In vivo, the biodistribution and tumor targeting<br />

potential of all 99mTc-Nanobodies was evaluated<br />

using pinhole SPECT/micro-CT in nude mice bearing<br />

HER2 positive SKOV3 xenografts. The Nanobody<br />

showing the most favourable in vivo characteristics<br />

was additionaly evaluated in nude mice<br />

bearing HER2 positive LS174T and HER2 negative<br />

MDAMB4357d xenografts.<br />

Results: Nanobodies were labeled with 99mTc and<br />

purified to a final radiochemical purity of ≥99%.<br />

Saturation binding studies showed that 99mTc labeling<br />

was not associated with a significant reduction of<br />

immunoreactivity. The Nanobodies appeared to be<br />

not or only weakly competitive with the commercial<br />

therapeutic antibodies. In vivo biodistribution demonstrated<br />

that tumor accumulation varied between<br />

0,78 and 4,44 percent injected activity per gram<br />

(%IA/g). Of the eleven 99mTc-Nanobodies tested in<br />

SKOV3 xenografts, three presented a tumor uptake<br />

above 4 %IA/g. One was selected and also showed<br />

high tumor uptake (3,76±0,82 %IA/g) in LS174T xenografts,<br />

but low uptake in MDAMB435d xenografts<br />

(0,71±0,07 %IA/g). In addition, all 99mTc-Nanobodies<br />

displayed high renal uptake but low non-specific<br />

accumulation in liver, muscle and blood, resulting<br />

in high tumor-to-background ratios.<br />

Conclusions: Using a standardized selection procedure<br />

for identificiation of high affinity Nanobodies<br />

for molecular imaging of cancer, we identified one<br />

Nanobody as the lead compound for a phase I clinical<br />

trial. This Nanobody meets with all criteria that<br />

characterize a good diagnostic tracer.<br />

Acknowledgement: The research at ICMI is funded<br />

by the Interuniversity Attraction Poles Program–<br />

Belgian State–Belgian Science Policy. Tony Lahoutte<br />

is a Senior Clinical Investigator of the Research<br />

Foundation–Flanders (Belgium) (FWO).<br />

References:<br />

1. Gainkam LO, Huang L, Caveliers V, et al. Comparison of<br />

the biodistribution and tumor targeting of two 99mTclabeled<br />

anti-EGFR nanobodies in mice, using pinhole<br />

SPECT/micro-CT. J Nucl Med. 2008;49(5):788-795.<br />

2. Hicks,D.G. et al (2008). HER2+ breast cancer: review of<br />

biologic relevance and optimal use of diagnostic tools.<br />

American Journal of Clinical Pathology, 129, 263-273.<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

YIA applicant<br />

day1<br />

Parallel Session 1: CANCER I - together with the ESR


YIA applicant<br />

30 Introduction:<br />

WarSaW, poland May 26 – 29, 2010<br />

[18F]-FDG/[18F]-FLT-PET and bioluminescence imaging of therapy response in B-cell<br />

lymphoma mice treated with cytotoxic and antiproliferative agents<br />

De Saint-Hubert M. (1) , Brepoels L. (1) , Devos E. (1) , Degroot T. (1) , Ibrahimi A. (1) , Tousseyn T. (1) , Verbruggen A. (1) ,<br />

Mortelmans L. (1) , Mottaghy F. (2) .<br />

(1) KULeuven, Belgium<br />

(2) RWTH Aachen.<br />

marijke.desainthubert@med.kuleuven.be<br />

Early and reliable evaluation of therapy<br />

outcome is essential for optimal medical care<br />

in oncology. Shortly after treatment the influx of<br />

inflammatory cells can interfere with [18F]-FDG<br />

uptake while [18F]-FLT is less hampered by this<br />

phenomenon. The aim of this study was to image<br />

[18F]-FDG and [18F]-FLT response to either cytotoxic<br />

or antiproliferative therapy and to correlate the<br />

response to the amount of viable cells assessed with<br />

Bioluminescence Imaging (BLI).<br />

Methods: Daudi cells (Burkitt lymphoma) were<br />

transduced with a lentiviral vector (blasticidin selection<br />

marker and firefly luciferase reporter gene) and<br />

inoculated in SCID-mice (n=57). When the tumors<br />

reached a diameter of 15mm2 animals were treated<br />

with either cyclophosphamide (125mg/kg, n=25)<br />

or temsirolimus (50mg/kg, n=25). [18F]-FDG and<br />

[18F]-FLT small-animal PET was performed on<br />

day0 (d0, before treatment) day (d) 2, 4, 7, 9 and 14.<br />

On these days we also imaged the tumor cells with<br />

BLI. At each time point, 2 mice of each treatment<br />

condition were sacrificed and tumors were excised<br />

for histopathology (H&E, Ki-67, CD20, TUNEL).<br />

Quantitative imaging data were expressed as % signal<br />

of the baseline signal measured on d0 and expressed<br />

as mean ± standard error of the mean (SEM).<br />

Results: [18F]-FDG uptake decreased immediately<br />

(-38% on d2) after cyclophosphamide treatment<br />

without a significant reduction in [18F]-FLT uptake<br />

which could be due to DNA repair as DNA FACS<br />

showed an increased amount of cells in the S-fase<br />

early after cyclophosphamide. From d7 [18F]-FLT<br />

uptake decreased significanlty corresponding to histology<br />

data (ki-67). [18F]-FDG uptake stabilized between<br />

d7 and d9 (-57% and -59% respectively) likely<br />

due to a high influx of inflammatory cells observed<br />

in histology (±40% on d7-d9). Caliper measurements<br />

showed tumor shrinkage from d7. Surprisingly the<br />

BLI measured an increased signal early after therapy<br />

(d2-d4) while only late after therapy a reduction in<br />

the signal was observed (d9-d14).<br />

Temsirolimus treatment reduced both the [18F]-<br />

FDG and [18F]-FLT uptake immediately (d2) after<br />

therapy. [18F]-FDG stabilized between d9 and d14<br />

imaging life<br />

maybe due to inflammation which was only observed<br />

on d14 but this might as well be due to regrowth. The<br />

early [18F]-FLT decrease corresponded well to ki-67<br />

stainings and to DNA FACS where an increase of the<br />

G0/G1 fase is observed. Tumor size reduced already<br />

from d4 after temsirolimus treatment. BLI showed<br />

the same signal increase as cyclophosphamide early<br />

after therapy and only in a late stage after therapy<br />

we observed a reduced amount of viable cells. This<br />

early increase might be due to an upregualtion of the<br />

promotor after therapy but further evalution of this<br />

phenomenon is on the way.<br />

Conclusions: [18F]-FDG reduced early after therapy<br />

but stabilized due to a temporary rise in inflammatory<br />

cells. [18F]-FLT-PET was able to detect a<br />

reduced proliferation after therapy. However, after<br />

cytotoxic therapy [18F]-FLT did not change probably<br />

due to DNA repair. Therefore caution should be<br />

made when imaging [18F]-FLT response immediately<br />

after cytotoxic therapy. Bioluminescence imaging<br />

showed an early increase which may have important<br />

implication for BLI monitoring of therapy.<br />

Acknowledgement: This work was financially supported<br />

by the Center of Excellence ‘MoSAIC’ of the<br />

K.U.Leuven.


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

TF-Pimonidazole: an hypoxia marker suitable for in vivo 19 F MRS imaging<br />

Heerschap, A. .<br />

Centre for Molecular Life Science, University Nijmegen, The Netherlands<br />

a.heerschap@rad.umcn.nl<br />

Introduction: Hypoxia in tumours is associated<br />

with enhanced progression, increased aggressiveness<br />

and metastatic potential and poor prognosis.<br />

Moreover, hypoxic tumour cells are resistant to<br />

radiotherapy and some forms of chemotherapy.<br />

Over the years several approaches to assess hypoxia<br />

in vivo have been explored, ranging from<br />

needles measuring local pO 2 invasively to a range<br />

of non-invasive (imaging) methods using oxygen<br />

sensitive agents. Unfortunately until today none<br />

of these methods or agents has entered widespread<br />

clinical practice. Here we demonstrate a<br />

novel hypoxia marker completely analogous to<br />

the commonly used histological hypoxia marker<br />

Pimonidazole (PIMO) and labelled with fluorine<br />

for in vivo 19 F MRS imaging.<br />

Methods:1-(2-nitro-1H-imidazol-1-yl)-3-[4-<br />

(trifluoromethyl)piperidin-1-yl]propan-2-ol<br />

(TF-PIMO) was synthesized in a similar way as<br />

described in [1]. C57BL/6 mice carrying a C38<br />

colon carcinoma on the upper leg (size approx.<br />

250 mm 3 ) were given either 80 or 200 mg / kg TF-<br />

PIMO intraperitoneal (IP) at least 3 hours before<br />

MR investigations. The mice were anesthetized<br />

using a single urethane IP injection. This avoids<br />

any spectral interference by fluorinated inhalation<br />

anaesthetics. Experiments were performed<br />

on a 7 T horizontal bore MR system. A homemade<br />

14 mm solenoid coil was used for transmit/receive<br />

of 19 F and 1 H. After initial localization and basic<br />

1 H MR imaging (T2*w GRE) an unlocalized pulse<br />

acquire sequence (FID) on 19 F was used to detect<br />

TF PIMO validating correct injection of the compound.<br />

Subsequently a series of 3D 19 F chemical<br />

shift imaging (CSI) FID experiments using an<br />

ultrashort adiabatic half passage pulse was used<br />

for TF-PIMO 19 F imaging. Further settings of the<br />

MRSI sequence: FOV 32 × 32 ×32 mm, matrix<br />

8 × 8 × 8, TR 597 ms, acquisition time 58 m 02<br />

s, 256 averages, weighted phase encoding scheme.<br />

After MR, tumours were removed immediately<br />

and stored in liquid nitrogen. Frozen tumour sections<br />

of 5 μm thickness were cut for staining and<br />

further analysis. The tumor sections were subsequently<br />

stained and scanned for Hoechst and rabbit<br />

anti-pimonidazole.<br />

Results: Figure 1 shows a heat map generated from<br />

the 3D 19 F MRSI representing the in vivo TF-PIMO<br />

accumulation in a hypoxic C38 colon carcinoma as<br />

was validated by subsequent immunohistochemical<br />

analysis of the tumour tissue.<br />

Figure 1: 1H MRI of a C38 murine colon carcinoma on a mouse leg<br />

with a heat map generated from a 3D 19F MRSI representing the in<br />

vivo TF-PIMO accumulation over-layed.<br />

Conclusions: TF-PIMO was synthesized and shown<br />

to be a potential non-invasive marker to image tissue<br />

hypoxia in tumours by 19F MRSI. Background<br />

free functional images are obtained that can be coregistered<br />

with conventional MRI. In addition this<br />

marker has the advantage that it can be stained with<br />

the same anti-body as used to detect the common<br />

clinical used marker Pimonidazole and thus allows<br />

for easy matching of hypoxia by histology and by<br />

MR on the same tumour sample.<br />

Acknowledgement: This work is supported in part<br />

by EMIL (LSHC-CT-2004-503569) and NWO (VIS-<br />

TA and INV911-06-021)<br />

References:<br />

1. Raleigh, J. A. et al; Magn Reson Med 22:451-466 (1991)<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

day1<br />

Parallel Session 1: CANCER I - together with the ESR


32 Detailed<br />

WarSaW, poland May 26 – 29, 2010<br />

[11C](R)-PK11195- a tricky tracer. Do we really need it to image microglial activation?<br />

Gerhard A. .<br />

Wolfson Molecular Imaging Centre Manchester, UK<br />

alex.gerhard@manchester.ac.uk<br />

experimental and human post mortem<br />

studies have shown that activation of microglia, the<br />

brain’s resident tissue macrophages, has a particularly<br />

close association with active brain pathology. One<br />

of the molecules expressed de novo during the<br />

‘activation’ of microglia is the translocator protein<br />

(18kDa), TSPO.PK11195 is a selective ligand for the<br />

TSPO.<br />

In vivo and in the absence of invading blood-borne<br />

cells the de novo expression of TSPO occurs primarily<br />

in activated microglia. Based on this relative cellular<br />

selectivity [11C]PK11195 PET has been used to<br />

image microglial activation in vivo for about 20 years<br />

now and it has been possible to demonstrate patterns<br />

of increased signal distribution in the brain that<br />

correspond well with the localisation of pathological<br />

changes in a number of neurological disorders (1).<br />

Recently [11C](R)- PK11195 PET has even<br />

successfully been used to show in vivo the effect of a<br />

pharmacological intervention in a neurodegenerative<br />

disorder on microglial activation (2).<br />

imaging life<br />

Nevertheless there are methodological challenges<br />

when using [11C]PK11195 particularly an<br />

unfavourable signal to noise ratio. Therefore<br />

numerous new ligands for the TSPO have been<br />

evaluated at different stages over the last years<br />

with some of them having also been assessed in<br />

diseases known to be accompanied by microglial<br />

activation (3).<br />

The talk will give a brief overview of relevant studies<br />

with PK11195, as well as newer TSPO tracers, and<br />

will try to highlight their potential advantages and<br />

problems.<br />

References:<br />

1. Venneti S, Lopresti BJ, Wiley CA. The peripheral<br />

benzodiazepine receptor (Translocator protein<br />

18kDa) in microglia: from pathology to imaging. Prog<br />

Neurobiol. 2006;80(6):308-322. Epub 2006 Dec 2006.<br />

2. Dodel R, Spottke A, Gerhard A, et al. Minocycline 1-year<br />

therapy in multiple-system-atrophy: Effect on clinical<br />

symptoms and [(11)C] (R)-PK11195 PET (MEMSA-trial).<br />

Mov Disord. 2010;25(1):11.<br />

3. Chauveau F, Boutin H, Van Camp N, Dolle F, Tavitian<br />

B. Nuclear imaging of neuroinflammation: a<br />

comprehensive review of [(11)C]PK11195 challengers.<br />

Eur J Nucl Med Mol Imaging. 2008;1:1.


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

Preclinical imaging of the type 1 cannabinoid receptor in neurodegenerative diseases<br />

Casteels C. (1) , Van Laere K. (1) .<br />

University Leuven, Division of Nuclear Medicine, Belgium<br />

cindy.casteels@med.kuleuven.be<br />

The endocannabinoid system (ECS) is an important<br />

modulatory system in the brain. It consists of a family<br />

of naturally occurring lipids, the endocannabinoids,<br />

of transport and degradation proteins, and of<br />

cannabinoid receptors. Type 1 cannabinoid (CB1)<br />

receptors are abundantly expressed in all brain areas,<br />

especially those involved in the control of motor<br />

function. CB1 receptor stimulation modulates<br />

GABA, glutamate and dopamine neurotransmitter<br />

release in a dynamic activity manner.1<br />

Dysregulation of cannabinoid-mediated control<br />

of basal ganglia function have been suggested to<br />

play a critical role in the pathogenesis and symptom<br />

development of Parkinson’s disease (PD) and<br />

Huntington’s disease (HD), providing rationale for<br />

potential ECS-targeted therapy in these diseases.2<br />

However, at present, limited clinical pilot trials have<br />

been inconclusive. As only in vitro, ex vivo and<br />

post mortem data on the ECS existed until recently,<br />

functional in vivo imaging may play an important<br />

role in the further evaluation of the neurobiological<br />

and clinical impact of the ECS in PD and HD.<br />

Here, we present the in vivo characterization of CB1<br />

receptor alterations in preclinical models of PD and<br />

HD using PET and [18F]MK-94703. Both genetic<br />

and toxin-based experimental models will be presented.<br />

Their relevance to mimic the human condition<br />

will be discussed as well.<br />

Acknowledgement: Financial support of the Research<br />

Council of the Katholieke Universiteit Leuven<br />

(OT/05/58), the Fund for Scientific Research,<br />

Flanders, Belgium (FWO/G.0548.06), and the Institute<br />

for the Promotion of Innovation by Science and<br />

Technology in Flanders (SBO50151) is gratefully<br />

acknowledged. This work is performed under European<br />

Commission FP6-project DiMI,LSHB-CT-<br />

2005-512146<br />

References:<br />

1. Katona et al., Nat. Med. 2008;<br />

2. Maccarrone et al., Prog. Neurobiol. 2007;<br />

3. Burns et al., Proc. Natl. Acad. Sci. U.S.A. 2007.<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

YIA applicant<br />

day1<br />

Parallel Session 2: NEUROSCIENCE I


YIA applicant<br />

34<br />

WarSaW, poland May 26 – 29, 2010<br />

Longitudinal non-invasive detection of amyloid plaques by combined molecular, functional<br />

and morphological imaging in transgenic mouse models of Alzheimers Disease<br />

Maier F. C. (1) , Wehrl H. F. (1) , Schmid A. (1) , Odenthal J. (2) , Reischl G. (3) , Wiehr S. (1) , Mannheim J. (1) , Stiller D. (4) , Jucker M. (2) ,<br />

Pichler B. (1) .<br />

(1) Laboratory for Preclinical Imaging and Imaging Technology of the Werner Siemens-Foundation,<br />

(2) Hertie Institute for Clinical Brain Research,<br />

(3) University Hospital Tuebingen, Radiopharmacy,<br />

(4) Drug Discovery Support, Boehringer Ingelheim Pharma GmbH & Co. KG.<br />

florian.maier@med.uni-tuebingen.de<br />

Introduction: Current small animal imaging<br />

instrumentation provides powerful tools to study<br />

disease characteristics and progression. However,<br />

biomarkers for imaging amyloid plaque deposition<br />

and associated physiological changes are not yet<br />

fully understood – impeding diagnosis in daily<br />

clinics. Thus our aims were to assess the ongoing<br />

plaque deposition in an animal model of Alzheimers<br />

Disease longitudinally using [11C]PIB, to compare<br />

the binding properties in different transgenic<br />

mouse models and to monitor perfusion differences<br />

with [15O]H2O and Arterial Spin Labeling (ASL) –<br />

reflecting disease induced changes in physiology.<br />

Methods: APPPS1 tg mice and Tg2576 mice<br />

with respective littermate controls were injected<br />

intravenously with 7.6±2.8 MBq [11C]PIB (specific<br />

activity > 50GBq/µmol) and with 28.5±3 MBq<br />

[15O]H2O for measurement of cerebral perfusion.<br />

Dynamic small animal PET scans were performed<br />

for 1h p.i. and the mice were anesthetised with<br />

1.5% isofluran in 100% oxygen. In addition, 3D MR<br />

images and ASL were acquired for each mouse. The<br />

PET images were analysed using cortical regions<br />

as target and the cerebellum as internal reference.<br />

The obtained time activity curves were processed<br />

with the Logan Plot and the simplified reference<br />

tissue model (SRTM) by Lammertsma. ASL-MRI<br />

data were analyzed with an inhouse programmed<br />

Matlab routine using a simplified version of the<br />

Bloch equation. Furthermore, the animals were<br />

analysed histopathologically.<br />

Results: Both analysis methods revealed<br />

significant differences in the binding potential<br />

(BP) of [11C]PIB in cortical regions of transgenic<br />

APPPS1 mice (Logan 0.28±0.10; SRTM 0.32±0.18)<br />

in comparison to littermate controls (Logan<br />

0.13±0.07; SRTM 0.05±0.07, n=8, p


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

Serotonergic neurotransmission in early Alzheimer’s disease<br />

Hasselbalch S. G. (1) , Marner L. (1) , Madsen K. (1) , Lehel S. (1) , Barré W. (1) , Knudsen G. M. (1) .<br />

(1) Copenhagen University, Denmark<br />

sgh@nru.dk<br />

Introduction: Post mortem studies suggest involvement<br />

of the serotonin system in Alzheimer’s disease<br />

(AD) and serotonin 2A (5-HT2A) receptors are<br />

globally reduced early in the course of the disease<br />

(1). However, few studies have investigated other<br />

aspects of the serotonergic system, including presynaptic<br />

markers (serotonin transporters, SERT) as<br />

a measure of serotonergic degeneration (2). Further,<br />

recent evidence points to a disease-modifying effect<br />

of 5-HT4 agonism through a decrease in beta-amyloid<br />

load (3). Therefore, we have investigated SERT<br />

and 5-HT4 in early and compared these measures to<br />

the reduction in 5-HT2A receptors.<br />

Methods: For the SERT/5-HT2A comparison, we<br />

included 12 patients (mean age 73.7 ±7.6 years, 8<br />

males) with AD (average MMSE of 24, range 19-26)<br />

and 11 healthy age-matched subjects (mean age 72.5<br />

±6.8 years, 6 males). Subjects were investigated with<br />

a 90 min dynamic [11C]DASB-PET recording to<br />

measure SERT (4) and a 40 min steady-state [18F]<br />

altanserin-PET recording to measure 5-HT2A receptors<br />

(5). In a separate study of 5-HT4 receptors,<br />

the novel radioligand [11C]SB207145 was used in<br />

twelve healthy individuals (mean age 67.2 y, 6 males)<br />

and eleven newly diagnosed AD patients (mean age<br />

70.6 y, 6 males, mean MMSE 24, range 19-27) using<br />

the simplified reference tissue model. Volumes of<br />

interest (VOIs) were delineated automatically on coregistered<br />

3T MRIs, and partial volume correction<br />

was applied to correct for differences in atrophy.<br />

Results: The 5-HT2A receptors were markedly reduced<br />

(25-66%) in AD patients in all regions but<br />

the striatum. Inc contrast, we found a reduction of<br />

SERT binding by 34% (p=0.0003) in the hippocampus,<br />

while most other regions were unaffected.<br />

Midbrain showed no change in binding (p=0.30).<br />

No statistically significant differences in 5-HT4 receptor<br />

binding between healthy individuals and AD patients<br />

were found in any of the included brain regions:<br />

Hippocampus (p = 0.55), posterior cingulate gyrus<br />

(p = 0.22), amygdala (p = 0.77), parietal cortex (p =<br />

0.44), temporal cortex (p = 0.74) and prefrontal cortex<br />

(p = 0.43).<br />

Conclusions: We showed a marked decrease of<br />

5-HT2A binding in patients with mild AD, consistent<br />

with previous findings in MCI (1). The SERT<br />

binding was unaffected by the disease in almost all<br />

cortical regions and in midbrain, suggesting that<br />

the serotonergic innervations and the neuron bodies<br />

in dorsal nucleus raphe are intact, at least early<br />

in the disease. We interpret the SERT reduction in<br />

hippocampus in patients as a decreased serotonergic<br />

innervation, which could be secondary to the neuronal<br />

degeneration taking place in hippocampus of<br />

AD patients. The marked reduction in 5-HT2A may<br />

be related to beta-amyloid accumulation. In contrast<br />

to the 5-HT2A receptor subtype, the 5-HT4 receptor<br />

levels seemed to be unaffected in AD. However,<br />

5-HT4 binding in relation to cognitive function,<br />

neuropsychiatric symptoms and beta-amyloid load<br />

should be further investigated.<br />

Acknowledgement: Supported by The Lundbeck<br />

Foundation, Rigshospitalet, and the Danish Medical<br />

Research Council. The John and Birthe Meyer Foundation<br />

is gratefully acknowledged for the donation<br />

of the Cyclotron and PET-scanner. These studies<br />

were funded in part by the EC - FP6-project DiMI,<br />

LSHB-CT-2005-512146.<br />

References:<br />

1. S. G. Hasselbalch et al., Neurobiol. Aging. 29, 1830<br />

(2008).<br />

2. K. Nielsen et al., Synapse. 59, 270 (2006).<br />

3. S.J. Robert, et al., Neurodegener Dis 5(3-4):163 (2008).<br />

4. M. Ichise et al., J Cereb. Blood Flow Metab. 23, 1096<br />

(2003). L. H. Pinborg et al., J. Cereb. Blood Flow Metab.<br />

23, 985 (2003).<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

day1<br />

Parallel Session 2: NEUROSCIENCE I


36 Introduction:<br />

WarSaW, poland May 26 – 29, 2010<br />

Qualitative and quantitative assessment of florbetapir F-18 PET ( 18 F-AV45) amyloid<br />

deposition PET imaging - validation by pathology. Preliminary report.<br />

Romanowicz G. (1) , Saha K. (2) , Krautkramer M. (2) , Joshi A. (2) , Pontecorvo M. (2) , Clark C. (2) , Carpenter A. (2) , Skovronsky D. (2) .<br />

(1) Gdański Uniwersytet Medyczny, Poland<br />

(2) Avid Radiopharmaceuticals Inc..<br />

greg@amg.gda.pl<br />

Various methods for analyzing amyloid<br />

brain PET scans have been proposed and tested.<br />

The lack of reliable truth standards has complicates<br />

the validation and implementation of these methods,<br />

and investigators have relied on theoretical modeling<br />

and clinical diagnoses as surrogates for true<br />

pathology. We applied three methods for assessing<br />

florbetapir F18 PET (18F-AV45) amyloid images: a<br />

semiquantitative visual read, a mean SUVr quantitation<br />

based on VOIs, and a novel automated histogram<br />

based quantitative approach. Each method<br />

was validated by comparison with amyloid burden<br />

measured at post-mortem.<br />

Methods: 6 subjects (3 men; 3 women; mean age at<br />

imaging 76y (47-86); diagnosis at enrollment, 1 MCI,<br />

4 AD, 1 PDD) with a life expectancy of < 6 months<br />

had a 10 minute PET scan 50 minutes after IV injection<br />

of 10 mCi of florbetapir F 18. Images were reconstructed<br />

by iterative technique with 4i/16s and 3<br />

mm FWHM post-reconstruction Gaussian filter. For<br />

visual assessment (VR) an experienced reader rated<br />

florbetapir cortical uptake intensity (VR score) on a<br />

5 point scale ranging from 0, (no difference in uptake<br />

between cortex and cerebellum), to 4 (uptake<br />

in all cortical brain regions significantly higher vs.<br />

both cerebellum and white matter). For the SUVr<br />

technique, PET images were spatially normalized by<br />

12 parameter affine registration to Talairach/MNI<br />

space and counts extracted from six regional VOIs<br />

(frontal, temporal, precuneus, parietal, anterior<br />

cingulated and posterior cingulated). Average standardized<br />

uptake value ratios (SUVr) were calculated<br />

with cerebellum as reference. The values for the six<br />

cortical regions were averaged to generate a global<br />

SUVr. For the histogram based analysis image data<br />

were processed automatically to generate a128 bin<br />

count histogram. The first order derivative Gaussian<br />

(FWHM= 3mm) convolved histogram curve was<br />

searched in the intensity direction to estimate separation<br />

of high/low intensity areas. The high/low intensity<br />

curve area ratio (CAR) was calculated to estimate<br />

specific amyloid binding. After the patient’s<br />

death (mean interval from imaging 43 days (range<br />

12-158)) their brain was evaluated for of β-amyloid<br />

deposition using immunohistochemistry (IHC) and<br />

by neuritic plaque rating (modified CERAD method).<br />

imaging life<br />

CAR score, VR score and SUVr were compared with<br />

IHC/CERAD using Spearman’s (ρ) or Pearson’s correlation<br />

coefficient (r) as appropriate.<br />

Results: A strong correlation was obtained between<br />

each measure of PET amyloid binding and both<br />

measures of amyloid deposition at autopsy (VR and<br />

IHC (ρ=0.88), VR and CERAD (ρ =0.92); SUVr and<br />

IHC (r=0.89), SUVr and CERAD (ρ=0.93); CAR and<br />

IHC (r=0.97), CAR and CERAD (ρ =0.64)).<br />

Conclusions: The results confirm that florbetapir F<br />

18 PET imaging provides relevant in vivo estimate of<br />

brain amyloid pathology irrespectively of the choice<br />

of image assessment method. The novel histogram<br />

based method is particularly interesting because it<br />

is quantitative and does not require fitting images<br />

to a template or reorientation of images, kinetic data<br />

acquisition, input functions or drawing / applying<br />

regions of interest.<br />

Acknowledgement: This work is supported by AVID<br />

Radiopharmaceuticals Inc. We would also like to<br />

thank A.S. Fleisher, J.A. Schneider, T.G. Beach, B.J.<br />

Bedell, S.P. Zehntner and M. Mintun for their contribution<br />

to the work.


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

In-vivo imaging of a mouse traumatic brain injury model using dual-isotope quantitative<br />

spect/ct<br />

Máthé D. (1) , Szigeti K. (2) , Fekete K. (3) , Horváth I. (2) , Benyó Z. (3) .<br />

(1) CROmed Ltd,<br />

(2) Nanobiotechnology and In Vivo Imaging Centre Semmelweis University Faculty of Medicine,<br />

(3) Semmelweis University Faculty of Medicine.<br />

domokos.mathe@gmail.com<br />

Introduction: Using a standardized mouse neurotrauma<br />

model we aimed at determining the role<br />

of high-resolution SPECT/CT in detecting and<br />

quantifying the volumetric and functional extent of<br />

blood-brain barrier disruption. We chose the freezing<br />

trauma model because of its reproducibly standard<br />

damage.<br />

Methods: Mice were subjected to a standardized<br />

freezing method transcranially using a cooled<br />

needle tip. In the presented initial studies, we compared<br />

the imaging pattern of the animals at equally 5<br />

hours post the freezing trauma. We used a dedicated,<br />

quantitative multiplexed multipinhole SPECT imaging<br />

system for laboratory animals. X-ray CT of the<br />

structures was also performed together with SPECT<br />

data acquisition.<br />

We correlated BBB disruption and blood perfusion<br />

with quantitative imaging of the decrease in glial<br />

and neuronal potassium uptake. We applied the validated<br />

intravenous tracer 99mTc-DTPA to detect the<br />

disruption of the BBB. Blood perfusion of the brain<br />

was assessed using the also validated tracer 99mTc-<br />

HMPAO. A radioactive isotopic potassium analogue,<br />

201Tl was used to track K+ uptake changes quantitatively.<br />

201Tl ions were passed through the BBB by<br />

means of diethyl-dithiocarbamate (DDC) complex<br />

that redistributes to ionic 201Tl after being taken<br />

up in brain tissues.<br />

Uptake volumes (in mm3) and total brain as well as<br />

lesion radioactivity values (in kBq) were evaluated<br />

on 3D reconstructed brain images of treated mice<br />

(n=5 per group) and a non-treated group. Besides<br />

imaging of single tracers (3 groups), simultaneous<br />

multi-channel imaging was used in two other groups<br />

of mice having received 99mTc-DTPA plus 201Tl-<br />

DDC or, 99mTc-HMPAO plus 201Tl-DDC.<br />

After imaging, histological control of the brain<br />

trauma size and localization was performed in the<br />

formaldehyde-fixed brains of all animals.<br />

Results: The uptake of 99mTc-DTPA correlated well<br />

with the size and localization of the lesion whereas<br />

the cold spot of the lesion at the perfusion image<br />

was evident in all animals having received 99mTc-<br />

HMPAO. However the cold spot was surrounded by<br />

an area of increased perfusion as compared to the<br />

same area in control animals. When comparing with<br />

201Tl-DDC images, the penumbra effect became evident,<br />

especially in the animals imaged with 99mTc-<br />

HMPAO and 201Tl-DDC in the same time. The<br />

area of non-active glial and neuronal potassium uptake<br />

was significantly larger than the non-perfused<br />

area and the hyper-perfused area co-localized with<br />

the edge of the disappeared neural potassium uptake<br />

volumes in 4 animals out of 5. SPECT/CT identified<br />

the dislocation of brain due to increased intracranial<br />

pressure in all treated animals.<br />

Conclusions: In vivo whole-animal imaging using<br />

quantitative SPECT is a very promising method to<br />

dissect different activation patterns and regulation<br />

of different mechanisms behind the events following<br />

neurotrauma (and consequently stroke too). Using<br />

a unique dual-isotopic approach to detect multiple<br />

events in the same time in the same animals, the size<br />

and presence of a penumbral region where perfusion<br />

is elevated but neural (both glial and neuronal) K+<br />

utilization is decreased, could be identified.<br />

Acknowledgement: We are grateful for Dr. Jürgen<br />

Goldschmidt (Magdeburg) and Roberto Pasqualini<br />

(Orsay) for valuable advice and kind hints in 201Tl-<br />

DDC application and preparation.<br />

References:<br />

1. Goldschmidt et al. NeuroImage 49 (2010) 303–315<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

YIA applicant<br />

day1<br />

Parallel Session 2: NEUROSCIENCE I


38 Introduction:<br />

WarSaW, poland May 26 – 29, 2010<br />

Molecular analysis of adaptive immune function through microscopic and mesoscopic<br />

imaging<br />

Stein J. .<br />

University of Bern, Switzerland<br />

jstein@tki.unibe.ch<br />

Cell migration is of central importance<br />

during development, cancer metastasis and the<br />

functioning of the immune system. A powerful<br />

approach to dissect molecular mechanisms during<br />

cell trafficking on a single cell level requires the<br />

adaptation of microscopy techniques to anesthetized<br />

animals, usually mice.<br />

Twophoton microscopy (2PM) was developed to<br />

examine the molecular mechanisms governing<br />

transmigration through endothelium and within<br />

tissue. Here, a brief overview is given over the<br />

techniques and potential applications of 2PM in the<br />

field of lymphocyte trafficking.<br />

imaging life<br />

The overall internal organization of organ<br />

microenvironments, such as B cell follicles and<br />

blood microvessels in lymph nodes, has thus far<br />

been mainly determined by two-dimensional tissue<br />

sectioning, whereby three-dimensional information<br />

is lost. Optical Projection Tomography (OPT) and<br />

selective plane illumination (SPIM) are recently<br />

developed mesoscopic imaging approaches to dissect<br />

the overall organization of specimen of 1-15 mm<br />

diameter. Their applications in lymphoid structure<br />

analysis will be discussed.


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

Magnetic resonance imaging – from structure to molecular interactions<br />

Rudin M. .<br />

ETH and University of Zürich, Switzerland<br />

rudin@biomed.ee.ethz.ch<br />

Magnetic resonance imaging (MRI) yields images<br />

characterized by high spatial resolution and<br />

unsurpassed soft-tissue contrast. The latter is<br />

the consequence of the fact that the MRI signals<br />

depends on multiple parameters such a various<br />

relaxation rates, water diffusivity, proton exchange<br />

reactions, which are tissue specific. By choice of<br />

experimental parameters contrast can be varied to<br />

highlight structures of interest. Paramagnetic or<br />

superparamagnetic contrast agents can be used to<br />

further enhance specific structures. It is therefore<br />

not surprising that today MRI has become one of the<br />

most important modalities for structural imaging.<br />

Yet MRI provides information beyond mere anatomy.<br />

The analysis of dynamic signal changes - e.g. during<br />

administration of a contrast agent - yields relevant<br />

quantitative information on physiological processes<br />

such as blood flow, vascular permeability, or renal<br />

function. Probably the most important physiological<br />

MRI application is functional MRI (fMRI), which<br />

has become an indispensable tool for studying<br />

brain function, or more precisely the hemodynamic<br />

response to neuronal activity, under normal and<br />

pathological conditions. fMRI measures local<br />

hemodynamic changes in the CNS that are linked to<br />

neural activity through neurovascular coupling.<br />

More recently, MRI has tapped into the field of<br />

molecular imaging. By coupling the MRI contrast<br />

agent to a targeting moiety, molecular information<br />

can be derived at high spatial resolution. Yet the<br />

MRI molecular imaging approach suffers from two<br />

limitations. MRI is insensitive due to the small<br />

quantum energy involved in the process, typically<br />

concentrations have to be in the micromolar range<br />

to induced chances detectable by MRI. This can be<br />

in part be accounted for by increasing the relaxivity<br />

of MRI reporters by increase of the payload or by<br />

using physiological amplification. The second<br />

limitation is that MRI contrast agents are in<br />

general bulky, which renders target accessibility<br />

difficult. Straightforward molecular targets that<br />

can be reached by MRI contrast agents are those<br />

expressed on the endovascular side. On the other<br />

hand the efficiency of MRI probes, similar to that<br />

of fluorescent probes, can be modulated through<br />

the molecular interaction with the target. This<br />

potentially allows discriminating target-bound from<br />

free floating probe, thereby enhancing the contrastto-background<br />

ratio. An attractive application of<br />

targeted MRI imaging are studies of cell trafficking.<br />

As many cells are rather tolerant to the amount of<br />

contrast agent they can carry, sensitivity appears<br />

to be less an issue. In fact there are some reports,<br />

that under favorable conditions single cells might be<br />

detected.<br />

The lecture will discuss the various aspects of MRI with<br />

focus on molecular and cellular imaging applications.<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

day1<br />

Parallel Session 3: TECHNOLOGY


40<br />

WarSaW, poland May 26 – 29, 2010<br />

Imaging modalities: PET and SPECT<br />

Ziegler S. .<br />

TU Munich, Department of Nuclear Medicine, Germany<br />

sibylle.ziegler@tum.de<br />

Nuclear medical imaging methods, positron emission<br />

tomography (PET) and single photon emission<br />

computed tomography (SPECT), utilize the detection<br />

of gamma rays leaving the body after a radioactive<br />

tracer has been administered in vivo. The choice<br />

of tracer substance as well as radio-nuclide for labelling<br />

depends on the biological process of interest<br />

and the organ which is imaged. While electronic<br />

collimation is used in PET, mechanical collimators<br />

made of high-Z materials need to be employed in<br />

SPECT imaging. Depending on the imaging situation,<br />

these collimators can be designed for parallel,<br />

imaging life<br />

diverging, or converging projection. Gamma ray detection<br />

in either modality is accomplished by scintillation<br />

detectors or semiconductor detectors, as in<br />

more recent developments. Raw data are projections<br />

of the activity distribution, which is reconstructed<br />

using analytical or statistical algorithms. Sensitivity<br />

of PET allows detecting pico-molar tracer amounts<br />

in vivo and current technology offers mm (PET) or<br />

sub-mm (SPECT) spatial resolution. Quantitative<br />

measurement of activity concentration in vivo is the<br />

basis for more advanced dynamic studies including<br />

biokinetic modelling.


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

in vivo fluorescence kinetic imaging for improved contrast and studies of temporal and<br />

quantitative biodistribution<br />

Mansfield J. (1) , Agarwal A. (2) , Curtis A. (2) , Krucker T. (2) .<br />

(1) Cambridge Research & Instrumentation,<br />

(2) Novartis Institutes for BioMedical Research.<br />

jmansfield@cri-inc.com<br />

Introduction: In vivo fluorescence imaging has<br />

added an easy and economical modality to the rapidly<br />

growing field of molecular imaging. It offers<br />

the possibility to perform experiments analogous<br />

to bioluminescence imaging through fluorescent<br />

proteins, but at longer and more efficient wavelengths.<br />

A further distinction is the translational<br />

and research aspects enabling the imaging of fluorophore-labelled<br />

biologics (e.g., antibodies, peptides,<br />

siRNA) and activatable reagents. One unique<br />

aspect limiting the sensitivity of in vivo fluorescence<br />

methodologies is the confounding effect of<br />

tissue autofluorescence, which can be addressed<br />

through the proper use of spectral imaging [1,2].<br />

However, like other molecular imaging modalities,<br />

reagent-based in vivo fluorescence imaging also<br />

has to contend with sensitivity and contrast problems<br />

due to non-specific signals and long wash-out<br />

times, both limiting the detection of specifically<br />

bound reagent and preventing accurate determination<br />

of uptake rates.<br />

Methods: A kinetic imaging modality, Dynamic<br />

Contrast Enhancement, or DyCETM, that combines<br />

rapid imaging (up to 15 frames/sec monochrome<br />

or 10 sec/frame multispectrally) with an advanced<br />

data processing methodology was developed. When<br />

combined with analysis software, these allow determination<br />

of (1) the rates of change of the intensity<br />

of the fluorophore in each pixel of the image and<br />

(2) the rate of uptake and wash-out in the animal.<br />

Mice were injected with a near-infrared fluorescent<br />

agent (IgG antibody) that was taken up in the liver<br />

and imaged for 80 minutes following injection at<br />

a rate of 1 multispectral dataset per minute. Each<br />

timepoint’s data were unmixed and kinetic data<br />

and “movies” assembled from these.<br />

Results: By utilizing the uptake and wash-out rate<br />

information, a much higher contrast image of the<br />

accumulating fluorophore was obtained in a much<br />

shorter period of time (minutes and hours vs. days).<br />

In addition, body compartment data determined<br />

from the kinetic data can provide information on<br />

the temporal distributions of a fluorescent agent<br />

during the experiment and can act as inputs for<br />

rate-of-change or body compartment models.<br />

Conclusions: This broadly applicable temporalbiodistribution<br />

methodology can be used, for<br />

example, to differentiate between the rate of liver<br />

uptake vs. the rate of tumor uptake and quantitate<br />

each signal relative to general body and/or bladder<br />

distribution. This information can then be<br />

combined with multispectral imaging and used to<br />

quantitate the biodistribution of multiple fluorophores<br />

simultaneously, each without interference<br />

from autofluorescence. When combined with<br />

models of peripheral, tumor, blood and wash-out<br />

(bladder) distributions, this kinetic imaging data<br />

can be transformed into a physiologically based<br />

pharmacokinetic model of agent distribution that<br />

provides an estimate of the pK values between the<br />

various compartments. Potentially such a method<br />

can also be used to establish optimal drug treatment<br />

schedules aiding drug discovery and development.<br />

References:<br />

1. Levenson, Mansfield, Cytometry A. 2006 Aug;<br />

69(8):748-58.<br />

2. Tam et al., Mol Imaging. 2007 Oct-Dec;6(4):269-76.<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

day1<br />

Parallel Session 3: TECHNOLOGY


42 Introduction:<br />

WarSaW, poland May 26 – 29, 2010<br />

Cerenkov radiation imaging of a xenograft murine model of mammary carcinoma<br />

Boschi F. (1) , Calderan L. (1) , D’ambrosio D. (2) , Marengo M. (2) , Fenzi A. (1) , Calandrino R. (3) , Sbarbati A. (1) , Spinelli A. E. (3) .<br />

(1) University of Verona,<br />

(2) S. Orsola – Malpighi University Hospital,<br />

(3) S. Raffaele Scientific Institute.<br />

federico@anatomy.univr.it<br />

2-[18F]fluoro-2-deoxy-D-glucose<br />

( 18 F-FDG) is a well known radiotracer for the in<br />

vivo studies of several diseases. In a previous paper<br />

[1] we showed that the positrons emitted by 18 F-FDG,<br />

travelling into tissues faster than the speed of light<br />

in the same medium, are responsible of Cerenkov<br />

Radiation (CR) emission which is prevalently in the<br />

visible range. The purpose of this work was to show<br />

that Cerenkov radiation escaping from tumour tissues<br />

of small living animals injected with 18 F-FDG<br />

can be detected with optical imaging (OI) techniques<br />

using a commercial optical instrument equipped<br />

with charged coupled detectors.<br />

Methods: In order to demonstrate as a proof of<br />

principle the possibility of imaging tumours using<br />

CR we studied an experimental model of mammary<br />

carcinoma named BB1. BB1 tumours were obtained<br />

by subcutaneous injection of BB1 cells, which are<br />

epithelial cells, from spontaneous mammary carcinomas<br />

of FVB transgenic mice for HER-2/neuT<br />

oncogene. The BB1 carcinomas exhibit histopatological<br />

and vascular features very similar to those<br />

of the parent spontaneous tumours. The BB1 mouse<br />

tumour model was well explained by Galiè and coworkers<br />

[2] and will not be described here.<br />

The images show a comparison between images acquired pre injection (left image) and 1 hour after 18F-<br />

FDG injection for two mice. White arrows indicate the position of the tumour masses.<br />

imaging life<br />

Mice injected with 18 F-FDG or saline solution underwent<br />

dynamic OI acquisition and a comparison<br />

between images were performed. Multispectral analysis<br />

of the radiation was used to estimate the deepness<br />

of the source of Cerenkov light. Small animal<br />

PET images were also acquired in order to compare<br />

the 18 F-FDG bio-distribution measured using OI<br />

and PET scanner.<br />

Results: The first Cerenkov in vivo whole body images<br />

of tumour bearing mouse and the measurements<br />

of the emission spectrum (560-660 nm range) were<br />

presented. Brain, kidneys and tumour were identified<br />

as a source of visible light in the animal body:<br />

the tissue time activity curves reflected the physiological<br />

accumulation of 18 F-FDG in these organs.<br />

The identification is confirmed by the comparison<br />

between CR and 18 F-FDG images.<br />

Conclusions: These results will allow the use of<br />

conventional optical imaging devices for the in<br />

vivo study of the cancer glucose metabolism and<br />

the assessment, for example, of anti-cancer drugs.<br />

Moreover this demonstrates that 18 F-FDG can be<br />

employed as it is as bimodal tracer for PET and OI<br />

techniques.<br />

References:<br />

1. Spinelli A E et al; Phys.<br />

Med. Biol. 55(2):483-<br />

495 (2010)<br />

2. Galiè M et al;<br />

C a r c i n o g e n e s i s .<br />

26:1868-1878 (2005)


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

A system for 4D (3D+Kinetics) molecular imaging in bioluminescence and fluorescence<br />

Maitrejean S. (1) , Kyrgyzov I. (1) , Bonzom S. (1) , Levrey O. (1) , Le Masne Q. (1) , Hernandez L. (1) , Raphael B. (2) .<br />

(1) Biospace Lab,<br />

(2) CEA/DSV/ I2BM / SHFJ / LIME.<br />

smaitrejean@biospacelab.com<br />

Introduction: Several technologies and systems are<br />

now capable of delivering tri-dimensional data in Bioluminescence<br />

and Fluorescence imaging. However, all<br />

are based on sequential acquisitions, and therefore any<br />

of them allow the production of real kinetic data in<br />

3D. In this work we demonstrate a new device which<br />

is able to acquire Bioluminescence and Fluorescence<br />

data from several views simultaneously, allowing the<br />

reconstruction of tri-dimensional images while keeping<br />

all kinetic properties of the acquisition.<br />

Methods: Multiple views of Bioluminescence and Fluorescence<br />

data are acquired simultaneously using a photon<br />

counting device (Photon ImagerTM) in which a<br />

specific add-on with four mirrors is installed. This device<br />

(named 4Views) allows the concurrent acquisition<br />

of the ventral, dorsal, right, and left views of the animal<br />

in a single image. The size and intensity of the four subimages<br />

(i.e. the four views) are corrected by taking the<br />

true optical path into account. The corrected photon<br />

counting data are recorded in a list mode file with time<br />

information (43 frame/s) as can be seen in Fig1.<br />

In a second step a micro video projector is used to<br />

project a moving spot on the animal. For each position<br />

of the spot, the<br />

height of the animal<br />

is calculated<br />

by triangulation<br />

and a map of the<br />

surface of the animal<br />

is produced.<br />

As the chosen<br />

reconstruction<br />

Fig1: simultaneous acquisition of four views<br />

in bioluminescence imaging.<br />

method is based<br />

on the finite elements<br />

method,<br />

the volume of the animal is represented by a tetrahedral<br />

mesh (about 30 000 tetrahedrons) using the Delaunay<br />

algorithm, while the surface is approximated<br />

by triangles, using a marching cube method. Then, the<br />

forward problem is solved for given light sources that<br />

are placed at the nodes of tetrahedrons and the light<br />

intensities on the surface of the triangles are computed.<br />

The forward problem is based in this first version on a<br />

diffusion model with average constant absorption and<br />

diffusion parameters. In a next version, a registration of<br />

the surface of the animal with an anatomical atlas will<br />

allow to use optical parameters associated with each<br />

organ. In the following step measured data is extracted<br />

from the list mode file and mapped on the triangulated<br />

surface. Since the detected events are stored separately,<br />

any time interval from 22 ms (one frame) up to the<br />

total scan duration can be used to create the data. In<br />

the last step of the process, the inner light sources are<br />

estimated as the result of the inverse problem using a<br />

least square criterion with a Tikhonoff regularization<br />

term. This regularisation term is chosen as an entropic<br />

term in order to favour connected solution. The two<br />

last steps can be performed using any time interval of<br />

the list mode file and therefore 3D kinetic data can be<br />

computed for times scales larger than 22 ms.<br />

Results: The validity of the method has been first tested<br />

using light beads (Microtek) inside a half cylinder of a<br />

scattering material (delrin). It has been demonstrated<br />

that two light beads as close as three millimetres could<br />

be separated, at a depth of one centimetre. A second<br />

set of tests has been realised on an animal (nude mice)<br />

by placing the light beads in the rectum of the animal.<br />

On the second image (Fig 2) , two beads 9 mm far from<br />

each other were placed in the rectum. It was possible to<br />

reconstruct the two positions of the light sources. The<br />

distance between the two reconstructed sources is 9.77<br />

mm in good agreement.<br />

Fig2: Reconstruction of two light sources based<br />

on the four views data of fig1.<br />

Conclusions: 3D optical reconstruction is known to<br />

be approximate, but can give useful and satisfactory<br />

results in a large number of applications. We have<br />

demonstrated that this four view approach is able to<br />

provide 3D kinetic molecular imaging data with a<br />

spatial accuracy similar to other methods.<br />

Acknowledgement: This work was supported in part<br />

by the DIMI and ENCITE networks.<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

day1<br />

Parallel Session 3: TECHNOLOGY


44 Magnetic<br />

WarSaW, poland May 26 – 29, 2010<br />

The alignment of target-specific lipoCEST MRI contrast agents<br />

Langereis S. (1) , Burdinski D. (1) , Pikkemaat J. (1) , Gruell H. (1) , Huskens J. (2) .<br />

(1) Philips Research Europe,<br />

(2) University of Twente.<br />

sander.langereis@philips.com<br />

resonance imaging (MRI) offers a unique<br />

combination of advantages such as the recording<br />

of contrast enhanced and anatomical images with<br />

a high spatial resolution, while avoiding the use of<br />

ionizing radiation. MRI for imaging sparse molecular<br />

epitopes on diseased cells is hampered by its low<br />

sensitivity, which can potentially be overcome with<br />

liposomal chemical exchange saturation transfer (lipoCEST)<br />

contrast agents (CAs).1-3 In this case, MR<br />

detection is based on the saturation of the intraliposomal<br />

water signal with a selective radiofrequency<br />

pulse. Water exchange across the liposomal membrane<br />

causes partial saturation of the bulk water<br />

signal and, as a consequence, negative contrast enhancement<br />

in the MR image. For in vivo applications,<br />

it is crucial to achieve large intraliposomal chemical<br />

shifts of the water protons, as larger shifts allow for<br />

better lipoCEST contrast enhancement, reduce the<br />

interference with background magnetization transfer<br />

effects, and allow for frequency-based multiplexing.<br />

Large chemical shift differences have been<br />

obtained upon aspherical deformation of liposomes<br />

encapsulating a chemical shift agent in response to<br />

osmotic shrinkage, and the additional incorporation<br />

of amphiphilic, paramagnetic lanthanide complexes<br />

within the liposomal bilayer.<br />

The direction of the chemical shift is governed by<br />

the alignment of the aspherical liposomes in the external<br />

magnetic field, which in turn is dictated by<br />

the sign of the magnetic anisotropy of the incorporated<br />

amphiphilic lanthanide complex. The use of<br />

lipoCEST CAs as targeted probes for molecular MRI<br />

applications entails their specific binding and immobilization<br />

at the target site, for example, the surface<br />

of a biological structure or a cell. To maximize the<br />

imaging life<br />

attractive interactions, such aspherical liposomes<br />

will tend to align with the target structure (Scheme<br />

1).4 This enforced orientation may, however, be<br />

different from the magnetic alignment. As a consequence,<br />

the chemical shift of the intraliposomal<br />

water of the bound lipoCEST CA may differ from<br />

that of the unbound CA. It is therefore essential<br />

to understand the interplay between the preferred<br />

magnetic and the enforced mechanical alignment of<br />

such aspherical lipoCEST CAs. Herein, we address<br />

the alignment change of such aspherical liposomes<br />

upon multivalent binding to a target surface, which<br />

was studied by using routine CEST MR methods.4<br />

References:<br />

Fig1.: Reorientation of aspherical<br />

lipoCEST contrast agents upon<br />

binding to a target surface with<br />

respect to an external magnetic<br />

field (B0).4<br />

1. Aime, S.; Delli Castelli, D.; Terreno, E. Angew. Chem. Int.<br />

Ed. 2005, 44, 5513-5515.<br />

2. Terreno, E.; Cabella, C.; Carrera, C.; Delli Castelli, D.;<br />

Mazzon, R.; Rollet, S.; Stancanello, J.; Visigalli, M.; Aime,<br />

S. Angew. Chem. Int. Ed. 2007, 46, 966-968.<br />

3. Langereis, S.; Keupp, J.; van Velthoven, J. L. J.; de Roos,<br />

I. H. C.; Burdinski, D.; Pikkemaat, J. A.; Grüll, H. J. Am.<br />

Chem. Soc. 2009, 131, 1380-1381.<br />

4. Burdinski, D.; Pikkemaat, J. A.; Emrullahoglu, M.;<br />

Costantini, F.; Verboom, W.; Langereis, S.; Grüll, H.;<br />

Huskens, J. Angew. Chem. Int. Ed. 2010, 49, 2227-2229.


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

Lanthanide-based in vivo luminescence imaging<br />

Petoud S. .<br />

CNRS, France<br />

spetoud@pitt.edu<br />

Introduction: Fluorescence and luminescence<br />

are detection techniques that possess important<br />

advantages for bioanalytical applications and<br />

biologic imaging: high sensitivity, versatility and<br />

low costs of instrumentation. A common characteristic<br />

of biologic analytes is their presence in<br />

small quantities among complex matrices such as<br />

blood, cells, tissue and organs. These matrices<br />

emit significant background fluorescence (autofluorescence),<br />

limiting detection sensitivity.<br />

The luminescence of lanthanide cations has several<br />

complementary advantages over the fluorescence<br />

of organic fluorophores and semiconductor<br />

nanocrystals, such as sharp emission bands for<br />

spectral discrimination from background emission,<br />

long luminescence lifetimes for temporal<br />

discrimination and strong resistance to photobleaching.<br />

In addition, several lanthanides emit<br />

near-infrared (NIR) photons that can cross deeply<br />

into tissues for non-invasive investigations and<br />

that result in improved detection sensitivity due<br />

to the absence of native NIR luminescence from<br />

tissues and cells. The main requirement to obtain<br />

lanthanide emission is to sensitize them with an<br />

appropriate chromophore.<br />

Methods: An innovative concept for such sensitization<br />

of lanthanide cations is proposed herein;<br />

the current limitation of low quantum yields<br />

experienced by most mononuclear lanthanide<br />

complexes is compensated for by using larger<br />

numbers of lanthanide cations and by maximizing<br />

the absorption of each discrete molecule,<br />

thereby increasing the number of emitted photons<br />

per unit of volume and the overall sensitivity<br />

of the measurement. To apply this concept, we<br />

are developing a family of dendrimer-naphthalimide<br />

ligands that are able to incorporate several<br />

lanthanide cations. Polyamidoamine (PAMAM)<br />

dendrimers have been chosen as a basis for these<br />

complexes because the oxygen atoms of the amido<br />

groups located along their branches can bind<br />

and protect the lanthanide cations inside the<br />

dendrimer core.1,2 Derivatives of naphthalimide<br />

groups, required for the sensitization of the lanthanide<br />

cations, are located at the branch termini.<br />

Our synthetic approach allows facile modification<br />

of the dendrimer complex for control over<br />

photophysical properties and solubility. It also<br />

provides for the attachment of different types<br />

of targeting agents such as peptides, oligonucleotides<br />

or proteins, as well as other sensing agents,<br />

to provide functionality to these compounds in a<br />

broad range of applications.<br />

Results: In this paper, we will describe several<br />

examples of luminescent polymetallic lanthanide<br />

complexes based on dendrimers. We will also<br />

present examples of their applications as reporters<br />

and sensors for biologic imaging in living cells<br />

and small animals.<br />

References:<br />

1. J. P. Cross, M. Lauz, P. D. Badger, S. Petoud, Journal of<br />

the American Chemical Society, 2004, 126, 16278.<br />

2. D. R. Kauffman, C. M. Shade, H. Uh, A. Star and S. Petoud,<br />

Nature Chemistry 2009, 1, 500.<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

day1<br />

Parallel Session 4: PROBES - supported by COST


46<br />

WarSaW, poland May 26 – 29, 2010<br />

New perspectives for imaging molecules and metabolism in vivo<br />

Gruetter R. .<br />

University of Lausanne and Geneva, Switzerland<br />

rolf.gruetter@epfl.ch<br />

Magnetic resonance imaging is subjected to continuous<br />

advances and discoveries, much of which are lead<br />

by either improved data acquisition techniques and/<br />

or higher magnetic fields or the detection of novel<br />

principles/contrast mechanisms altogether. This presentation<br />

will highlight on one hand the possibility<br />

to detect contrast agents at very low concentrations<br />

using a new approach to MR, based on hyperpolarized<br />

media using dissolution DNP. The potential<br />

will be demonstrated especially for long-lived nuclei<br />

with long T1, such as Lithium-6, Yttrium-89 and<br />

N-15, but also C-13 will be illustrated. The ability to<br />

detect low-concentration nuclei or rare spin nuclei<br />

imaging life<br />

opens new possibilities for molecular imaging. On<br />

the other side, for imaging molecules (metabolism)<br />

recent advances have allowed to attain in rodent<br />

brain a spatial resolution of below 1umol at 14.1<br />

Tesla that matches or even exceeds that possible by<br />

nuclear imaging modalities, in particular PET. The<br />

quantitative nature of the MR spectroscopic imaging<br />

approach allows insights into cellular biochemistry<br />

and metabolism that complements the capabilities<br />

of PET and SPECT and opens new windows on<br />

characterizing disease evolution and treatment opportunities,<br />

as will be illustrated with the example of<br />

ischemia and cancer, among others.


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

In vivo biodistribution of radiolabeled matrix metalloproteinase-2 activatable cell<br />

penetrating peptides<br />

Van Duijnhoven S. (1) , Robillard M. (2) , Nicolaij K. (1) , Gruell H. (1) .<br />

(1) University of Technology Eindhoven, The Netherlands<br />

(2) Philips Research Laboratories. The Netherlands<br />

s.m.j.v.duijnhoven@tue.nl<br />

Introduction: Activatable cell penetrating peptides<br />

(ACPPs) are a new class of promising molecular<br />

imaging probes for the visualization of proteolytic<br />

activity in vivo [1-3] . The cell penetrating function of<br />

a polycationic peptide is efficiently blocked by intramolecular<br />

electrostatic interactions with a polyanionic<br />

peptide. Proteolysis of a cleavable linker<br />

present between the polycationic cell penetrating<br />

peptide and the polyanionic peptide affords dissociation<br />

of both domains and enables the activated<br />

cell penetrating peptide to enter cells. Fluorescently<br />

labeled ACPPs cleavable by matrix metalloproteinase-2<br />

(MMP-2) have been reported as specific<br />

probes for MMP-2 expressing tumors in mice [1-3] .<br />

Here, we developed MMP-2 activatable dual isotope<br />

radiolabeled ACPPs and assessed their in vivo<br />

biodistribution in HT-1080 tumor-bearing mice.<br />

These probes enabled us to discriminate activated<br />

from intact ACPP.<br />

Methods: MMP-2 activatable and non-activatable<br />

cell penetrating peptides (ACPP and non-ACPP, respectively)<br />

containing an n-terminal tyrosine were<br />

prepared by Fmoc solid-phase peptide synthesis,<br />

site specifically conjugated with DOTA chelate succinimidyl<br />

ester at the c-terminus, purified by HPLC,<br />

and characterized by LC-MS. Furthermore, a DOTAconjugated<br />

cell penetrating peptide (CPP) was synthesized<br />

as positive control. Nude mice were injected<br />

subcutaneously with approximately 3.0 x 10 6 MMP-2<br />

positive HT-1080 fibrosarcoma cells. When the<br />

tumors reached a size of 8-50 mm 3 , the mice (n=6<br />

per probe) were used for in vivo studies. Therefore,<br />

the cell penetrating peptide and polyanionic peptide<br />

of ACPP or non-ACPP (60 nmol) were labeled<br />

with 177 Lu and 125 I, respectively, analyzed by iTLC<br />

and HPLC, injected into the tail vein, and in vivo<br />

biodistribution was determined 24h post-injection.<br />

Results: Radiolabeled ACPP (>98% radiochemical<br />

purity for both 177 Lu and 125 I) showed a significant<br />

~3-fold increase in tumor uptake relative to a negative<br />

control peptide with a scrambled linker (t-test,<br />

p


YIA applicant<br />

48 Introduction:<br />

WarSaW, poland May 26 – 29, 2010<br />

Bioresponsive MRI contrast agents based on self-assembling β-cyclodextrin nanocapsules<br />

Martinelli J. (1) , Fekete M. (1) , Tei L. (1) , Botta M. (1) .<br />

Università del Piemonte Orientale “A. Avogadro”, Italy<br />

jonathan.martinelli@unipmn.it<br />

One of the most exciting research area<br />

in the development of MRI contrast agents is the design<br />

of responsive or “smart” probes, whose performance<br />

is modulated by changes in physiological environment<br />

such as pH, partial oxygen pressure, metal<br />

ion concentration, enzyme activity etc. We have designed<br />

and synthesized a new type of nanosized Gdbased<br />

MRI probe containing disulfide bonds, that<br />

can be activated (i.e. switched on or off) under reducing<br />

conditions like those where specific enzymes<br />

or high radical concentrations are associated with a<br />

disease state (e.g. tumors, strokes etc.). The activation<br />

of the agent is signaled by a large change in the<br />

relaxation properties.<br />

Methods: The macromolecular architecture of the<br />

agent is based on nanocapsules prepared from perthiolated<br />

β-cyclodextrins via oxidation of the thiol<br />

groups to form S-S bridges.1 The assembly was<br />

carried out in the presence of either an equimolar<br />

amount or an excess of Gd-benzyl-AAZTA2 as the<br />

MRI-active paramagnetic complex, able to form<br />

inclusion adducts with the cyclodextrin buildingblocks<br />

and thus being “trapped” inside the capsules<br />

during their formation. After purification of the<br />

nanomaterials and removal of the external Gd-complexes<br />

by prolonged dialysis, the macromolecular<br />

systems were characterized by analytical and NMR<br />

relaxometric techniques. Reduction kinetic experiments<br />

were carried on by monitoring the variation<br />

in the longitudinal relaxation rate vs. time upon addition<br />

of tris(2-carboxyethyl)phosphine (TCEP)3 as<br />

a reducing agent able to cleave the disulfide bonds<br />

between the CD units.<br />

Results: High relaxivity nanoparticles made up of several<br />

β-CD units were formed which include in the inner<br />

core a relatively large number of Gd-chelates. The<br />

ICP analyses and NMRD profiles confirmed that a<br />

significant amount of complex was caged within the<br />

macromolecular structures. The relaxivity is as high<br />

as 22 mM-1 s-1 at 20 MHz and 25 °C for the system<br />

prepared with a 5:1 complex/CD ratio. Dynamic light<br />

scattering measurements also showed that the size of<br />

the nanocapsules prepared in the presence of the Gdcomplex<br />

is sensibly bigger than in its absence (120-<br />

200 nm vs. 30 nm). Following addition of TCEP and<br />

imaging life<br />

cleavage of the S-S bridges, a significant decrease (ca.<br />

30%) in the water proton relaxation rate was observed.<br />

Conclusions: The high relaxivity values measured for<br />

the Gd-loaded nanocapsules imply a high permeability<br />

of water through the cyclodextrin units of the surface<br />

shell. Upon disruption of the nanoparticles following<br />

the cleavage of the disulfide bonds between<br />

the CD units by the reducing agent TCEP, the complexes<br />

are released and originate an equilibrium between<br />

the free form and their adduct with the monomeric<br />

CDs. This is signaled by a remarkable decrease<br />

in the relaxivity as a consequence of the shortening<br />

of the reorientational correlation time τR.<br />

According to these data, our β-cyclodextrin nanocapsules<br />

containing Gd-complexes appear to be<br />

promising as MRI contrast agents responsive to<br />

reducing biological environments.<br />

Acknowledgement: This work is supported by the<br />

Regione Piemonte (Italy) as part of the NanoIGT<br />

project.<br />

References:<br />

1. Jones, L. C.; Lackowski, W. M.; Vasilyeva, Y.; Wilson, K.;<br />

Chechik, V. Chem. Commun. 2009, 1377-1379.<br />

2. Aime, S.; Calabi, L.; Cavallotti, C.; Gianolio, E.; Giovenzana,<br />

G. B.; Losi, P.; Maiocchi, A.; Palmisano, G.; Sisti, M. Inorg.<br />

Chem. 2004, 43, 7588-7590.<br />

3. Burns, J. A.; Butler, J. C.; Moran, J.; Whitesides, G. M. J.<br />

Org. Chem. 1991, 56, 2648-2650.


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

Photochemical activation of endosomal escape of MRI-Gd-agents in tumor cells<br />

Gianolio E. (1) , Arena F. (1) , Hogset A. (2) , Aime S. (3) .<br />

(1) Centro di Imaging Molecolare,<br />

(2) PCI Biotech AS Laboratories,<br />

(3) Molecular Imaging Center, Torino Italy.<br />

eliana.gianolio@unito.it<br />

Introduction: The cellular uptake of xenobiotics often<br />

procedes through the entrapment into endosomes. As<br />

recently reported1,2, in the case of cellular labeling with<br />

Gd-based complexes, the confinment into endosomal<br />

vesicles negatively affects the attainable relaxation enhancement<br />

of water protons. In fact it has been shown<br />

that, upon increasing the number of Gd(III) per cell, a<br />

quenching effect on the observed relaxivity takes place.<br />

It is the consequence of the fact that the term |R1end<br />

– R1cyt| is > than the water exhange rate between the vesicular<br />

and cytosolic compartments. In this communication<br />

we report an efficient method for endosomal escape<br />

of paramagnetic Gd(III) chelates that yields to a marked<br />

improvement of the efficiency in cellular labeling.<br />

Methods: The Photosensitizer TPPS2a (LumiTrans®)<br />

and the Lumisource® lamp were provided by PCI Biotech<br />

AS laboratories (Oslo, Norway). For cellular labeling,<br />

ca. 3-4×106 HTC,K562, NEURO2A and C6 cells<br />

were incubated for 18 hours at 37°C with different concentrations<br />

(5-100mM) of Gd-HPDO3A in the absence<br />

and in the presence of the Photosentisizer (2µl/ml). After<br />

this incubation time cells were washed three times<br />

and incubated for additional 4 hours at 37°C. Then the<br />

cells containing the Photosentisizer were exposed to LumiSource<br />

light for 5 minutes. Then cells were detached,<br />

washed and transferred into glass capillaries for registraion<br />

of MR-images on a Bruker Avance300 spectrometer<br />

operating at 7.1T.<br />

K562 cells<br />

B<br />

K562 cells<br />

A<br />

C<br />

D<br />

E<br />

F<br />

G<br />

R1obs (s -1 )<br />

10<br />

9<br />

8<br />

7<br />

6<br />

5<br />

4<br />

3<br />

2<br />

1<br />

0,0 5,0x10 9<br />

1,0x10 10<br />

Number of Gd 3+ / cell<br />

1,5x10 10<br />

Fig.1: A) T 1 -weighted spin echo images of K562 cells labeled with Gd-HPDO3A. Phantoms E, F<br />

and G contain cells treated with the PCI technology while phantoms B, C, and D contain cells<br />

simply labeled by pynocitotic uptake. B) Observed relaxation rates of cells (HTC stars; NEURO-2a<br />

circles; C6 triangles) labelled with Gd-HPDO3A with endosomic (black symbols) distribution as<br />

a function of the number of Gd(III) found in each cell.<br />

Results: The cellular labeling has been pursued<br />

by pynocitosis (18h at 37°C) at different concentration<br />

of Gd-HPDO3A in the presence and in<br />

the absence of PCI treatment. As shown in the<br />

figure 1A, a marked enhancement in the labeling<br />

efficiency has been observed upon application<br />

of photochemical stimulus to cells entrapping<br />

Gd-HPDO3A and TPPS2a. This considerable<br />

gain in signal intensity achieved when cells are<br />

processed with PCI tehnology is due to the release<br />

of Gd-units from endosomes to cytosol. As<br />

shown in Fig. 1B, the “quenching” effect on the<br />

relaxivity of cells processed with PCI technology<br />

is reached when the number of Gd-HPDO3A per<br />

cell is ca. One order of magnitude higher than the<br />

number causing the same effect when the paramagnetic<br />

complexes are confined into endosomes.<br />

Thus, on going from the endosome-entrapped to<br />

cytoplasm-entrapped Gd(III), the paramagnetic<br />

loading can be several times higher thus allowing<br />

a marked improvement in the MRI detection of<br />

labelled cells.<br />

Conclusions: The PCI methodology appears an<br />

excellent route to pursue the endosomal escape of<br />

Gd-HPDO3A molecules entrapped by pynocitosis<br />

in different types of cells. It as been shown that<br />

it allows to exploit high payload of Gd-HPDO3A<br />

before the quenching effect becomes detectable.<br />

Fig. 1: A) T1-weighted spin echo<br />

Neuro<br />

images of K562 cells Neuro_lumi labeled with<br />

Gd-HPDO3A. Phantoms Htc E, F and G<br />

contain cells treated Htc_lumi with the PCI<br />

C6<br />

technology while phantoms C6_Lumi B,C and<br />

D contain cells simply labeled by<br />

pynocitotic uptake. B) Observed<br />

relaxation rates of cells (HTC stars;<br />

NEURO-2a circles; C6 triangles)<br />

labelled with Gd-HPDO3A with<br />

endosomic (black symbols) or<br />

cytoplasmatic (red symbols)<br />

distribution as a function of the<br />

number of Gd(III) found in each cell.<br />

References:<br />

1. Terreno, E et al., Magnetic<br />

Resonance in Medicine,<br />

2006, 55, 491-497.<br />

2. Strijkers G.J. et al. 2009,<br />

61, 1049-58.<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

day1<br />

Parallel Session 4: PROBES - supported by COST


50 Education<br />

WarSaW, poland May 26 – 29, 2010<br />

and Research Experience:<br />

Degree in Medicine and Surgery at the Catholic University of Rome (Italy).<br />

Membership of the College of Physicians and Surgeons of Rome (Italy).<br />

Specialization in Neurology at the Catholic University of Rome (UCSC).<br />

Post-graduate research at the laboratory of muscular disease of the neurological department at the Catholic<br />

University of Rome (UCSC).<br />

PhD in Neuroscience at the Neuroscience Department of the Catholic University of Rome (UCSC) within a<br />

joint collaboration with the department of Neuroimmunology of the Max Planck Institute for Neurobiology<br />

(Martinsried, Germany).<br />

Post doctoral position at the Max Planck Institute of Neurobiology, department of Neuroimmunology<br />

(Martinsried, Germany).<br />

Group leader position at the Institute for Multiple Slerosis Research (IMSF) of the Georg-August Universität<br />

of Göttingen.<br />

Dissertation: Visualization of antigen presentation and T cell activation after treatment with high doses of<br />

soluble antigen.<br />

Selected references:<br />

• I. Bartholomäus, N. Kawakami, F. Odoardi, C. Schläger, D. Miljkovic, J.W. Ellwart., W.E.F Klinkert., C. Flügel-Koch, T.B.<br />

Issekutz, H. Wekerle, A. Flügel. Effector T cell interactions with meningeal vascular structures in nascent autoimmune<br />

lesions, Nature. 2009 Nov 5; 462 (7269):94-8.<br />

• W. Dammermann+, B. Zhang+, M. Nebel+, C. Cordiglieri+, F. Odoardi, T. Kirchberger, N. Kawakami, J. Dowden, F.<br />

Schmid, K. Dornmair, M. Hohenegger, A. Flügel*, A.H. Guse*, B.V.L. Potter* (2009) NAADP mediated Ca2+ signaling via<br />

type 1 ryanodine receptor in T cells revealed by a synthetic NAADP antagonist, PNAS, 2009 Jun 30; 106 (26): 10678-83.<br />

+,* equal contribution.<br />

• Odoardi F, Kawakami N, Klinkert WE, Wekerle H, Flügel A. Blood-borne soluble protein antigen intensifies T cell activation<br />

in autoimmune CNS lesions and exacerbates clinical disease. Proc Natl Acad Sci U S A. 2007 Nov 20;104(47):18625-30.<br />

• Flügel A, Odoardi F, Nosov M, Kawakami N. Autoaggressive effector T cells in the course of experimental autoimmune<br />

encephalomyelitis visualized in the light of two-photon microscopy. J Neuroimmunol. 2007 Nov;191(1-2):86-97.<br />

• Odoardi F, Kawakami N, Li Z, Cordiglieri C, Streyl K, Nosov M, Klinkert WE, Ellwart JW, Bauer J, Lassmann H, Wekerle H,<br />

Flügel A. Instant effect of soluble antigen on effector T cells in peripheral immune organs during immunotherapy of<br />

autoimmune encephalomyelitis. Proc Natl Acad Sci U S A. 2007 Jan 16;104(3):920-5.<br />

• Kawakami N, Nägerl UV, Odoardi F, Bonhoeffer T, Wekerle H, Flügel A. Live imaging of effector cell trafficking<br />

and autoantigen recognition within the unfolding autoimmune encephalomyelitis lesion. J Exp Med. 2005 Jun<br />

6;201(11):1805-14.<br />

imaging life<br />

Francesca Odoardi


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

Immune surveillance and autoimmunity: how encephalitogenic T cells enther their target organ<br />

Odoardi F. .<br />

Max Planck Institute of Neurobiology, Germany<br />

odoardi@neuro.mpg.de<br />

Introduced in the immunological field in 2002<br />

two-photon microscopy is the method of choice<br />

for visualizing living cells in their anatomical<br />

compartment because it allows to image deep<br />

in the tissue with negligible phototoxicity and<br />

photobleaching. We applied this technique in order<br />

to visualize in living Lewis rat the fate of genetically<br />

labeled MBP specific effector T cells in the menigeal<br />

vessels during the initial phase of experimental<br />

autoimmune encephalomyelitis, a classical model of<br />

multiple sclerosis. We observed that the incoming<br />

cells remained in close association with pial blood<br />

vessels, crawling on surfaces within the outline of<br />

the vessels. This behavior was specific to the CNS:<br />

in peripheral organs, for example in peripheral<br />

nerves, muscle or subcutaneous tissue, MBP T<br />

cells mainly rolled along the inner surface of the<br />

vessels. The crawling was completely abolished by<br />

the treatment with anti VLA-4 and LFA-1 antibodies.<br />

After diapedesis, the cells continued their scan on<br />

the abluminal vascular surface and the underlying<br />

leptomeningeal (pial) membrane. Here they<br />

established contact with local meningeal phagocytes<br />

and these interactions were crucial to induce the<br />

production of proinflammatory and to trigger<br />

tissue invasion.<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

day1<br />

<strong>ESMI</strong> Plenary Lecture 2 by Francesca Odoardi


52 Introduction:<br />

WarSaW, poland May 26 – 29, 2010<br />

Exendin-4 derivatives labeled with radiometals for the detection of insulinoma: a “from<br />

bench to bed approach”<br />

Wild D. (1) , Wicki A. (1) , Mansi R. (1) , Béhé M. (1) , Keil B. (1) , Bernhardt P. (1) , Christofori G. (1) , Ell P. J. (1) , Reubi J. C. (1) , Maecke H. (1) .<br />

(1) University of Freiburg, Department of Nuclear Medicine<br />

damian.wild@uniklinik-freiburg.de<br />

Strong overexpression of glucagonlike<br />

peptide-1 (GLP-1) receptors in human insulinoma<br />

provides an attractive target for imaging.<br />

We have shown that GLP-1 receptor SPECT/CT<br />

using a Lys(Ahx-DOTA)NH2 C-terminal extended<br />

Exendin-4 derivative labeled with 111In shows<br />

high sensitivity in the detection of hardly detectable<br />

insulinoma [1]. For obvious reasons 111In<br />

is not an ideal nuclear imaging probe label. We<br />

therefore aimed at the development of 99mTc-<br />

(SPECT/CT) and 68Ga-, 64Cu-(PET/CT) labeled<br />

peptides. In addition we extended our clinical<br />

studies with a DTPA-modified peptide for higher<br />

specific activity labeling with 111In.<br />

Methods: Internalisation, biodistribution and<br />

imaging studies were performed in the Rip1Tag2<br />

mouse model and the corresponding cell line.<br />

The Results were compared with the “gold standard”<br />

Lys40(Ahx-DOTA-111In)NH2-Exendin-4.<br />

Kidney blocking was studied with poly-glutamic<br />

acid and Gelofusine. Clinical data were obtained<br />

with the 111In-labeled DTPA-analog.<br />

Results: The tumor uptake of Lys40(Ahx-DOTA-<br />

68Ga)NH2-Exendin-4 was very high, at 205±59<br />

%IA/g (1h pi) and was very similar to Lys40(Ahx-<br />

DOTA-111In) NH2-Exendin-4. > 90% of this<br />

uptake could be blocked by the preinjection of<br />

cold peptide. Normal organs showed much lower<br />

uptake resulting in a high lesion-to-background<br />

ratio. Kidney uptake was high and comparable<br />

to the tumor uptake. Lys40(Ahx-HYNIC-99mTc/<br />

EDDA)NH2-Exendin-4 shows distinctly lower<br />

tumor but also normal tissue uptake. Tumors<br />

with 1-3.2 mm in size could be visualised easily<br />

by SPECT and PET.<br />

The kidney uptake could be reduced by 49-78%<br />

using poly-glutamic acid, Gelofusine or a combination<br />

of both.<br />

Clinical studies using SPECT/CT with Lys40(Ahx-<br />

DTPA-111In)NH2-Exendin-4showed 100% sensitivity<br />

in the localisation of benign insulinoma<br />

of the first consecutive 13 patients. Some of the<br />

localisations such as an ectopic insulinoma could<br />

imaging life<br />

only be localised with SPECT/CT. Due to the long<br />

residence time in the tumor intraoperative localisation<br />

with an endoscopic gamma probe was possible<br />

even 2 weeks i.p..<br />

Conclusions: These very promising pharmacokinetic<br />

data show that the two new imaging probes<br />

are good candidates for clinical translation: Especially<br />

the 99mTc-labeled peptide may increase<br />

the availability and distribution of the method<br />

whereas the 68Ga derivative may even allow to<br />

image and localise very small lesions<br />

Acknowledgement: Oncosuisse grant No OSC-<br />

01778-08-2005 and grants from the Novartis<br />

Foundation and Swiss National Science Foundation<br />

are gratefully achnowledged.<br />

References:<br />

1. Wild D, Macke H, Christ E, Gloor B, Reubi JC. Glucagonlike<br />

peptide 1-receptor scans to localize occult<br />

insulinomas. N Engl J Med. 2008;359(7):766-768.


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

Assessment of HIF transcriptional activity in a mouse tumor model using GPI anchored avidin–<br />

a novel protein reporter for in vivo imaging<br />

Lehmann S. (1) , Garcia E. (2) , Blanc A. (2) , Schibli R. (2) , Keist R. (1) , Rudin M. (1) .<br />

(1) Animal imaging center,<br />

(2) Paul Scherrer Institute, Villigen, Switzerland.<br />

lehmann@biomed.ee.ethz.ch<br />

Introduction: With the emergence of multimodal<br />

imaging approaches genetic reporters, which can be<br />

flexibly combined with multiple imaging methods,<br />

are highly attractive. Here we present the feasibility<br />

of using glycosylphosphatidylinositol anchored avidin<br />

(av-GPI) (1) as a novel reporter for multimodal<br />

in vivo imaging. Expressed on the extracelluar side<br />

of cell membranes, av-GPI can be targeted with<br />

biotinylated imaging probes. In this study, we employed<br />

av-GPI to read out on the activity of hypoxia<br />

inducible factors (HIFs) in tumors. Induced by tumor<br />

hypoxia to mediate the adaptation of cells to<br />

low oxygen tensions these transcription factors play<br />

an important role in cancer progression. Typically,<br />

the expression of HIF in human cancer patients is<br />

associated with more aggressive tumor phenotypes<br />

and poor patient prognosis. Imaging HIF activity<br />

in live tumors hence provides an important tool to<br />

study the mechanisms leading to its activation in<br />

cancer.<br />

Methods: Mouse C51 cells were stably transfected<br />

with pH3SVG, a reporter construct driving the<br />

expression of avidin-GPI from a minimal SV40<br />

promoter and 3 hypoxia response element (HRE),<br />

bound by HIF, from the human transferrin gene<br />

(2). To monitor HIF activity in vivo, pH3SVG transfected<br />

C51 cells were subcutanouesly implanted into<br />

Balb/C nude mice. 10 days after tumor inoculation,<br />

mice received an i.v. injection of alexa-594-biocytin<br />

and were imaged using fluorescence reflectance<br />

imaging.<br />

Results: Fluorescence stainings of av-GPI expressing<br />

cells demonstrated that this protein is specifically expressed<br />

on the extracellular side of cell membranes.<br />

Moreover, upon pharmacological activation of HIF,<br />

we observed a shift in fluorescence indicative of<br />

an increased expression of av-GPI in fluorescent<br />

activated cell sorting (FACS) experiments involving<br />

pH3SVG transfected cells. In vivo fluorescence<br />

imaging showed a specific uptake of a biotinylated<br />

dye (alexa-594-biocytin) in the tumor from 60 minutes<br />

after contrast injection, whilst there was no<br />

accumulation of an unbiotinylated control probe<br />

(alexa-594-cadaverine). On ex vivo tissue sections<br />

alexa-594 biocytin was found to co-localize with<br />

zones positive for pimonidazole, a commonly used<br />

hypoxia marker (3) but also showed staining in regions<br />

devoid of pimonidazole uptake. In additional<br />

experiments, biotinylated 67Ga-DOTA was shown<br />

to specifically label avidin expressing cells in vitro.<br />

Conclusions: Overall, we demonstrate the utility<br />

of av-GPI as a reporter for in vivo imaging of HIF<br />

transcriptional activity in an optical, fluorescence<br />

reflectance approach. In vitro binding studies with<br />

67Ga-DOTA showed a high specificity of the probe<br />

in targeting av-GPI in cells, which implies that<br />

this reporter can indeed be combined with different<br />

imaging modalities. Its application in SPECT is<br />

currently being tested.<br />

References:<br />

1. Pinaud, F., King, D., Moore, H.-P. & Weiss, S. (2004)<br />

Bioactivation and Cell Targeting of Semiconductor<br />

CdSe/ZnS Nanocrystals with Phytochelatin-Related<br />

Peptides. Journal of the American Chemical Society<br />

126: 6115-6123<br />

2. Wanner, R. M., et al. (2000) Epolones induce<br />

erythropoietin expression via hypoxia-inducible<br />

factor-1 alpha activation. Blood 96: 1558-65<br />

3. Raleigh, J. A., et al. (1998) Hypoxia and vascular<br />

endothelial growth factor expression in human<br />

squamous cell carcinomas using pimonidazole as a<br />

hypoxia marker. Cancer Res 58: 3765-8<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

YIA applicant<br />

day1<br />

Plenary Session on Excellent and Late Breaking Abstracts


YIA applicant<br />

54 Introduction:<br />

WarSaW, poland May 26 – 29, 2010<br />

A leukocyte ligand of vascular adhesion protein-1 as an imaging tool in PET<br />

Autio A. (1) , Saanijoki T. (1) , Sipilä H. (1) , Jalkanen S. (2) , Roivainen A. (3) .<br />

(1) Turku PET Centre,<br />

(2) MediCity Research Laboratory, National Public Health Institute,<br />

(3) Turku PET Centre, Turku Centre for Disease Modeling.<br />

akauti@utu.fi<br />

Vascular adhesion protein-1 (VAP-1)<br />

is both an endothelial glycoprotein and a semicarbazide-sensitive<br />

amine oxidase (SSAO) enzyme playing<br />

a critical role in leukocyte trafficking to the sites of<br />

inflammation. Although VAP-1 was identified more<br />

than 15 years ago, the leukocyte ligand has remained<br />

unknown until very recently. Last year it was shown<br />

that Siglec-10 (sialic acid-binding immunoglobulinlike<br />

lectin) expressed on a subpopulation of lymphocytes<br />

can bind to VAP-1 and serve as its substrate [1].<br />

According to phage display screening and structural<br />

modeling also Siglec-9 expressed on granulocytes<br />

and monocytes interacts with VAP-1. In this study,<br />

we investigated a Siglec-9 peptide as a potential imaging<br />

tool in positron emission tomography (PET).<br />

Methods: A cyclic peptide binding to recombinant<br />

human VAP-1 was conjugated with DOTA-chelator<br />

through PEG-linker and 68Ga-labeled for PET<br />

studies as previously described [2]. The interaction<br />

between VAP-1 and 68Ga-Siglec-9 peptide<br />

was evaluated in vitro in human plasma samples<br />

possessing different SSAO levels. The VAP-1 specificity<br />

was further tested with competition assay in<br />

mice bearing melanoma xenografts by PET imaging<br />

and autoradiography. In vivo imaging of inflammation<br />

was examined in a rat model. All in vivo<br />

studies were confirmed by ex vivo measurements.<br />

Standardized uptake value<br />

a Tumor<br />

b Tumor<br />

c<br />

500<br />

P


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

Clinical translation of ex vivo sentinel lymph node mapping for colorectal cancer using<br />

invisible near-infrared fluorescence light<br />

Hutteman M. (1) , Choi H. S. (2) , Mieog S. (1) , Van Der Vorst J. (1) , Ashitate Y. (2) , Kuppen P. (1) , Löwik C.W.G.M. (1) , Van De Velde C. (1) ,<br />

Frangioni J. (2) , Vahrmeijer A. (1) .<br />

(1) Leiden University Medical Center,<br />

(2) BIDMC / Harvard Medical School.<br />

m.hutteman@lumc.nl<br />

Introduction: Sentinel lymph node (SLN) mapping<br />

in colorectal cancer may have prognostic and therapeutic<br />

significance, however, currently available<br />

techniques are not optimal. We hypothesized that<br />

the combination of invisible near-infrared (NIR)<br />

fluorescent light and ex vivo injection could solve<br />

remaining problems in the field.<br />

Methods: The FLARE imaging system was used<br />

for real-time identification of SLNs after injection<br />

of the lymphatic tracer HSA800 in the colon and<br />

rectum of (n = 4) pigs. A total of 32 SLN mappings<br />

were performed in pigs, in vivo and ex vivo after oncologic<br />

resection, using an identical injection technique.<br />

Guided by these results, SLN mappings were<br />

performed in ex vivo tissue specimens of 6 colorectal<br />

cancer patients undergoing resection (Figure 1).<br />

Results: Using NIR fluorescent imaging, lymph<br />

flow could be followed in real-time from the injection<br />

site to the SLN. Pre-clinically, in pigs, the SLN<br />

was identified in 32/32 (100%) SLN mappings in<br />

both colon and rectum, under both in vivo and ex<br />

COLON<br />

RECTUM<br />

Color<br />

Fig1.: NIR Fluorescence-Guided SLN Mapping in Patients with Colorectal Cancer<br />

vivo conditions. Clinically, SLNs were identified in<br />

all patients (n = 6) using the ex vivo strategy (Figure<br />

1). No false negatives were found. SLNs were<br />

identified within 5 minutes after injection of the<br />

fluorescent dye.<br />

Conclusions: The current study shows proof of<br />

principle that ex vivo NIR fluorescence-guided SLN<br />

mapping can provide high-sensitivity, rapid, and accurate<br />

identification of SLNs in colon and rectum.<br />

This will permit optimized, non-FDA-approved NIR<br />

fluorescent lymphatic tracers to be translated immediately<br />

to the clinic.<br />

NIR Fluorescene Color-NIR Merge<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

YIA applicant<br />

day1<br />

Plenary Session on Excellent and Late Breaking Abstracts


YIA applicant<br />

56<br />

WarSaW, poland May 26 – 29, 2010<br />

Development of strategies for in vivo MRI and Optical Imaging of neural stem cells distribution<br />

for the treatment of traumatic spinal cord injury<br />

Lui R. (1) , Merli D. (1) , Libani I. V. (1) , Madaschi L. (1) , Marra F. (1) , Marfia G. (1) , Carelli S. (1) , Clerici M. S. (2) , Gorio A. (3) ,<br />

Lucignani G. (1) , Ottobrini L. (1) .<br />

(1) University of Milan,<br />

(2) University of Milan and Don Gnocchi Foundation IRCCS,<br />

(3) University of Milan and Humanitas IRCCS, Rozzano, Milan.<br />

ramona.lui@unimi.it<br />

Introduction: The use of adult stem cells in cell-mediated<br />

therapies is an area of considerable interest within<br />

tissue regeneration research. However, important<br />

parameters such as the distribution of the injected<br />

cells, cell survival, target organ localisation, cell proliferation<br />

and differentiation cannot be evaluated in<br />

vivo. Here we propose multiple labelling protocols<br />

for in vivo visualisation by MRI, and Optical fluorescence<br />

imaging of murine neural stem cells (mNSC)<br />

originating from the subventricular zone of the adult<br />

murine brain in spinal cord injury animal models.<br />

Methods: mNSC were isolated and cultured as<br />

described[1]. Cells were directly labelled for 24 hours<br />

with 200 μg Fe/ml of SPIOs (Endorem®) in presence<br />

of different amount of Protamine Sulphate (PS) (ratio<br />

Fe/PS 1:0,025; 1:0,05) and immediately analysed<br />

for viability, iron content (Perl’s Staining and spectrophotometer<br />

analysis), morphology, staminality<br />

and differentiation capability. Labelled cells were<br />

injected into the tail vein of a spinal cord injury<br />

murine model and followed by MRI for a month to<br />

visualize their distribution. Initial cell distribution<br />

was also followed with scintigraphy after cell labelling<br />

with 111Indium-oxine (60 μCi/106 cells). Cells<br />

localization, distribution e viability, over time, were<br />

analysed in vivo with CCD camera after injection of<br />

mNCS infected with a viral vector expressing Luciferase<br />

under a PGK promoter (PLW vector).<br />

Results: mNSC incubated for 24 with 200 μg Fe/ml<br />

Endorem® did not show significant differences in<br />

terms of viability and proliferation rate between labelled/non-labelled<br />

cells in presence or absence of<br />

different amount of PS[2]. The percentage of viable<br />

cells was 85% for Fe/PS 1:0.025 ratio, and 77% for<br />

Fe/PS 1:0.05 ratio compared to the 95% of the control.<br />

On the contrary, the percentage of iron-positive<br />

cells increased in proportion to the PS content in the<br />

medium. In particular we obtained 94,5% of ironpositive<br />

cells in the samples incubated with Fe/PS<br />

1:0.025 ratio and 99% iron-positive cells in the samples<br />

incubated with Fe/PS 1:0.05 ratio. The iron content<br />

increased from 110 pg Fe/cell to 210 pg Fe/cell<br />

increasing PS concentration. In both cases, labelled<br />

cells were able to give rise to floating neurospheres<br />

imaging life<br />

as observed by optical microscopy after further 5<br />

days of culture, demonstrating their maintenance<br />

of the self-renewal capability. Immune fluorescence<br />

analysis for Nestin confirmed these data. Differentiation<br />

capability was also maintained as confirmed<br />

by β-TubulinIII and GFAP expression. Scintigraphic<br />

techniques confirmed initial distribution to lung,<br />

spleen and liver while MRI showed iron signal due<br />

to stem cell localization into the lesion site at 21 and<br />

28 days after injection. Neural stem cells, infected<br />

with the viral vector PLW, were detected up to one<br />

week after i.v. injection within the lesion of injured<br />

mice. Infected cells intramedullary injected as control<br />

were detected at the same timepoint as well.<br />

Conclusions: These results showed that adult neural<br />

stem cells can be efficiently labelled with different<br />

molecules without significantly perturbing physiological<br />

stem cell features and self-renewal capability.<br />

This labelling protocols can be applied for<br />

the in vivo visualisation by MRI, scintigraphy, and<br />

Luminescence imaging of the distribution of stem<br />

cells after their transplantation into murine model<br />

of disease.<br />

Acknowledgement: this work is supported by CAR-<br />

IPLO Foundation grant. Dr Lui R is supported by a<br />

fellowship from the Doctorate School of Molecular<br />

Medicine, University of Milan.<br />

References:<br />

1. Bottai D et al. Mol Med (2008) 14:634-644<br />

2. Politi LS et al. Neuroradiology (2007) 49:523–534


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

Validation of bone marrow-derived stromal cell graft position by colocalisation of histology,<br />

bioluminescence and magnetic resonance imaging.<br />

De Vocht N., Van Der Linden A. .<br />

Universiteit Antwerpen – Bio Imaging Lab<br />

nathalie.devocht@ua.ac.be<br />

Introduction: The use of stem cell transplantation<br />

as a therapeutic tool to treat neurodegenerative disorders<br />

(e.g. traumatic brain injury, stroke, multiple<br />

sclerosis) has gained increasing interest over the<br />

last decade. However, a profound knowledge of cell<br />

implant migration, differentiation and survival will<br />

be necessary to understand the physiological mechanisms<br />

involved in regeneration of injured brain tissue.<br />

For this, the development of multimodal cell<br />

imaging techniques, both in vivo and post-mortem,<br />

is currently of highest importance.<br />

Methods: We optimised a multimodal labelling<br />

strategy for bone marrow-derived stromal cells<br />

(BMSC), based on (i) genetic modification with<br />

both the eGFP and Luciferase reporter genes, and<br />

(ii) endocytic uptake of blue fluorescent magnetite-containing<br />

micron-sized particles (MPIO).<br />

This combined labelling strategy allows unambiguous<br />

identification of cell localisation, survival<br />

and differentiation using pre-mortem bioluminescence<br />

/ magnetic resonance imaging (BLI/MRI)<br />

and post-mortem histological analysis.<br />

Results: For this study, we labelled Luciferase/<br />

eGFP-expressing BMSC (BMSC-Luc/eGFP, Bergwerf<br />

et al. 2009) with 1.63 µm fluorescent (Glacial<br />

Blue) MPIOs from Bangs Laboratories. MPIO labelling<br />

of BMSC-Luc/eGFP did not influence their<br />

phenotypic properties and self-renewal capacity.<br />

In order to evaluate the suitability of these MPIO<br />

particles for in vivo multimodal imaging, we implanted<br />

4 x 105 MPIO labelled (n = 15) or unlabelled<br />

(n = 15) cells by stereotactic injection in the<br />

right hemisphere (DV: 2mm beneath the dura; ML:<br />

2mm; AP: -2mm) of syngeneic male FVB mice (10-<br />

12 weeks old). Survival of unlabelled and MPIOlabelled<br />

MSC-Luc/eGFP implants was monitored<br />

during 2 weeks by BLI, which demonstrated equal<br />

survival of both cell populations. Moreover, T2-<br />

and T2*-weighted MR images clearly demonstrated<br />

the presence of iron oxide particles in mouse<br />

brain. Additionally, combined immunohistochemistry<br />

and fluorescence microscopy was able to confirm<br />

cell identity and the presence of MPIO particles<br />

within BMSC-Luc/eGFP grafts.<br />

Conclusions: We here demonstrate that combining<br />

three labelling strategies allows unambiguous identification<br />

of MSC following implantation in mouse<br />

brain. The MPIO-labelling allows for pre-mortem<br />

MRI detection of cell implant localisation. In addition,<br />

BMSC survival was followed up using BLI up<br />

to two weeks post-injection. Finally, the combination<br />

of eGFP expression and MPIO-labelling allows<br />

for discrimination of cell implants (eGFP+/MPIO+)<br />

from inflammatory cells (green background fluorescence/MPIO-)<br />

and free particles (eGFP-/MPIO+),<br />

enabling an improved qualitative histological analysis<br />

of cell implantation.<br />

Acknowledgement: This work was supported in part<br />

by: (i) the EC-FP6-NoE funded DiMI (ii) the Flemish<br />

government-funded IWT-SBO BRAINSTIM<br />

Project and (iii) the FWO Flanders.<br />

References:<br />

1. Bergwerf I, De Vocht N, et al., BMC Biotechnology<br />

2009; 9:1<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

day1<br />

Plenary Session on Excellent and Late Breaking Abstracts


DAY<br />

2


• <strong>ESMI</strong> Plenary Lecture 3: Theodorus W.J. Gadella Jr. (Amsterdam, The Netherlands)<br />

New probe-based strategies for quantitative microscopy of signaling dynamics in single cells<br />

Chairs: Vasilis Nziachristos (Munich, Germany) , Bengt Långström (Uppsala, Sweden)<br />

• Parallel Session 5: Neuroscience II<br />

Chairs: Markus Rudin (Zürich, Switzerland), Veerle Baekelandt (Leuven, Belgium),<br />

Mathias Hoehn (Cologne, Germany)<br />

• Parallel Session 6: Cardiovascular I – together with ESR<br />

Chairs: Tony Lahoutte (Mons, Belgium), Nicolas Grenier (Bordeaux, France)<br />

• Parallel Session 7: Probes II – together with EANM<br />

Chairs: Frédéric Dollé (Orsay, France), Denis Guilloteau (Tours, France)<br />

• Parallel Session 8: Gene and Cell Based Therapies – together with CliniGene<br />

Chairs: Ludwig Aigner (Salzburg, Austria), Cornel Fraefel (Zürich, Switzerland)<br />

• <strong>ESMI</strong> Plenary Lecture 4: Hans-Jürgen Wester (Munich, Germany)<br />

Molecular imaging of CXCR4 receptors<br />

Chairs: Adriana Maggi (Milano, Italy), Adriaan Lammertsma (Amsterdam, The Netherlands)<br />

• Plenary Session on Current Contribution of Imaging Technologies to Drug Development<br />

Chairs: Adriana Maggi (Milano, Italy), Adriaan Lammertsma (Amsterdam, The Netherlands)<br />

Day 2 - Friday May 28, 2010


60<br />

WarSaW, poland May 26 – 29, 2010<br />

Chairman of section of Molecular Cytology, Director Centre for Advanced Microscopy<br />

Swammerdam Institute for Life Sciences, University of Amsterdam, The Netherlands<br />

www.mc.bio.uva.nl<br />

Dr. Gadella is founder/director of the Centre for Advanced Microscopy (CAM) at the University of Amsterdam<br />

and full professor in Molecular Cytology chairing a group of 25-30 scientists including 4 assistant<br />

professors. The CAM and chairgroup are positioned within the Swammerdam Institute for Life Science at<br />

the Science Faculty of the University of Amsterdam and fully integrated within the Netherlands Institute for<br />

Systems Biology founded in 2007. Dr Gadella personally supervises a research team on spatiotemporal cell<br />

signaling. His team wants to understand how cells can achieve and maintain a local signal in order to drive<br />

morphogenesis, to define new cytoskeletal anchorage or vesicle-docking sites. The focus is on signal flow<br />

across and in the plane of the membrane of living mammalian cells. To this end genetic encoded fluorescent<br />

biosensors are employed and the in situ molecular interactions between signaling molecules including phospholipid-second<br />

messengers, receptors, G-proteins and downstream targets are analyzed. By multiparameter<br />

imaging approaches several signaling events are visualized and quantified simultaneously in individual living<br />

cells with sub-second temporal and submicron spatial resolution. The in situ cellular imaging research<br />

heavily depends on advanced bioimaging. To this end advanced automated microscopy approaches such<br />

as fluorescence resonance energy transfer (FRET) microscopy (including bleaching-, spectral-, ratio- and<br />

fluorescence lifetime-imaging approaches), fast live cell microscopy (including spinning disk, line-scanning<br />

and controlled light exposure confocal microscopy, and total internal reflection microscopy (TIRF)), and<br />

photochemical microscopy approaches such as photoactivation, uncaging, fluorescence recovery after photobleaching<br />

(FRAP), fluorescence loss in photobleaching microscopy (FLIP), fluorescence correlation spectroscopy<br />

(FCS), cross-correlation (FCCS), lifetime-correlation (FCLS) have been implemented developed<br />

and applied to quantifying cell signalling phenomena. Currently, the palette of advanced imaging instrumentation<br />

is expanded with superresolution localization microscopy approaches such as PALM and STORM.<br />

The research group is funded from university resources (roughly 40%), national grant agencies (NWO-ALW,<br />

CW, FOM and STW) and several international grants (ESF and EU-FP6/7 projects including integrated projects,<br />

STREPs and TMR programs).<br />

The Gadella lab has numerous (inter)national collaborations in the field of signal transduction (Dr. C. Hoffman,<br />

Univ. Würzburg; M. Wymann, Univ. Basle; K. Jalink, NKI Amsterdam) and collaborations on probe<br />

development (dr. C. Schultz, EMBL Heidelberg; dr. K. Lukyanov & D. Chudakov, Moscow) and collaborations<br />

on advanced light microscopy (prof. van Noorden (AMC); dr. A. Houtsmuller, Erasmus MC; dr. H.<br />

Gerritsen, Utrecht; dr. Dobrucki, Univ. Krakow; prof. M. Carmo Fonseca, Lisbon). In 2007 prof Gadella<br />

was elected president of the Netherlands Society for Advanced Microscopy. Recently he was appointed as<br />

national coordinator of the advanced light microscopy activities of the Netherlands for the new large EU<br />

infrastructure (ESFRI) programme EuroBioimaging that recently entered the startup phase.<br />

imaging life<br />

Theodorus W.J. Gadella Jr.


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

New probe-based strategies for quantitative microscopy of signaling dynamics in single cells<br />

Gadella T.W.J. jr, Goedhart J., Van Weeren L., Crosby, K., Hink M.A.<br />

Section of Molecular Cytology and Centre for Advanced Microscopy, Swammerdam Institute for Life Sciences & Netherlands Institute for Systems Biology,<br />

University of Amsterdam, Science Park 904, NL-1098 XH Amsterdam.<br />

Th.W.J.Gadella@uva.nl<br />

Since the cloning of the green fluorescent protein<br />

from Aequoria victoria, numerous fluorescence<br />

microscopy applications have been described in<br />

the literature. By exploiting the spectroscopic axis<br />

in microscopy (excitation/emission wavelength,<br />

fluorescence lifetime) utilizing spectral variants<br />

of GFP colocalization and FRET applications<br />

were enabled. In the other direction, the spectroscopical<br />

axis in microscopes can also be used to<br />

obtain novel GFP variants with optimized properties.<br />

Here we report a screening method that,<br />

in addition to fluorescence intensity, quantifies<br />

the excited state lifetime of a fluorescent protein,<br />

providing a direct measure for the quantum yield<br />

of the fluorescent protein. The novel approach<br />

was used to screen a library of cyan fluorescent<br />

protein (CFP) variants yielding the brightest cyan<br />

fluorescent protein variant described thus far<br />

which we dubbed mTurquoise. mTurquoise has a<br />

marked increased quantum yield of 0.84, and a<br />

seriously increased lifetime of 3.7 ns. Because of<br />

its monoexponential decay and high R0 for FRET<br />

to SYFP2 (5.7 nm), it is the preferred donor in<br />

FRET studies. In addition, the lifetime screening<br />

method also yielded several other CFP lifetime<br />

variants with identical spectra. It is demonstrated<br />

that three spectrally identical CFP variants can<br />

be separated in single FLIM experiment in living<br />

cells and their distribution & molecular stoichiometry<br />

can be quantified. The triple unmixing<br />

technique using the new lifetime variants can be<br />

combined with spectral separation of other GFP<br />

color variants with a great potential for multiparameter/multiplexed<br />

imaging.<br />

We apply the different probes for the study of<br />

GPCR-triggered signaling in single mammalian<br />

cells. The design of a Gq FRET sensor reporting<br />

on activation by endogenous expressed receptors,<br />

multiparametric imaging of lipid-derived second<br />

messengers, PtdInsP2-dependent PLC translocation,<br />

and RhoGEF activation using a variety of<br />

FRET-, ratio imaging- and TIRF-microscopic applications<br />

will be presented.<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

day2<br />

<strong>ESMI</strong> Plenary Lecture 3 by Theodorus Gadella


62<br />

WarSaW, poland May 26 – 29, 2010<br />

Potential to use transgenic animals for imaging of neurological diseases<br />

Aigner L. .<br />

Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Austria<br />

ludwig.aigner@pmu.ac.at<br />

Transgenic animal models represent an elegant and<br />

innovative tool to image the multitude of events<br />

including degenerative, inflammatory and regenerative<br />

responses in the brain. However, there are<br />

currently no transgenic animal models that would<br />

image specifically one or another neurological<br />

disease. Typically, a transgenic reporter animal<br />

highlights the activity of a gene promoter that is<br />

specifically active in one or another cell type during<br />

a certain physiological process, upon response<br />

to injury, along the course of neurodegeneration<br />

or during a regenerative response. Thus, transgenic<br />

animal models allow the imaging of physiological<br />

and pathophysiological responses that are<br />

associated with neurological / neurodegenerative<br />

diseases, but not the disease itself. For example,<br />

currently available models allow the elucidation of<br />

several aspects of neurodegeneration: 1) Inflammatory<br />

events can be visualized using transgenic<br />

animals with the TLR2 promoter controlling the<br />

expression of reporter genes; herein, these reporters<br />

are up-regulated in response to the onset<br />

imaging life<br />

of an inflammatory response hand in hand with<br />

endogenous TLR2 and microglial activation (Lalancette-Hebert,<br />

Gowing et al. 2007). 2) Reactive<br />

gliosis may be pictured using GFAP-reporter mice<br />

as a result of stroke in living animals (Cordeau,<br />

Lalancette-Hebert et al. 2008). 3) Hypoxia might<br />

be imaged due to the application of models like<br />

the HRE-GFP transgenic animals where cells that<br />

suffer from oxygen deprivation express reporter<br />

proteins induced by low-oxygen-responsive transcription<br />

factors (Berchner-Pfannschmidt, Frede<br />

et al. 2008). 4) Neuroplasticity after brain ischemia<br />

comprises the recovery of dendritic structure, and<br />

this event may be imaged using transgenic models<br />

with fluorescent proteins expressed throughout a<br />

distinct cell population and including the cellular<br />

protrusions (Li and Murphy 2008). 5) Neurogenesis<br />

and the migration of young neurons can be<br />

visualized using DCX-promo luciferase mice. This<br />

presentation will provide a summary and an overview<br />

on the current state of transgenic models in<br />

the field of neurological diseases.


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

Specific cell labeling with imaging reporters in vivo by viral vectors: the challenges<br />

Reumers V. (1) , Ibrahimi A. (2) , Toelen J. (2) , Aelvoet S. A. (1) , Van Den Haute C. (1) , Debyser Z. (2) , Baekelandt V. (1) .<br />

(1) Neurobiology and Gene Therapy, Katholieke Universiteit Leuven,<br />

(2) Molecular Virology and Gene Therapy, Katholieke Universiteit Leuven. From MoSAIC, the Molecular Small Animal Imaging Center, K.U.Leuven, Belgium<br />

Veerle.Baekelandt@med.kuleuven.be<br />

Viral vectors are extremely efficient tools for gene<br />

transfer into living cells. We are using lentiviral<br />

vectors and adeno-associated viral (AAV) vectors<br />

for stable overexpression and suppression of gene<br />

expression in rodent brain. Stereotactic injection<br />

of these viral vectors into different brain regions<br />

of mouse and rat brain induces overexpression or<br />

inhibition of disease-related genes, which can be<br />

used for target validation, functional genomics and<br />

generation of disease models. We have invested<br />

over the last years into reporter gene technology<br />

and non-invasive imaging in rodent brain. We have<br />

optimized bioluminescence imaging to monitor<br />

gene expression in mouse brain and to track<br />

neuronal stem cell migration and differentiation.<br />

In order to specifically target certain neuronal cell<br />

populations we have recently developed conditional<br />

lentiviral and AAV viral vectors based on Cremediated<br />

recombination. Application of these viral<br />

vectors in specific cre-transgenic mouse strains<br />

allows to specifically label endogenous neural stem<br />

cells in the subventricular zone or dopaminergic<br />

cells in the substantia nigra. This approach holds<br />

promise to non-invasively follow up endogenous<br />

neurogenesis and neurodegeneration over time.<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

day2<br />

Parallel Session 5: NEUROSCIENCE II


YIA applicant<br />

64<br />

WarSaW, poland May 26 – 29, 2010<br />

Optimization of an MRI method to measure microvessel density: application to monitor<br />

angiogenesis after stroke<br />

Boehm-Sturm P. (1) , Adamczak J. (1) , Farr T. D. (1) , Jikeli J. (2) , Kallur T. (1) , Hoehn M. (1) .<br />

(1) MPI for Neurological Research in Cologne, Germany<br />

(2) Center of Advanced European Studies and Research in Bonn.<br />

philipp.boehm-sturm@nf.mpg.de<br />

Introduction: Angiogenesis is the growth of new vessels<br />

from an existing vascular network and is highly<br />

upregulated following ischemia. Increases in microvessel<br />

density (MVD) have been reported to occur<br />

in the peri-infarct zone of stroke patients, which was<br />

correlated with increased survival time [1]. Steadystate-contrast-enhanced<br />

(SSCE) MRI provides<br />

the ability to monitor MVD in vivo. To do so, the<br />

changes in relaxivities ΔR2/ΔR2* due to an i.v. injection<br />

of a paramagnetic blood pool contrast agent are<br />

evaluated by spin-echo (SE) and gradient-echo (GE)<br />

imaging, and the MVD index Q=ΔR2/(ΔR2)2/3 is<br />

mapped [2,3]. However, noise in the MR images can<br />

significantly distort the results of the method. Therefore,<br />

the goal of this study is to optimize the SSCE<br />

MRI parameters in simulations in order to find a<br />

noise-resistant protocol. We aim to apply the method<br />

to monitor angiogenesis after stroke in rats and to<br />

validate this by immunohistochemistry.<br />

Left: simulated relative error in Q (in %) over TESE and TEGE at a fixed noise level; the arrow indicates the optimal set of TEs. Right: lesion in SE<br />

image 48 h after MCAO (a) and Q at 7 days prior (b), and 7 (c), and 14 days (d) post MCAO of the same animal. Arrow indicates an area with high Q<br />

value. Typical RECA (red)/BrdU (green) (e) and Hoechst(blue)/RECA(red)/laminin (green) (f) fluorescence microscopy images close to the infarct.<br />

Methods: Q-maps were simulated for different levels<br />

of noise and echo times TESE/TEGE of the SE/<br />

GE images. Male Wistar rats (n=7) were scanned<br />

with SSCE MRI 7 days prior, and 7 and 14 days post<br />

60 min transient MCAO. An SE image was acquired<br />

48 h post MCAO to depict the lesion extent. Rats<br />

received Bromodeoxyuridine (BrdU) injections between<br />

3-7 days post MCAO in order to label proliferating<br />

cells. Following the final SSCE MRI scan<br />

at 14 days, animals were perfused and brain sections<br />

stained for BrdU, rat endothelial cell antigen<br />

(RECA), and laminin.<br />

imaging life<br />

Results: The simulations revealed that the error in<br />

Q is minimal for a TESE of 30-40 ms and a TEGE of<br />

10-15 ms. Areas with increased Q could be detected<br />

close to the infarct in a few of the rats as early as 7<br />

days and in most of the rats at 14 days post MCAO.<br />

BrdU/RECA double staining was established to<br />

identify newly formed endothelial cells and laminin<br />

staining confirmed the presence of an intact basement<br />

membrane in the vessels.<br />

Conclusions: Our results show that scanner noise<br />

can severely limit the capabilities of SSCE MRI but<br />

a set of optimal scan parameters can minimize this<br />

problem. The method has been shown to monitor<br />

potential changes of the microvasculature longitudinally<br />

and non-invasively in the ischemic rat brain.<br />

It will in the future allow to study neo-angiogenesis<br />

in the course of (stem)cell-based therapies after cerebral<br />

lesions.<br />

Acknowledgements: Generous supply of ENDOREM<br />

by Drs. C. Corot and P. Robert is gratefully acknowledged.<br />

This work was supported in part by grants from<br />

the European Union under FP6 program (StemStroke,<br />

LSHB-CT-2006-037526), and under FP7 program<br />

(ENCITE, 201842), and an Alexander von Humboldt<br />

Research Fellowship to TDF.<br />

References:<br />

1. Krupinski et al, Stroke (1994) 25:1794-1798;<br />

2. Jensen et al., MRM (2000) 44:224–230;<br />

3. Wu et al., NMR Biomed (2004) 17:507–512


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

MRI study of intra-arterial bone marrow-derived macrophage administration after acute<br />

focal ischemic stroke in rats<br />

Riou A. (1) , Chauveau F. (1) , Cho T.H. (1) , Nataf S. (2) , Berthezene Y. (1) , Nighoghossian N. (1) , Wiart M. (1) .<br />

(1) CREATIS-LRMN : CNRS UMR5220, INSERM U630, Univ. Lyon1, INSA-Lyon, France<br />

(2) CNRS UMR5220, INSERM U630.<br />

adrien.riou@live.fr<br />

Introduction: In the past few years numerous studies<br />

have been published to assess if cell-based therapy<br />

after stroke can enhance long-term recovery. It<br />

is well established that cerebral ischemia results in<br />

a complex inflammatory cascade that mainly involves<br />

cells from the mononuclear phagocyte system1,<br />

although the beneficial or deleterious effect<br />

of this activation following stroke is still a matter<br />

of controversy. Furthermore, therapeutic benefits<br />

gained from cell-based therapy depend on migration<br />

and localization of grafted cells within the target<br />

tissue that is closely related to the cell delivery<br />

route2 and therapeutic time window3 chosen for<br />

the therapy. The aim of this study was to assess the<br />

feasibility of in vivo early intra-arterial (IA) bone<br />

marrow-derived macrophage (BMDM) administration<br />

for acute focal ischemic stroke treatment, using<br />

multiparametric magnetic resonance imaging<br />

(MRI).<br />

Methods: BMDM were obtained from a sacrificed<br />

littermate of recipient rats by flushing out one<br />

hindlimb. Bone marrow cells were then seeded on<br />

uncoated flasks at 5 x 105 cells/ml in medium supplemented<br />

with murine macrophage-colony stimulating<br />

factor and flt3 ligand (mM-CSF and flt3L at<br />

10ng/ml) for one week. At day 6, cells were labeled<br />

with anionic superparamagnetic nanoparticles<br />

overnight (AMNP, CNRS UMR 7612, Paris, France,<br />

[Fe]=1mM). Male Sprague Dawley rats (250 to 350g,<br />

n=17) were subjected to 1-h intraluminal transient<br />

middle cerebral artery occlusion (tMCAO) (n=12)<br />

or sham procedure (n=5). IA administration of<br />

AMNP-labeled cells was performed at the time of<br />

reperfusion in all tMCAO animals and in 3 shams<br />

animals (4 million in 1-ml except for one sham: 1<br />

million in 1-ml). Ischemia / reperfusion and cell<br />

injection were monitored by transcranial laser doppler<br />

flowmetry. MRI was performed on a Bruker<br />

Biospec 7T/12cm magnet at D0 just after cells administration<br />

and from D1 to D9. The MR exam included<br />

T2-, T2*-, diffusion- and perfusion-weighted<br />

imaging and multi-echo 3D imaging. Animals<br />

were sacrificed after completion of the MRI exams<br />

and brains were prepared for immunohistological<br />

analysis with Prussian blue for iron detection and<br />

Ox-42 for macrophage detection.<br />

Results: IA administration in tMCAO group lead to<br />

heterogeneous results: 3 rats died following injection,<br />

2 had cells detectable only in the temporalis<br />

muscle and 7 had cells detectable in the ipsilateral<br />

parenchyma but with heterogeneous patterns: 3 had<br />

a widespread persistent hypointense signal distribution,<br />

while 4 had only few local spots of signal<br />

loss on follow-up scans. Furthermore, IA administration<br />

in the sham group caused lesion formation<br />

on follow-up scans in all injected animals (n=3),<br />

even when cell number was decreased, as opposed<br />

to the non-injected sham rats that did not have any<br />

lesions. Immunohistological analysis is in progress<br />

to ascertain iron-labeled macrophage localization.<br />

Conclusions: Our results were consistent with that<br />

of previous studies2, 4 showing that IA delivery<br />

route efficiently brought a large number of cells to<br />

the brain soon after transplantation, but severely<br />

increased mortality. More importantly, lesions observed<br />

in the sham group (undemonstrated to date<br />

to our knowledge) suggested that this high mortality<br />

rate resulted from cell embolisation in cerebral<br />

vessels leading to formation or worsening of the<br />

ischemic lesion. This result points out a serious limitation<br />

for the translation of this cell delivery route<br />

into the clinics.<br />

References:<br />

1. Huang J, et al. Surg Neurol.;66:232-45 (2006).<br />

2. Li L, et al. J Cereb Blood Flow Metab (2009).<br />

3. de Vasconcelos Dos Santos A, et al. Brain Res.1306:149-<br />

58 (2010).<br />

4. Walczak P, et al. Stroke. 2008;39:1569-74 (2008).<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

day2<br />

Parallel Session 5: NEUROSCIENCE II


YIA applicant<br />

66<br />

WarSaW, poland May 26 – 29, 2010<br />

Inactivated paramagnetic tissue plasminogen activator predicts thrombolysis outcome<br />

following stroke<br />

Gauberti M. (1) , Montagne A. (1) , Vivien D. (1) , Orset C. (1) .<br />

INSERM U919 SP2U Serine protease and pathophysiology of the neurovascular unit, France<br />

gauberti@cyceron.fr<br />

Introduction: Ischemic stroke is the third leading<br />

cause of death in developed countries. Despite numerous<br />

clinical trials, tissue plasminogen activator<br />

(tPA) induced thrombolysis remains the only treatment<br />

of the acute phase. However there is growing<br />

body of evidences that parenchymal tPA has deleterious<br />

effects including increased risk of intra cranial<br />

hemorrhages and neurotoxicity. To limit these side<br />

effects, only rigorously selected patients (onset of<br />

symptoms < 4.5 hrs, age < 85 hrs, etc.) benefit from<br />

thrombolysis, with more than 90% of patients being<br />

untreated. In previous in vitro studies we showed<br />

that vascular tPA can cross the intact blood brain<br />

barrier (BBB) by Low Density Lipoprotein Receptor<br />

Protein (LRP) mediated transcytosis [1]. Accordingly,<br />

increasing data of the literature suggest that<br />

BBB specific permeability to tPA could be critical for<br />

brain outcome following thrombolysis [2] [3].<br />

Figure 1. Ex-vivo NIRF brain imaging of fluorescent tPA and albumin<br />

administered at 1 or 4 hours after permanent middle cerebral artery<br />

occlusion in mice. Unlike albumin which has the same molecular<br />

weight, tPA can cross the BBB at 4 hours post ischemia.<br />

Results: Here in a model of permanent middle cerebral<br />

artery occlusion in mice, we show that intravenous<br />

tPA aggravates ischemic lesion size and<br />

BBB permeability if administrated at 4 hours but<br />

not at 1 hour after ischemia. In parallel, although<br />

BBB remains impermeable to albumin, we provide<br />

evidences both by microscopic epifluorescence and<br />

near infrared fluorescence (NIRF) brain imaging of<br />

a passage of tPA across the BBB only when administrated<br />

at 4 hours post ischemia. Using MRI and inactivated<br />

paramagnetic tPA as molecular probe, we<br />

imaging life<br />

are tempting to demonstrate that such type of brain<br />

imaging could allow us to pre-select stroke patients<br />

who would not be susceptible to haemorrhagic transformations<br />

following rtPA-induced thrombolysis.<br />

Conclusions: Non invasive imaging of inactivated<br />

paramagnetic tPA could allow clinicians to include<br />

stroke patients for thrombolysis with objective brain<br />

imaging data independently of the time post-stroke<br />

onset. As long as the BBB remains impermeable to<br />

tPA, thrombolysis seems safe.<br />

References:<br />

1. Benchenane et al.; Circulation 111:2241-2249 (2005).<br />

2. Benchenane et al.; Stroke 38:1036 (2005).<br />

3. Roussel et al.; Thromb Haemost. 102:602-608 (2009).


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

Matrix metalloproteinase-9 – a possible therapeutic target in intractable epilepsy<br />

Wilczynski G. .<br />

Nencki Institute, Poland<br />

g.wilczynski@nencki.gov.pl<br />

Introduction: Temporal lobe epilepsy (TLE) is a<br />

chronic, devastating, pharmacologically intractable<br />

disease in which aberrant synaptic plasticity<br />

plays a major role. Recently, MMP-9, a matrix<br />

metalloproteinase, has been implicated in synaptic<br />

plasticity, long-term potentiation and learning and<br />

memory formation. We asked, whether MMP-9<br />

might play a pathogenic role in epileptogenesis.<br />

Methods: High resolution light- and electronmicroscopic<br />

immunocytochemistry, in situ hybridization,<br />

in situ zymography; use of transgenic<br />

animals; antibody microarrays<br />

Results: Our study revealed MMP-9 as a novel<br />

synaptic enzyme, and a key pathogenic factor<br />

in two distinct animal models of TLE: kainateevoked-epilepsy<br />

and pentylenetetrazole (PTZ)<br />

kindling-induced epilepsy. In particular, sensitivity<br />

to PTZ-induced epileptogenesis is decreased<br />

in MMP-9 knockout (KO) mice, whereas it is<br />

increased in MMP-9-overexpressing rats. Moreover,<br />

confocal- and immunoelectron-microscopic<br />

analyses demonstrated that MMP-9 associated<br />

with hippocampal dendritic spines bearing asymmetric<br />

(excitatory) synapses. In addition, both<br />

MMP-9 protein levels as well as its enzymatic activity<br />

became strongly increased upon seizures.<br />

Furthermore, MMP-9-deficiency diminished seizure-evoked<br />

pruning of dendritic spines, and it<br />

decreased aberrant synapse formation following<br />

mossy-fibers sprouting.<br />

We then asked whether MMP-9 could play an important<br />

role also in human intractable epilepsy.<br />

Accordingly, we have studied the expression and<br />

localization of MMP-9 in samples of human epileptic<br />

brain tissue, obtained upon surgical treatment<br />

of childhood intractable epilepsy. By antibody<br />

microarrays, we found MMP-9, but none of<br />

the other seven MMPs studied, to be consistently<br />

upregulated in epileptic lesions, as compared to<br />

autopsy brain tissue. By immunohistochemistry<br />

of epileptic tissue, increased MMP-9 localized<br />

specifically to affected neurons.<br />

Conclusions: Taken together, the aforementioned<br />

results suggest that the synaptic pool of MMP-9 is<br />

critically involved in the sequence of events that underlie<br />

epileptogenesis, both in rodents and humans.<br />

Therefore, the enzyme should be considered as a<br />

candidate target for therapeutic pharmacological<br />

intervention.<br />

Acknowledgement: The work was supported by<br />

Polish-Norwegian Research Grant<br />

References:<br />

1. Wilczynski et al., Important role of matrix<br />

metalloproteinase 9 (MMP-9) in epileptogenesis. J Cell<br />

Biol 180: 1021-1035, 2008.<br />

2. Konopacki et al., Synaptic localization of seizureinduced<br />

matrix metalloproteinase-9 mRNA.<br />

Neuroscience; 150:31-39, 2007<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

day2<br />

Parallel Session 5: NEUROSCIENCE II


68<br />

WarSaW, poland May 26 – 29, 2010<br />

Potentials of new contrast agents for vascular molecular imaging in patients<br />

Douek P., Boussel L., Sigovan M., Cannet E. .<br />

Creatis, Hopital Louis Pradel, Lyon University, France<br />

philippe.douek@creatis.univ-lyon1.fr<br />

Introduction: Atherosclerosis is a diffuse and<br />

multisystem, chronic inflammatory disorder<br />

involving vascular, metabolic, and immune systems<br />

leading to plaque instability and / or vascular<br />

trombosis. The traditional risk assessment relies on<br />

clinical, biological and conventional imaging tools.<br />

However, they fall short in predicting near future<br />

events particularly in patients with unstable carotid<br />

artery disease or coronary artery disease. The plaque<br />

instability is dictated in part by plaque morphology,<br />

which in turn is influenced by pathophysiologic<br />

mechanisms at the cellular and molecular level.<br />

In current clinical practice, anatomic imaging<br />

modalities such as intravascular ultrasound, highresolution<br />

magnetic resonance imaging can identify<br />

several morphologic features supporting the unstable<br />

plaque, but give little or no information regarding<br />

molecular and cellular mechanisms. optical imaging,<br />

PET, Molecular MR imaging, or more recently<br />

spectral CT may identify some of these molecular<br />

and cellular processes.<br />

Methods: We aimed to review<br />

1) the role of relevant biological, factors and<br />

advances in atherosclerosis biology, that illuminate<br />

key biological aspects of atherosclerosis, including<br />

macrophage activity, protease activity, lipoprotein<br />

presence, apoptosis, and angiogenesis<br />

imaging life<br />

2) imaging agent chemistry (nanotechnology,<br />

chemical biology screens)<br />

3) recent advances in the field of molecular<br />

imaging that have led to the development of novel<br />

paramagnetic and superparamagnetic targeted<br />

contrast agents that bind exclusively to cells such as<br />

macrophages, or molecules such as albumin, fibrin,<br />

angiogenesis markers... with either PET, MR, US or<br />

CT .and<br />

Results: 4) a few clinically promising applications of<br />

molecular imaging for high-risk atherosclerosis and<br />

its potential for current and emerging treatments in<br />

clinical trials.<br />

Conclusions: A multimodal assessment of plaque<br />

instability involving the combination of systemic<br />

markers, high resolution imaging and molecular<br />

imaging targeting inflammatory and thrombotic<br />

components and the potential of new drugs<br />

targeting plaque stabilization may lead to a new<br />

stratification of the atherothrombotic risk and to<br />

a better prevention of atherothrombotic stroke or<br />

myocardial infarction.


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

Protease specific nanosensors in atherosclerosis<br />

Schellenberger E. .<br />

Charité Berlin, Germany<br />

eyk.schellenberger@charite.de<br />

Introduction: Atherosclerosis and especially the<br />

consequences of rupture of atherosclerotic plaques<br />

are the foremost cause of death in industrialized<br />

societies. Up to date the identification of vulnerable<br />

atherosclerotic plaques prone to rupture has not been<br />

solved for the clinics. The pathological processes<br />

leading to destabilization of such plaques are highly<br />

complex and involve inflammatory action and<br />

pathological tissue remodeling of the extracellular<br />

matrix performed by proteases. Therefor the imaging<br />

of specific protease activity by high resolution<br />

MR imaging could be an important tool to find<br />

dangerous plaques that need urgent treatment.<br />

Results: Optical imaging with activatable fluorescent<br />

smart probes has become the modality of choice<br />

for experimental in vivo detection of protease<br />

activity. Recently we introduced a novel highrelaxivity<br />

nanosensors that are suitable for in vivo<br />

imaging of protease activity by MRI. Upon specific<br />

protease cleavage, the nanoparticles rapidly switch<br />

from a stable low-relaxivity stealth state to become<br />

adhesive, aggregating high-relaxivity particles.<br />

To demonstrate the principle, we chose a cleavage motif<br />

of matrix metalloproteinase 9/2 (MMP-9 and MMP-2),<br />

proteases that important during the destabilization of<br />

the extracellular matrix of plaques. Based on clinically<br />

tested very small iron oxide particles (VSOP), the<br />

MMP-9-activatable protease-specific iron oxide<br />

particles (PSOP) have a hydrodynamic diameter of<br />

only 25 nm. PSOP are rapidly activated, resulting in<br />

aggregation and increased T2*-relaxivity of the particles.<br />

Conclusions: PSOP could be useful sensors for the<br />

in vivo identification of vulnerable plaques in the<br />

future.<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

day2<br />

Parallel Session 6: CARDIOVASCULAR I - together with ESR


YIA applicant<br />

70<br />

WarSaW, poland May 26 – 29, 2010<br />

Imaging of inflamed carotid artery atherosclerotic plaques with the use of 99mTc-HYNIC-<br />

IL-2 scintigraphy in the end-stage renal disease patients<br />

Opalinska M. (1) , Hubalewska-Dydejczyk A. (2) , Stompor T. (3) , Krzanowski M. (4) , Mikołajczak R. (5) , Garnuszek P. (5) ,<br />

Karczmarczyk U. (6) , Maurin M. (5) , Rakowski T. (7) , Sowa-Staszczak A. (2) , Glowa B. (2) .<br />

(1) Nuclear Medicine Unit, Chair and Department of Endocrinology, Jagiellonian University Medical School, (2) Nuclear Medicine Unit, Chair and Department of<br />

Endocrinology, Jagiellonian University Medical School, (3) Chair and Department of Nephrology, Hypertesiology and Internal Medicine, University of Warmia<br />

and Mazury, (4) Chair and Department of Nephrology, Jagiellonian University Medical School, (5) Radioisotope Center POLATOM, (6) Departament of Radiopharmaceuticals,<br />

National Medicines Institute, (7) 2nd Cardiology Clinic, Institute of Cardiology, Jagiellonian University Medical School.<br />

mkal@vp.pl<br />

Introduction: Cardiovascular diseases are the main<br />

cause of death in developed countries. In some patients’<br />

populations, including those with end-stage<br />

renal disease (ESRD), cardiovascular related mortality<br />

is 30 times higher compared to the general<br />

population.<br />

Such a high cardiovascular mortality is associated<br />

with significantly accelerated atherosclerosis in<br />

ESRD mainly due to intense inflammatory state.<br />

Therefore serum concentrations of inflammatory<br />

agents that promote atherosclerosis are often used<br />

to evaluate the cardiovascular risk.<br />

The histological studies of the atherosclerotic<br />

plaque revealed that at least 20% of inflammatory<br />

cells within unstable plaque comprise lymphocytes<br />

T, which contain receptors for interleukin 2 (IL-2);<br />

those receptors can be identified by scintigraphy<br />

with radiolabeled IL-2.<br />

Methods: 28 patients (13 men, 15 women, aged 55,2<br />

± 9,6, 17 on peritoneal dialysis, 11 on hemodialysis)<br />

underwent scintigraphy with the use of 99mTc-<br />

HYNIC-IL-2. In all cases ultrasound examination<br />

of the carotid arteries was performed to obtain information<br />

about localization and morphology of<br />

atherosclerosis plaques and intima-media thickness<br />

(IMT) measurement. Furthermore, levels of selected<br />

proinflammatory factors, atherogenic markers and<br />

calcium-phosphate balance parameters were measured.<br />

Finally, target/non-target (T/nT) ratio of IL-2<br />

uptake in atherosclerotic plaques confirmed by carotid<br />

USG with IMT, presence of calcifications in<br />

atherosclerotic plaques and concentration of the<br />

measured agents were compared.<br />

Results: Increased 99mTc-IL-2 uptake in atherosclerotic<br />

plaques previously visualized by neck ultrasound<br />

in 38/41 (91%) cases was detected. Median<br />

T/nT ratio of focal 99mTcIL-2 uptake in atherosclerotic<br />

plaque was 2,35 (range 1,23– 3,63, average 2,35<br />

± 0,70). Mean IMT value on the side of plaque assessed<br />

in scintigraphy was 0,79 ± 0,18 mm (median<br />

0,8, range 0,5 – 1,275).<br />

imaging life<br />

No statistically significant association was found<br />

between 99mTc-IL-2 T/nT ratio and mean value<br />

of either IMT or classical cardiovascular risk factors.<br />

Inversely, proportional dependence between<br />

scintigraphy results and hemoglobin concentration<br />

(R = - 0,21, p = 0,02) was found. Furthermore, relationships<br />

between T/nT ratio and homocysteine (R<br />

= 0,22, p = 0,037), ApoB (R = 0,31, p = 0,008), ApoB/<br />

ApoA-I ratio (R= 0,29, p= 0,012) and triglycerides<br />

concentration (R = 0,26, p = 0,021) were detected.<br />

Lower T/nT ratio in patients with better parameters<br />

of nutrition state (hemoglobin, albumin, adiponectin)<br />

in comparison with patients with worse nutritional<br />

parameters (3,20 ± 0,5 vs 2,16 ± 0,68, p =<br />

0,025) was revealed as well as the difference between<br />

values of T/nT ratio in groups of patients with values<br />

of ApoB, sCD40L and ADMA above and below median<br />

(3,18 ± 0,52 vs 2,16 ± 0,68 p = 0,031). An interesting<br />

relationship between T/nT ratio in atherosclerotic<br />

plaques with and without calcifications (2,35 ±<br />

0,68 vs 1,924 ± 0,55 p = 0,088) was observed.<br />

Conclusions: Scintigraphy with the use of labeled<br />

IL-2 can be a tool for inflamed atherosclerotic (vulnerable)<br />

plaque visualization within common carotid<br />

arteries in ESRD patients. Quantitative results<br />

of the carotid arteries scintigraphy with labeled IL-2<br />

correlate with serum concentration of selected cardiovascular<br />

risk markers<br />

Acknowledgement: This work is supported by the<br />

Polish Committee for Scientific Research (KBN)<br />

within Research Project 2 P05B 003 28<br />

References:<br />

1. Annovazzi A et al; 99mTc-interleukin-2 scintgraphy for<br />

the in vivo imaging of vulnerable atheroslerotic plaque.<br />

Eur J Nucl Med Mol Imaging 2006; 33: 117 – 126.


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

Uptake of 68 Ga-Chloride in Atherosclerotic Plaques of LDLR -/- ApoB 100/100 Mice<br />

Silvola J. (1) , Laitinen I. (2) , Sipilä H. (1) , Laine J. (3) , Leppänen P. (4) , Ylä-Herttuala S. (4) , Knuuti J. (1) , Roivainen A. (5) .<br />

(1) Turku PET Centre, University of Turku,<br />

(2) Nuklearmedizinische Klinik der TU Muenchen, Technische Universitaet Muenchen,<br />

(3) Department of Pathology, Turku University Hospital,<br />

(4) A.I. Virtanen Institute, University of Kuopio,<br />

(5) Turku Centre for Disease Modelling, University of Turku.<br />

jmuhau@utu.fi<br />

Introduction: Atherosclerosis is an inflammatory<br />

disease in which monocytes/macrophages have essential<br />

role in the development of plaques. Radiogallium<br />

has been used for decades for in vivo imaging<br />

of inflammation. 68Ga is a positron emitter with a<br />

half-life of 68 min and it is suitable for PET imaging.<br />

Purpose of this study was to explore the uptake of<br />

68Ga-chloride in atherosclerotic plaques in mice.<br />

Methods: Uptake of intravenously administered<br />

68Ga-chloride (17 ± 2 MBq) was investigated in<br />

9 atherosclerotic LDL-/-ApoB100/100 mice and<br />

6 control mice at 3 hours after injection. LDL-/-<br />

ApoB100/100 mice were kept in on a high fat, Western-type<br />

diet for 3-4 months, starting at 7 months<br />

of age. Control mice were fed with regular chow.<br />

The biodistribution of the tracer was evaluated, and<br />

aortic cryosections were further analysed by digital<br />

autoradiography. Subsequently, the autoradiographs<br />

were combined with histological and immunohistological<br />

analysis of sections [1].<br />

Results: According to the autoradiography analysis,<br />

the radioactivity uptake in atherosclerotic plaques was<br />

higher compared to healthy vessel wall (ratio 1.8 ± 0.2,<br />

P = 0.0002) and adventitia (ratio 1.3 ± 0.2, P = 0.0011).<br />

Some 68Ga-radioactivity was also detected in calcified<br />

regions of plaques. Autoradiography signal<br />

co-localized with macrophages as demonstrated by<br />

Mac-3 immunohistochemistry. In both mice strains,<br />

the highest level of radioactivity was found in the<br />

urine and blood.<br />

Conclusions: We observed a moderate but significantly<br />

higher 68Ga-chloride uptake in the aortic<br />

plaques of atherosclerotic mice. While the uptake of<br />

68Ga-chloride was promising in this animal model,<br />

the slow blood clearance may limit the usability of<br />

68Ga-chloride in clinical imaging of atherosclerotic<br />

plaques.<br />

Acknowledgement: The study was conducted within<br />

the Finnish Centre of Excellence in Molecular Imaging<br />

in Cardiovascular and Metabolic Research, supported<br />

by the Academy of Finland, University of Turku, Turku<br />

University Hospital and Åbo Akademi University.<br />

This work was further funded by the Finnish Foundation<br />

for Cardiovascular Research, Instrumentarium<br />

Foundation and the Drug Discovery Graduate School.<br />

References:<br />

1. Haukkala J et al; Eur J Nucl Med Mol Imaging. 36:2058–<br />

2067 (2009)<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

day2<br />

Parallel Session 6: CARDIOVASCULAR I - together with ESR


YIA applicant<br />

72<br />

WarSaW, poland May 26 – 29, 2010<br />

Hybrid imaging using dual energy µCT and FMT for characterization of atherosclerotic<br />

plaques in ApoE -/- mice<br />

Gremse F. (1) , Rix A. (2) , Bzyl J. (1) , Lederle W. (1) , Schulz R. (2) , Perkuhn M. (3) Schober A. (3) , Weber C. (3) , Kiessling F. (1) .<br />

(1) Department of Experimental Molecular Imaging, Medical Faculty, RWTH Aachen University,<br />

(2) Chair for Biological Imaging, Technical University Munich, Germany,<br />

(3) Department of Diagnostic Radiology, Medical Faculty, RWTH Aachen University,<br />

(4) Institute of Molecular Cardiovascular Research RWTH Aachen University, Germany.<br />

fgremse@ukaachen.de<br />

Introduction: To evaluate the ability of combined<br />

µCT and fluorescence mediated tomography (FMT)<br />

to characterize early and late stages in atherosclerotic<br />

plaque formation.<br />

Methods: Three groups of ApoE -/- mice were<br />

scanned by dual energy µCT and FMT. Iodine<br />

based blood pool CT contrast agent and a fluorescent<br />

probe for active cathepsins were used. CT and<br />

FMT datasets were fused using automated marker<br />

detection and registration. Group 1 consisted of 5<br />

mice with an age of more than 40 weeks fed with<br />

normal chow. Group 2 included 6 mice (age < 20<br />

weeks) with 12 weeks of cholesterol diet and group 3<br />

consisted of 5 mice (age


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

Long circulating emulsion based ct contrast agents<br />

De Vries A. (1) , Custers E. (2) , Lub J. (2) , Nicolay K. (1) , Gruell H. (1) .<br />

(1) Eindhoven University of Technology, The Netherlands<br />

(2) Philips Research, Eindhoven, The Netherlands<br />

A.d.Vries@tue.nl<br />

Introduction: With X-ray and CT moving forward<br />

into the interventional care, such as stent placement,<br />

balloon dilatation, vascular surgery, and electrophysiological<br />

procedures, there is a need for contrast<br />

agents that have long circulation times [1]. Previously,<br />

we prepared iodinated emulsion-based CT contrast<br />

agents that remain in the blood pool for several hours<br />

[2]. The primary blood clearance occurs via uptake in<br />

the reticuloendothelial system (RES), which is known<br />

to be dose dependant. Here, we tested if circulation<br />

times of these iodinated emulsions can possibly be extended<br />

by a co-injection with liposomes to slow down<br />

RES uptake of the contrast agents. We co-injected the<br />

iodinated emulsion (radiolabeled with 111 Indium) together<br />

with liposomes (radiolabeled with 177 Lutetium)<br />

and investigated the biodistribution of iodinated nanoparticles<br />

and liposomes by in vivo CT and γ-counting.<br />

Methods: Liposomes were prepared using DPPC,<br />

cholesterol and DOTA-DSPE (mol ratio 65.7/33.3/1)<br />

and were radiolabeled with 177 Lutetium. Iodinated<br />

nanoparticles were prepared using 2% w/w PBD-<br />

PEO (with 1 mol% PBD-PEO-DOTA) and 20% w/v<br />

3,7-dimethyloctyl 2,3,5-triiodobenzoate and were<br />

radiolabeled with 111 Indium. Swiss mice were used<br />

in the study, in which 2 mL/kg body weight of iodinated<br />

contrast agent (113 mgI/mL) together with<br />

2 mL/kg body weight of a) physiological salt (n=5)<br />

(control) or b) liposomes (n=5) was injected, leading<br />

to an injected dose of 225 mg I/kg body weight.<br />

Helical CT scans were acquired pre-injection (reference)<br />

and up to 3 hours post-injection using a<br />

dedicated small animal SPECT/CT system (nano-<br />

SPECT/CT ® , Bioscan). The uptake of 177 Lutetium<br />

and 111 Indium in dissected organs was measured<br />

using a γ-counting wizard.<br />

Results: Fig. 1 shows the Hounsfield Unit (HU)<br />

increase of blood after the injection of emulsion<br />

and emulsion together with liposomes as a function<br />

of time. The blood half life increased by 40<br />

minutes to t ½ =116 min when liposomes were coinjected<br />

(t ½ =77 min for pure emulsion; blood half<br />

time was derived from a single-exponential fit).<br />

The CT and biodistribution data (γ-counting)<br />

showed that when co-injecting liposomes, the<br />

emulsion particles are taken up in a lower degree<br />

by the spleen as compared to the control group.<br />

Conclusions: Circulation times of the iodinated<br />

emulsions as CT contrast agents can be further<br />

extended when co-injecting liposomes. The liposomes<br />

are likely taken up by the RES system,<br />

thereby slowing down the clearance of the iodinated<br />

emulsion in a dose dependant manner. As<br />

liposomes are well tolerated with little toxicity at<br />

high doses, the above strategy can be exploited to<br />

formulate well tolerated long circulating iodinated<br />

contrast agents without increasing the administered<br />

iodine dose.<br />

Delta Hounsfield Units<br />

120<br />

100<br />

80<br />

60<br />

40<br />

20<br />

References:<br />

Emulsion<br />

Emulsion + Liposomes<br />

0<br />

0 25 50 75 100 125 150 175 200<br />

Time (min)<br />

1. Vera DR, Mattrey RF; Acad Rad. 9(7):784-792 (2002)<br />

2. de Vries, A. et al. Biomaterials, submitted (2010)<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

YIA applicant<br />

day2<br />

Parallel Session 6: CARDIOVASCULAR I - together with ESR


74<br />

WarSaW, poland May 26 – 29, 2010<br />

Application of microfluidics to the ultra-rapid preparation of fluorine-18 and carbon-11<br />

labelled compounds<br />

Miller P. .<br />

Imperial College London, UK<br />

philip.miller@imperial.ac.uk<br />

Introduction: The advantages associated with the<br />

miniaturisation of chemical reactions have been<br />

recognised for well over a decade now. Such benefits<br />

include controlled and predicable mixing regimes,<br />

efficient heat transfer, reduced reagent consumption,<br />

small reaction volumes (nL-µL), improved safety<br />

and enhanced processing capabilities. Microfluidic<br />

reactors, the devices used to perform and process<br />

small scale chemical reactions, contain enclosed<br />

microchannels typically 10-500 µm in diameter and<br />

can be fabricated from a wide range of materials including<br />

various polymers, silicon and glass (figure<br />

1). The use of microfluidic reactors for rapid synthesis<br />

with the short-lived isotopes 11C (t1/2 = 20.4<br />

min) and 18F (t1/2 = 109 min), commonly used in<br />

positron emission tomography, has been recognised<br />

since 2003. Interest in the application of microfluidics<br />

to PET radiosynthesis has come about primarily<br />

because miniaturised reaction systems have the<br />

potential to address several key challenges in PET<br />

radiochemistry, including increasing the speed,<br />

imaging life<br />

reducing the scale and improving the processing of<br />

radiolabelling procedures.[1,2] This talk will provide<br />

a succinct and critical overview, to the nonspecialist,<br />

of the emerging and developing field of<br />

using microfluidics for the rapid preparation of 11C<br />

and 18F labelling compounds.<br />

Acknowledgement: PWM is grateful to the EPSRC<br />

for the award of a Life Sciences Interface fellowship<br />

(EP/E039278/1).<br />

References:<br />

Figure 1<br />

1. P. W. Miller, J. Chem. Technol. Biotechnol. 2009, 84,<br />

309. [2] P. W. Miller, A. J. deMello and A. D. Gee, Curr.<br />

Radiopharmaceuticals.accepted for publication.


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

Radioligand for in vivo measuring neurotransmitters release<br />

Halldin C. (1) , Finnema S. (1) , Varrone A. (1) , Farde L. (1) .<br />

Karolinska Institutet, Sweden<br />

christer.halldin@ki.se<br />

Introduction: This report deals with an overview<br />

of radioligands useful for in vivo measuring<br />

neurotransmitter release. In particular this will<br />

be exemplified by a PET-study evaluating the<br />

effect of (±)-fenfluramine- induced serotonin<br />

release on the binding of the selective serotonin<br />

5-HT1B receptor radioligand [11C]AZ10419369 in<br />

cynomolgus monkeys. In a classical displacement<br />

paradigm after bolus administration of radioligand<br />

fenfluramine caused a dose-dependent reduction in<br />

specific binding ratios. The aim of this study was to<br />

confirm our previous findings by using a bolus plus<br />

continuous infusion approach in monkey.<br />

Materials and Methods: A total of 18 PET<br />

measurements were conducted using a bolus plus<br />

infusion paradigm of [11C]AZ10419369 in three<br />

cynomolgus monkeys. On six of the nine experimental<br />

days a baseline measurement was followed by a<br />

displacement measurement in which fenfluramine<br />

(1.0 or 5.0 mg/kg) was infused i.v. between 80 and<br />

85 minutes after the bolus radioligand injection.<br />

On three of the nine experimental days a baseline<br />

measurement was followed by a pretreatment<br />

measurement in which fenfluramine (5.0 mg/kg)<br />

was infused i.v. between 30 and 25 minutes before<br />

the bolus radioligand injection. Emission data were<br />

acquired for 125 or 155 minutes using the HRRT<br />

PET-system. For determination of the fenfluramine<br />

effect the binding potential (BPND) was calculated<br />

for different time frames with the cerebellum as<br />

reference region.<br />

Results: Administration of fenfluramine had no<br />

evident effect on radioactivity in the cerebellum.<br />

After administration of fenfluramine (1.0 and 5.0<br />

mg/kg), the respective binding potentials decreased<br />

in the occipital cortex by 19% (7-34%) and 47% (40-<br />

57%) in the displacement paradigm, and by 39%<br />

(32-45%) in the pretreatment paradigm (5.0 mg/kg).<br />

Conclusions: This study confirms that the 5-HT1Bligand<br />

[11C]AZ10419369 is sensitive to endogenous<br />

serotonin levels in vivo.<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

day2<br />

Parallel Session 7: PROBES II - together with EANM


76<br />

WarSaW, poland May 26 – 29, 2010<br />

18f-tracers for amyloid plaques<br />

Guilloteau D. .<br />

CHRU Tours, INSERM Imaging and Brain, France<br />

denis.guilloteau@univ-tours.fr<br />

As documented by the increasing literature, PET<br />

imaging using probes binding to amyloid plaques<br />

represents a major opportunity to investigate<br />

early symptoms and prodromal phase of AD, and<br />

in other hand to follow up the efficiency of new<br />

treatment.<br />

The most well-known tracer used for this molecular<br />

imaging is the [11C]-PIB. Numerous studies with<br />

PIB have demonstrated that the properties of this<br />

tracer permit the visualization and the quantification<br />

of amyloid deposits, but its [11C] labelling limits<br />

considerably its potential use due to its short half-life.<br />

A strong effort have been performed by different<br />

groups in order to develop a tracer labelled with [18F]<br />

for amyloid plaques. Several 18F amyloid binding<br />

compounds belonging to different families have<br />

been described. Of these 18F-AV-45 (Florbetapir)<br />

is most advanced in clinical development (end of<br />

imaging life<br />

Phase III) but Phase I / II clinical results have also<br />

been reported on 18F-FDDNP, 18F-Flumetametamol,<br />

18F-BAY94-9172 (AV-1). We will present these<br />

results and discuss the similarities and differences<br />

regarding the specificity and the sensitivity of the<br />

brain uptake.<br />

Some results obtained with [18F]-FDDNP and<br />

([18F]-AV45 in our University Hospital will be<br />

presented.


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

Chelators for radiocopper: biological/pharmacokinetic differences of [Tyr 3 ,Thr 8 ]octreotide<br />

conjugates<br />

Abiraj K. (1) , Fani M. (2) , Tamma M. L. (1) , Reubi J. C. (3) , Barnard P. (4) , Schibli R. (5) , Dilworth J. (6) , Maecke H. (2) .<br />

(1) University Hospital of Basel, Switzerland<br />

(2) University Hospital of Freiburg, Germany<br />

(3) University of Bern, Switzerland<br />

(4) University of Melbourne,<br />

(5) ETH Zurich,<br />

(6) University of Oxford.<br />

keelaraa@uhbs.ch<br />

Introduction: Copper-64 has attracted great deal<br />

of attention as radioisotope for targeted positron<br />

emission tomography (PET) and radionuclide therapy<br />

due to its dual decay characteristics (β+: 17.4%;<br />

E β+max= 653 keV; β-: 39%; E β-max= 579 keV),<br />

favorable half life (t1/2=12.7 h) and increased availability.1<br />

Owing to the instability of copper complexes<br />

under ‘in vivo’ condition, continuous efforts<br />

are being made to develop bifunctional chelators<br />

(BFC’s) which can form stable copper complexes for<br />

labeling of biomolecules.1-3 In the present study,<br />

we conjugated [Tyr3,Thr8]octreotide (TATE), to<br />

four different chelating systems, radiolabeled with<br />

64Cu and evaluated the biological/pharmacokinetic<br />

differences of these radioconjugates.<br />

Methods: The BFC’s DOTA(tBu)3 and<br />

NODAGA(tBu)3 are commercially available whereas<br />

CB-TE2A(tBu) and PHENDAM were synthesized<br />

using suitable synthetic methodologies. TATE was<br />

synthesized on solid phase using standard Fmoc<br />

strategy and coupled to each BFC. The conjugates<br />

were labeled with 64Cu using 0.1 M ammonium<br />

acetate buffer (pH 8.0) under different labeling<br />

conditions. Receptor affinity measurements were<br />

performed using radioligand assays and autoradiographic<br />

methods. The radiolabeled conjugates were<br />

evaluated in vitro and in vivo in tumor-bearing nude<br />

mice, using the HEK-sst2 cell line. Imaging studies<br />

were performed using a clinical PET/CT camera.<br />

Results: Synthesis of orthogonally protected crossbridged<br />

cyclam-based BFC (CB-TE2A(tBu)) allows<br />

facile conjugation to targeting biomolecules such<br />

as peptide on solid phase. Synthesis of PHENDAM<br />

provides a new N4-macrocyclic ligand for conjugation<br />

to biomolecule and subsequent labeling with<br />

64Cu. All the conjugates (DOTA-TATE, NODAGA-<br />

TATE, PHENDAM-TATE and CB-TE2A-TATE)<br />

showed high binding affinity to somatostatin receptor<br />

subtype-2 (sst2) with IC50 values ranging from<br />

0.6 nM to 2.5 nM. The labeling of NODAGA-TATE<br />

could be performed at room temperature (radiolabeling<br />

yield >97% at specific activity of >20 GBq/<br />

µmol) whereas other conjugates required elevated<br />

temperature. All the radioconjugates showed substantially<br />

high and receptor mediated uptake by<br />

HEK-sst2 cells. Biodistribution studies with nude<br />

mice showed high uptake of the radioconjugates in<br />

HEK-sst2 xenografts at 1h p.i. (64Cu-DOTA-TATE:<br />

20.29±2.74%, 64Cu-NODAGA-TATE: 29.39±4.13%,<br />

64Cu-PHENDAM-TATE: 16.97±1.46%, and 64Cu-<br />

CB-TE2A-TATE: 19.34±2.55% ID/g). The radioactivity<br />

in tumor persisted at 4h p.i., but 24h p.i. data<br />

showed fast washout in case of all the conjugates.<br />

Compared to 64Cu-DOTA-TATE, the other three<br />

radioconjugates showed improved pharmacokinetics.<br />

High in vivo stability of radiometal complex in<br />

case of 64Cu-NODAGA-TATE and 64Cu-CB-TE2A-<br />

TATE was apparent from their low uptake in liver<br />

and blood at all time points. Interestingly, 64Cu-<br />

PHENDAM-TATE showed fast washout from the<br />

kidneys resulting in high tumor-to-kidney ratio at<br />

early time points. The PET/CT images acquired at<br />

8 h p.i. clearly delineated the tumor and showed images<br />

in accordance with the biodistribution data.<br />

Conclusions: Among four different chelating systems<br />

evaluated, 64Cu-NODAGA-TATE and 64Cu-<br />

CB-TE2A-TATE showed superior in vivo stability<br />

of the radiocopper complex. The study infers that<br />

triazacyclononane and cross-bridged cyclam based<br />

chelating systems might be ideal for 64Cu labeling<br />

of biomolecules intended to be used in targeted PET<br />

imaging and radionuclide therapy but PHENDAM<br />

may be a new 64Cu chelating system of interest for<br />

protein labeling.<br />

Acknowledgement: Swiss National Science Foundation,<br />

European Molecular Imaging Laboratories<br />

(EMIL), COST BM0607.<br />

References:<br />

1. Shokeen W et al; J. Acc. Chem. Res. 42: 832-841 (2009)<br />

2. Boswell C A et al; J. Med. Chem. 47: 1465-1474 (2004)<br />

3. Prasanphanich A F et al; PNAS 104: 12462-12467<br />

(2007)<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

YIA applicant<br />

day2<br />

Parallel Session 7: PROBES II - together with EANM


78<br />

WarSaW, poland May 26 – 29, 2010<br />

[ 11 C]SOMADAM: potential sert ligand for pet studies and comparison with [ 11 C]MADAM<br />

Gourand F. (1) , Emond P. (2) , Bergström J. P. (3) , Takano A. (3) , Gulyas B. (3) , Guilloteau D. (2) , Barré L. (1) , Halldin C. (3) .<br />

(1) CEA/DSV CI-NAPS, France<br />

(2) INSERM U930-Université François Rabelais de Tours CHRU Bretonneau, France<br />

(3) Karolinska Institutet, Sweden<br />

gourand@cyceron.fr<br />

Introduction: Although [ 11 C]MADAM is an<br />

appropriate PET radioligand for the visualization<br />

and quantification of SERT in vivo, metabolite<br />

analysis in human and non-human plasma samples<br />

using HPLC separation have shown that [ 11 C]<br />

MADAM was rapidly metabolized. 1,2<br />

A possible metabolic pathway is the S-oxidation<br />

which could lead to SOMADAM and SO 2 MADAM.<br />

In vitro evaluation of these two potential metabolites<br />

have shown that SOMADAM exhibited a good<br />

affinity for SERT and a good selectivity for SERT<br />

over NET and DAT. 3 In view of its in vitro biological<br />

properties, SOMADAM has been labelled with<br />

carbon-11 and evaluated as a potential radioligand<br />

for in vivo quantification of SERT in the monkey<br />

brain using PET.<br />

Methods: The labeling of [ 11 C]SOMADAM is based<br />

on the N-alkylation reaction of the N-desmethyl<br />

precursor using [ 11 C]methyl triflate in acetone.<br />

Comparative PET imaging studies in cynomolgus<br />

monkey with [ 11 C]MADAM and [ 11 C]SOMADAM<br />

were carried out and plasma samples were analyzed<br />

using reverse phase HPLC.<br />

Results: The incorporation of [ 11 C]methyl triflate to<br />

[ 11 C]SOMADAM was in the range of 50% and after<br />

purification by reverse phase HPLC, [ 11 C]SOMADAM<br />

was obtained with a radiochemical purity higher<br />

than 99%. PET imaging studies in monkey using<br />

[ 11 C]SOMADAM showed 1) a rapid and high brain<br />

uptake and 2) an homogenous distribution of the<br />

imaging life<br />

radioactivity. A rapid washout in all regions was<br />

observed, indicating a high non-specific binding. As<br />

it has already been shown in previous PET studies,<br />

the radioactivity accumulation of [ 11 C]MADAM is<br />

consistent with the known densities of SERT sites.<br />

HPLC analysis of plasma samples obtained after<br />

either [ 11 C]MADAM or [ 11 C]SOMADAM injection<br />

gave radiochromatograms with a similar profile<br />

where all the radioactive metabolites detected were<br />

more hydrophilic than the parent compound. After<br />

[ 11 C]MADAM injection, one radioactive species has<br />

been identified as [ 11 C]SOMADAM and the fraction<br />

was approximatively 5% at 4 min and 1% at 15 min.<br />

Conclusions: From PET imaging studies in<br />

monkey, it appears that [ 11 C]SOMADAM does not<br />

present any advantages over [ 11 C]MADAM, either<br />

in terms of brain kinetics and specific binding.<br />

Nevertheless, [ 11 C]SOMADAM has been identified<br />

in plasma sample as a minor labeled metabolite of<br />

[ 11 C]MADAM.<br />

References:<br />

1. Halldin C et al; Synapse. 58:173-183 (2005)<br />

2. Lundberg J et al; J Nucl Med. 46:1505-1515 (2005)<br />

3. Vercouillie J et al; Bioorg Med Chem Lett. 16:1297-<br />

1300 (2006)


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

MR imaging of extracellular redox by a thiolsensitive Gd(III)-DO3A derivative<br />

Menchise V. (1) , Gianolio E. (2) , Digilio G. (3) , Cittadino E. (2) , Napolitano R. (2) , Fedeli F. (2) , Catanzaro V. (2) , Aime S. (2) .<br />

(1) Institute for Biostructures and Bioimages (CNR) c/o Molecular Biotechnology Center University of Turin, Italy<br />

(2) Department of Chemisty IFM & Center for Molecular Imaging, University of Turin, Italy<br />

(3) Department of Environmental and Life Sciences, University of Eastern Piedmont “A. Avogadro”, Alessandria, Italy<br />

valeria.menchise@unito.it<br />

Introduction: The characterization of the microenvironment<br />

around and within tumors is of great<br />

importance to evaluate the invasiveness of cancers<br />

and to predict resistance to radio- and chemotherapy.<br />

Severe hypoxia and acidosis has been related<br />

to aggressive cell phenotype, poor clinical outcome<br />

and low likelihood of patient survival. 1 More reducing<br />

microenvironment conditions are related<br />

to increased cancer cell proliferation and increased<br />

resistance to chemotherapies. Any method for the<br />

imaging of the parameters characterizing the tumor<br />

microenvironment would be therefore of immense<br />

value for the characterization/grading of the tumor<br />

and for the choice and calibration of therapies. We<br />

have recently described a series of Gd-DO3A based<br />

compounds that exploit exofacial protein thiols<br />

(EPTs) to be taken up by cells. The most effective<br />

of these compounds, named Gd-L1A, can reach an<br />

intracellular concentration as high as 1.2x10 10 Gd<br />

atom per single cell. 2 The fact that the uptake of Gd-<br />

L1A by cells is proportional to the amount of EPTs<br />

makes it an interesting candidate as a probe for the<br />

MR molecular imaging of the extracellular redox<br />

microenvironment.<br />

Methods: In labeling experiments, cells grown to a<br />

confluence of 80% were subjected to a 4 hours incubation<br />

with Gd-L1A (0.5 to 3mM) at 37 °C, washed<br />

three times, mechanically harvested and finally subjected<br />

to mineralization and quantitative analysis of<br />

Gd(III) by a relaxometric assay. MR images were acquired<br />

on mice grafted with B16 tumor before and after<br />

intratumor/intramuscle injection of Gd-L1A using<br />

a T1-weighted multislice multiecho protocol (TR/TE<br />

250/7.7) on an ASPECT spectrometer operating at 1T.<br />

Results: Uptake experiments of GdL1A carried<br />

out on B16 melanoma cells treated with chemicals<br />

known to block EPTs have shown that the extent of<br />

Gd uptake increases with the concentration of free<br />

EPTs on the cell membrane. Imaging of EPTs has<br />

been performed in an animal tumor model obtained<br />

by inoculating about 1 million of B16 melanoma<br />

cells subcutaneously in B57Bl/6 mice. Gd-L1A has<br />

been delivered to tumor areas in these mice models<br />

and signal enhancement monitored over time and<br />

compared to that found in non-tumor bearing tissue.<br />

Gd-DO3A has been used as a control.<br />

A significant signal enhancement has been found in<br />

the tumor area of mice treated with Gd-L1A, with<br />

wash-out kinetics of the contrast agent consistent<br />

with its internalisation by tumor cells.<br />

Conclusions: These results show that Gd-L1A is<br />

suitable to image EPTs, that are in turn sensitive and<br />

responsive to the extracellular redox.<br />

Acknowledgement: This work is supported by FP6<br />

Project NMP4-CT-2006-026668 (MediTrans) and by<br />

Regione Piemonte (PIIMDMT)<br />

References:<br />

1. Tatum et al. Int J Radiat Biol., 82: 699-757 (2006)<br />

2. Digilio G. et al., Chem. Commun. 893–895 (2009)<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

day2<br />

Parallel Session 7: PROBES II - together with EANM


80<br />

WarSaW, poland May 26 – 29, 2010<br />

Everything but not cell replacement with somatic neural stem cell transplantation<br />

Pluchino S. .<br />

San Raffaele Scientific Institute Milano, Italy<br />

pluchino.stefano@hsr.it<br />

Introduction: Since the first transplant of stem cells<br />

into the spinal cord of rodents in which an acute demyelinating<br />

lesion was induced, we have witnessed<br />

a spur of experimental cell-based transplantation<br />

approaches aimed at fostering biological and molecular<br />

mechanisms underlying CNS repair. Theories<br />

assuming that very little renewing potential is<br />

identified within the adult CNS have been contravened,<br />

new promising sources of myelinogenic cells<br />

for transplantation purposes have been characterized,<br />

and new cell-replacement strategies have been<br />

proposed and established. A better understanding of<br />

the dynamics of endogenous remyelination has been<br />

achieved, and insights concerning the process of remyelination<br />

driven by site-specific myelin-forming<br />

cell transplantation have been discovered.<br />

Methods: Some major limitations have – however –<br />

not been overcome yet:<br />

(i) the limited amount of highly myelinating cells<br />

that can be grown in vitro and<br />

(ii) the limited migratory capacity of myelinating<br />

cells once transplanted. Somatic stem cells might<br />

represent therefore an alternative and promising area<br />

of investigation with some potential in its essence.<br />

Results: New hopes have been recently raised by the<br />

encouraging preliminary results obtained by transplanting<br />

CNS-derived NPCs and bone marrow mesenchymal/stromal<br />

stem cells in rodents with experimental<br />

MS. However, most of the results with stem<br />

cells as therapeutic weapons for MS have consistently<br />

challenged the sole and limited view that stem cells<br />

therapeutically work exclusively throughout cell replacement.<br />

Indeed, the transplantation of somatic<br />

(non-hematopoietic) stem cells promotes substantial<br />

CNS repair via a number of bystander mechanisms,<br />

mainly exerted by undifferentiated stem cells releasing<br />

in vivo a milieu of tissue-trophic and immune<br />

modulatory molecules, whose release is likely to be<br />

temporally and spatially orchestrated by specific<br />

(micro)environmental cues. These molecules are indeed<br />

are pleiotropic and redundant in nature as well<br />

as ‘constitutively’ secreted by stem cells. In this view,<br />

the therapeutic plasticity of stem cell can be viewed<br />

imaging life<br />

as the capacity of stem cells to adapt their fate and<br />

function(s) to specific environmental needs occurring<br />

as a result of different pathological conditions.<br />

Conclusions: While further studies are certainly required<br />

to assess the overall safety, efficacy and in vivo<br />

therapeutic plasticity of NPCs, the great challenge for<br />

any future human application of NPC-based protocols<br />

in MS will be to develop more reliable and reproducible<br />

approaches optimizing both (tissue) trophic<br />

as well as immune regulatory capacities of stem cells<br />

for functional and anatomical rescuing of myelin architecture<br />

in MS patients.<br />

Detailed review of the most recent data on the highly<br />

peculiar immune regulatory stem cell signature will<br />

be here provided.


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

Mesenchymal stem cell transplantation: an option to promote remyelination?<br />

Aigner L. .<br />

Institute of Molecular Regenerative Medicine, Paracelsus Medical University Salzburg, Austria<br />

ludwig.aigner@pmu.ac.at<br />

Mesenchymal stem cells (MSCs) have been<br />

used previously in a number of animal models<br />

for neurological / neurodegenerative diseases.<br />

Moreover, clinical studies that are using MSCs for<br />

therapy of CNS diseases are currently ongoing. In<br />

animal models, systemic or local transplantation<br />

of MSCs provided functional improvement. The<br />

therapeutic effects of MSCs are derived from their<br />

immuno-modulatory activity and from the fact<br />

that MSCs secrete a number of neuroprotective<br />

cytokines. Moreover, there is evidence that MSCs<br />

might be able to influence the pool of endogenous<br />

stem or progenitor cells. Along this line, we have<br />

recently demonstrated that MSCs secrete activities<br />

that promote oligodendroglial fate and differentiation<br />

of neural progenitors (Rivera et al., 2006).<br />

Here, we will present data on the effects of MSC<br />

transplantation on endogenous spinal cord<br />

progenitors after spinal cord injury. Like in the<br />

in vitro situation, the presence of MSCs in the<br />

lesioned spinal cord promoted oligodendroglial and<br />

inhibited astroglial differentiation of endogenous<br />

progenitors. This supports the possibility of use<br />

MSC transplantation for strategies to remyelinate<br />

axons in the diseased CNS.<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

day2<br />

Parallel Session 8: GENE and CELL based therapies - together with CliniGene


82<br />

WarSaW, poland May 26 – 29, 2010<br />

Viral vector-mediated transcriptional targeting of dendritic cells for antigen-specific<br />

tolerance induction in EAE/MS.<br />

Dresch C. .<br />

Univesity of Zürich, Switzerland<br />

cdresch@vetvir.uzh.ch<br />

Introduction: The cause of multiple sclerosis (MS)<br />

is unknown and the pathogenic processes leading<br />

to disease development is incompletely understood.<br />

Current knowledge supports a T cell<br />

mediated autoimmune pathogenesis targeting myelin<br />

components or myelin-producing cells. Immunization<br />

of susceptible animals with myelin<br />

antigens or transfer of myelin antigen-reactive T<br />

cells induces experimental autoimmune encephalomyelitis<br />

(EAE), an inflammatory disorder of<br />

the CNS which closely resembles MS. Because the<br />

ethiology of MS is not yet completely understood,<br />

there is no curative treatment available at present.<br />

Methods: The aim of this project was to induce permanent,<br />

antigen-specific tolerance in EAE/MS. The<br />

strategy includes the ex vivo modification of autologous<br />

hematopoietic stem cells (HSC) with lentiviral<br />

vectors that express antigens involved in EAE/MS<br />

from a dendritic cell-specific promoter. After reinfusion,<br />

the modified HSC will give rise to all cells<br />

of the immune system including antigen expressing<br />

dendritic cells. As lentivirus vectors mediate the genomic<br />

integration of transgenes in HSC, there is a<br />

constant supply of antigen expressing “steady-state”<br />

dendritic cells. We hypothesized that the stable<br />

antigen presentation by these cells in thymus and<br />

periphery in a non-inflammatory condition would<br />

tolerize self-reactive T cells and, therefore, prevent/<br />

revert disease development.<br />

Results: We demonstrated the effectiveness of this<br />

strategy for inducing myelin oligodendrocyte glycoprotein<br />

(MOG)-specific tolerance in an EAE<br />

model in mice. We show the efficient deletion of<br />

MOG specific T cells in chimeras that received HSC<br />

transduced with a MOG-expressing lentivirus vector.<br />

Also, 0% of mice which received HSC transduced<br />

with the MOG-expressing lentivirus vector<br />

developed EAE upon induction (clinical score 0),<br />

while 100% of mice that received BM cells transduced<br />

with a GFP-expressing control lentivirus vector<br />

developed EAE.<br />

Conclusions: We confirmed the potential of lentivirus<br />

vectors that transcriptionally target transgene<br />

imaging life<br />

expression to dendritic cells for antigen-specific<br />

tolerance induction in autoimmune diseases. This<br />

strategy completely prevented MOG induced EAE<br />

in mice. We will further investigate the mechanisms<br />

of vector mediated tolerance induction, in particular<br />

the involvement of regulatory T cells and cytokines.<br />

The strategy presented here is particularly promising<br />

for clinical applications. Moreover, these tools<br />

will also prove useful for studying disease mechanisms<br />

and for addressing fundamental questions in<br />

immunity and tolerance.<br />

Acknowledgement: Swiss-MS Society.


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

Delivery of a bioluminescent transgene to a tumor via bone marrow engraftment and local<br />

control of gene expression by non invasive local hyperthermia<br />

Fortin P. Y. (1) , Lepetit-Coiffe M. (1) , Genevois C. (1) , Debeissat C. (1) , Quesson B. (1) , Moonen C. (1) , Couillaud F. (1) .<br />

(1) University of Bordeaux/CNRS, France<br />

py.fortin@imf.u-bordeaux2.fr<br />

Introduction: The success of a gene therapy strategy<br />

depends on several parameters including the<br />

right choice of therapeutic gene, efficient delivery<br />

method and reliable control of gene expression. In<br />

the present study, we investigate an in vivo strategy<br />

to deliver a transgene around a tumor and to control<br />

the expression of the transgene. This strategy<br />

includes bone marrow engraftment of genetically<br />

engineered cells into a wild type mouse to create<br />

a chimera with nucleated circulating blood cells<br />

(CBC) expressing the firefly luciferase (lucF) reporter<br />

gene under the transcriptional control of a<br />

thermosensitive promoter. Later on, a subcutaneous<br />

tumor is induced and as part of the physiological<br />

inflammatory process, CBC accumulate into<br />

and around the tumor. Finally, lucF expression<br />

detected by bioluminescence imaging (BLI) is<br />

induced “on demand” by local hyperthermia using<br />

MR guided high-intensity focused ultrasound<br />

(MRgHIFU).<br />

Methods: Bone marrow cells (BMCs) were obtained<br />

from homozygote C57/BL6 (CD45.2) transgenic<br />

mice (NLF-1) containing the lucF transgene under<br />

transcriptional control of the heat shock protein 70<br />

(Hspa1b) promoter. BMCs were transplanted into<br />

a congenic mouse (CD45.1) pre-treated with Busilvex®,<br />

an injectable form of busulfan (2 busulfan IP<br />

injections, 25 mg/Kg) to induce medullar aplasia.<br />

The level of engraftment was measured 2 months<br />

later by measuring CD45.1/CD45.2 ratio using Fluorescence<br />

Activated Cell Sorting (FACS). Carcinoma<br />

Mouse Tumor 93 (CMT-93, ATCC) were implanted<br />

subcutaneously (2 millions cells) to generate a tumor<br />

on the left leg. Tumors were heated (44°C, 8<br />

min) for local gene activation using either a water<br />

bath or MRgHIFU. LucF expression was evaluated<br />

in vivo 6 hours post heating by BLI.<br />

Results: After 2 months, about 80% of engrafted<br />

mice exhibited more than 65% of CBC from the<br />

donor mouse as demonstrated by FACS. CMT-93<br />

cells implanted into engrafted mice formed tumors<br />

ranging from 5 and 10 mm in one month. Tumors<br />

heated (44°C, 8 min) by dipping the tumor-bearing<br />

leg into a water bath, in mice exhibiting more<br />

than 65% of engraftment, induced lucF activation<br />

and transient light emission 6 hours later. Light<br />

emission was found in and around the tumor, for<br />

about 50% of the mice. The remaining mice did<br />

not exhibit any light emission. A week later lightemitting<br />

mice did not produce light anymore.<br />

They were heated again (44°C, 8 min) but using<br />

the MRgHIFU device. Six hours later, light emission<br />

was found around the heated focal region corresponding<br />

to both tumors and the surrounding<br />

region (n = 15). Light emission was also detected<br />

in an additional location (n = 6) corresponding to<br />

head and apical region of bone both on ipsi and<br />

contralateral legs.<br />

Conclusions: The bioluminescent chimera mice<br />

express the lucF reporter under transcriptional<br />

control of a thermosensitive promoter in hematopoietic<br />

cells. This model allows for studying<br />

gene delivery to tumor using circulating blood cells.<br />

Local gene expression was induced “on demand”<br />

by local hyperthermia. MRgHIFU heating reveals<br />

both expected (inside and around the tumor) and<br />

unexpected (contralateral leg) activation patterns<br />

further to be explored.<br />

Acknowledgement: This work was supported in<br />

part by Diagnostic Molecular Imaging and the<br />

Conseil Régional d’Aquitaine.<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

day2<br />

Parallel Session 8: GENE and CELL based therapies - together with CliniGene


YIA applicant<br />

84<br />

WarSaW, poland May 26 – 29, 2010<br />

Dendritic cell labelling with paramagnetic nanoparticles and 111 In-oxine for in vivo magnetic<br />

resonance imaging and scintigraphic imaging<br />

Martelli C. (1) , Borelli M. (1) , Rainone V. (1) , Ottobrini L. (1) , Degrassi A. (2) , Russo M. (2) , Texido G. (3) , Pesenti E. (2) , Fiorini C. (4) ,<br />

Clerici M. (5) , Trabattoni D. (1) , Lucignani G. (1) .<br />

(1) University of Milan ,<br />

(2) Nerviano Medical Sciences,<br />

(3) Pharmacology Department, BU Oncology, Nerviano Medical Sciences, Nerviano, MI,<br />

(4) Politecnico of Milan,<br />

(5) University of Milan and on Gnocchi Foundation.<br />

cristina.martelli@unimi.it<br />

Introduction: Better understanding of the biology<br />

and the role of dendritic cells (DCs) in regulating<br />

immune responses is driving the development of innovative<br />

anti-neoplastic DC based immunotherapies<br />

both at clinical and pre-clinical levels [1] . The aim of<br />

this study is the development and the evaluation of a<br />

tumour-specific DC vaccine, tested on a transgenic<br />

murine model of breast cancer (MMTV-v-Ha-Ras).<br />

MRI and SPET methodologies were tested for their<br />

feasibility in showing the migration to local draining<br />

lymph nodes (DLNs) of DC, properly labelled<br />

with paramagnetic nanoparticles (MNPs, Endorem ® )<br />

or 111 In-oxine [2] .<br />

Methods: Total bone marrow cells were extracted<br />

from wt mice. DC differentiation was studied by<br />

flow cytometry; at the 6 th day of culture DCs were<br />

labelled with commercial MNPs (200 ugFe/ml, for<br />

16h). Labelling efficiency was checked by optical<br />

microscopy after Perl’s staining and relaxometric<br />

analysis. Tumour lysates from breast cancer lesions<br />

of transgenic mice were used to load immature DCs<br />

(iDCs), and maturation was monitored by flow cytometry.<br />

Stimulatory activity of Ag-loaded DCs and<br />

migratory ability were evaluated in vitro. MNP labelled<br />

and Ag loaded DCs were then injected into<br />

the footpad of a transgenic tumour bearing mice.<br />

The same cells were labelled after antigen loading<br />

with 111 In-Oxine (30 uCi/10 6 cells). MRI and SPET<br />

imaging were performed at 4, 24 and 48h after cells<br />

injection. MRI was performed on a 7T Bruker Pharmascan<br />

instrument, and MSME and FLASH sequences<br />

were used. SPET imaging was carried out<br />

with a new prototype of gamma camera developed<br />

within an European Project by Politecnico of Milano.<br />

Ex vivo Perl’s staining and gamma counting of<br />

explanted DLNs was performed to validate imaging<br />

data and DC migration.<br />

Results: Perl’s staining showed constant iron content<br />

of DCs in all the experiments. Mean Iron<br />

content was 240 pg/cell. DCs labelling with 111 Inoxine<br />

showed a very high efficiency (83%). Vitality<br />

was not significantly affected by both labelling<br />

strategies. Labelling with MNPs did not affect DC<br />

imaging life<br />

immune-phenotype or functionality, as demonstrated<br />

by CD86 and CD83 expression levels, T-cell proliferation<br />

and INF-γ production. Migration assays<br />

showed that Ag-loaded DCs were able to migrate<br />

in the presence of stimulatory chemokines (6Ckine<br />

and MIP3β). MRI evidenced the presence of an hypointense<br />

signal in both axillary and popliteal LN,<br />

in relation to the injection site, 4h after cell injection.<br />

Signal was visible up to 24 h after DC injection.<br />

Perl’s staining of LN sections after in vivo injection<br />

of labelled DCs loaded with the antigens, showed<br />

the presence of iron within the node, indicating that<br />

mature and labelled DCs migrate in vivo from the<br />

site of injection to the DLN. SPET imaging demonstrated<br />

as well DCs migration from the injection<br />

site to the DLN. Ex vivo analysis of different organs<br />

showed a statistically significant higher signal within<br />

the LN omolateral to the injection site in comparison<br />

with the controlateral one.<br />

Conclusions: Labelling protocols do not perturb<br />

DC physiology and functionality. Dynamic in vivo<br />

MRI and SPET imaging monitoring of DCs distribution<br />

will shed light on the fundamental parameters<br />

responsible for anti-neoplasic efficacy [3] , while<br />

the use of clinically approved MNPs and tracers will<br />

speed up the transfer from pre-clinical studies to<br />

clinical trials.<br />

Acknowledgement: this work is supported by the FP6<br />

funded Hi-CAM project (LSHC-CT-2006-037737),<br />

PRIN (20082NHWH9) and AIRC (IG2009-9311)<br />

and by fellowships from the Doctorate School of<br />

Molecular Medicine, University of Milan.<br />

References:<br />

1. de Vries IJ et al. Nature Biotech, 2005,23(11):1407-<br />

1413<br />

2. Baumjohann D et al. Eur. J. Immunobiology 2006; 36:<br />

2544-2555<br />

3. Lucignani G et al. TRENDS in Biotechnology 2006;<br />

24(9): 410-418


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

The type 2 cannabinoid receptor as a new PET reporter gene for the brain<br />

Vandeputte C. (1) , Evens N. (1) , Toelen J. (1) , Deroose C. (1) , Ibrahimi A. (1) , Verbruggen A. (1) , Debyser Z. (1) , Bormans G. (1) ,<br />

Baekelandt V. (1) , Van Laere K. (1) .<br />

(1) K.U. Leuven, Belgium<br />

caroline.vandeputte@med.kuleuven.be<br />

Introduction: Reporter genes play an important role<br />

in the understanding of gene expression and function<br />

in living subjects. However, for the brain no<br />

successful PET reporter systems are available with<br />

low endogenous background gene expression and<br />

good blood-brain-barrier (BBB) penetration of the<br />

PET probe. The aim of this study was to develop<br />

a new PET reporter gene system which can be applied<br />

to the brain. The type 2 cannabinoid receptor<br />

(CB 2 ) has a very low brain expression in physiological<br />

conditions and CB 2 PET radioligands crossing<br />

the BBB were recently developed.<br />

Methods: We constructed lentiviral (LV) and<br />

adeno-associated viral vector (AAV) transfer plasmids<br />

encoding human CB 2 , harboring a point mutation<br />

at position 80 (D80N) referred to as CB 2 (D80N),<br />

as such or in combination with enhanced green fluorescent<br />

protein (eGFP) or firefly luciferase (FLuc).<br />

Rats were stereotactically injected with either 5 µl of<br />

AAV-eGFP-T2A-CB 2 (D80N) in the right striatum<br />

and 5 µl of AAV control vector in the left striatum<br />

or 5 µl of AAV-fLuc-T2A-CB 2 (D80N) in the right<br />

striatum. At different time points (6, 13, 18, 73, 96<br />

and 252 days) after stereotactic injection of the<br />

AAV, a CB 2 selective carbon-11 labeled radioligand<br />

[ 11 C]GW405833 was injected intravenously and<br />

dynamic µPET images (Focus 220, Siemens) were<br />

acquired. BLI scans were performed at 16, 58 and<br />

281 days after surgery. Time-activity curves (TAC)<br />

and parametric binding potential maps were determined.<br />

The animals were sacrificed and perfused<br />

and double immunohistochemical staining against<br />

CB 2 and eGFP of the brain slices was performed.<br />

Results: The observed CB 2 binding potential increased<br />

over time and reached a maximum in right<br />

striatum between 18 and 58 days after vector injection<br />

in the brain. The time-activity curves persistently<br />

expressed an increased uptake in right<br />

striatum compared to control left striatum and<br />

cerebellum. Immunohistochemical analysis showed<br />

colocalization of both CB 2 and eGFP in right striatum.<br />

In contrast, only eGFP expression was seen in<br />

the contralateral hemisphere.<br />

Conclusions: We have successfully developed a new<br />

PET reporter gene system consisting of a lentiviral<br />

or an adeno-associated viral vector expressing<br />

the CB 2 receptor as the reporter gene which can be<br />

quantified for several months.<br />

Acknowledgement: We gratefully acknowledge the<br />

financial support by the European Commission for<br />

EC-FP6-STREP-STROKEMAP, BRAINSTIM SBO-<br />

IWT-060838, DIMI LSHB-CT-2005-512146 and<br />

the K.U. Leuven Center of Excellence ‘MoSAIC’<br />

(Molecular Small Animal Imaging Center).<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

YIA applicant<br />

day2<br />

Parallel Session 8: GENE and CELL based therapies - together with CliniGene


86<br />

WarSaW, poland May 26 – 29, 2010<br />

Department of Nuclear Medicine , Klinikum rechts der Isar<br />

Technische Universität München, Ismaninger Straße 22<br />

D-81675 München, Germany<br />

Phone: +49-(0)89-4140-4586, Fax: +49-(0)89-4140-4841<br />

Graduation:<br />

1981-1985 Study of Chemistry, University of Cologne<br />

1992-1996 Diploma and PhD, Radiochemistry,<br />

Res. Center Juelich, Germany<br />

1995-1997 Scientific Assistant, Department of Nuclear Medicine,<br />

Technische Universität München (TUM)<br />

1997-2004 Research Associate, Department of Nuclear Medicine (TUM)<br />

Academic Appointments<br />

since 2003 five national and international offers (W3 Radiopharm. Chem. , C4 Bioinorg. and Radiopharm.<br />

Chem., C3 Radiopharm. Chem., Assoc. Prof. for Radiology, Assoc. Prof. for Radiat. Oncol.). Since 2004<br />

Professor for Radiopharmaceutical Chemistry at the TU München, Faculty of Medicine)<br />

Scientific Focus<br />

Development of specific molecular probes for the non-invasive imaging of diseases, new strategies for<br />

production of radiopharmaceuticals, radionuclide therapy, tissue selective targeting of imaging probes and<br />

therapeutic compounds, transfer of genomic and proteomic information into new imaging methods.<br />

imaging life<br />

Hans-Jürgen Wester


Molecular imaging of CXCR4 receptors<br />

<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

Wester H.J. (1) , Demmer O. (2) , Dijkgraaf I. (1) , D´Alessandria C. (1) , Kessler H. (2) .<br />

(1) Department of Nuclear Medicine, Klinikum rechts der Isar, Technische Universität München, 81675 Munich, Germany<br />

(2) Institute for Advanced Study, Technische Universität München, Lichtenbergstrasse 4, 85747 Garching, Germany<br />

H.J.Wester@lrz.tu-muenchen.de<br />

Chemokines play a key role in tumor metastasis and are<br />

also involved in tumor growth. The receptor subtype<br />

CXCR4 has been found to be highly expressed in >30<br />

different types of cancer. Similar to the homing of hematopoietic<br />

progenitor and stem cells, the chemokine<br />

receptor subtype CXCR4 and its endogenous ligand,<br />

the stromal cell derived factor 1-alpha (SDF1-alpha or<br />

CXCL12), have been found to be responsible for “hijacking”<br />

circulating tumor cells in organs and tissues<br />

expressing CXCL12. In primary tumors, expression<br />

and binding of CXCL12 to CXCR4 promotes tumor<br />

proliferation and activates cell migration. In addition,<br />

CXCL12 induces recruitment of progenitor cells, which<br />

allows for tumor angiogenesis. It has been demonstrated<br />

that CXCR4 gene expression in mammary cancer<br />

cells is coregulated by Her2/neu, and also activated by<br />

HIF-1-alpha, implicating hypoxia-induced metastasis.<br />

A high CXCR4 density has also been reported for cancer<br />

stem cells. Consequently, CXCR4 directed therapies<br />

are currently under development aiming to inhibit<br />

tumor growth and metastasis by blocking CXCL12<br />

binding to CXCR4 using antibodies, large and small<br />

peptides as well as small molecules. CXCR4-directed<br />

PET probes should be useful tracers for monitoring<br />

those CXCR4-directed therapies in the future, i.e. for<br />

patient selection, therapy planning and monitoring<br />

response and may also independently allow for imaging<br />

the ´metastatic potential` of primary tumors.<br />

Antibodies, radiolabeled SDF, medium sized peptides<br />

and newly developed cyclic CXCR4 peptides antagonists<br />

are currently assessed as probes for monitoring<br />

CXCR4 expression or CXCR4 targeted therapies<br />

using metastasizing tumor models in rodents.<br />

With the aim to develop suitable radiolabeled probes to<br />

image CXCR4 receptor expression, we initially started<br />

with the evaluation of 4-[ 18 F]Fluorobenzoyl-TE14011-<br />

Me. Initial experiments showed unfavourable in vitro<br />

characteristics and low labelling yields. Using a similar<br />

approach by Hanoka et al (2006), Ac-TZ14011<br />

(Acetyl-Arg 1 -Cit 6 -Arg 7 -Lys 8 (In-DTPA)-T140-amide),<br />

labelled with In-111, showed high uptake in liver and<br />

kidney of tumor bearing mice reached, whereas tumor<br />

uptake was only negligible.<br />

Exploiting our experience in the development of small<br />

peptides, i.e. on cyclic RGD- pentapetides as high affinity<br />

ligand for the alpha(v)beta(3) integrin, we focussed<br />

our research on small CXCR4-binding pentapeptides.<br />

We have synthesized approximately 150<br />

monomeric and dimeric peptides and evaluated their<br />

binding characteristics on Jurkat cells and transfected<br />

CMS5 cells stably expressing the hCXCR4 receptor.<br />

Selected candidates were labelled with 123,125 I-iodine,<br />

18 F-fluorine and 68 Ga-gallium and investigated in nude<br />

mice bearing CMS5/CXCR4+ fibrosacoma and metastasizing<br />

OH-1 SCLC tumors. Based on c(Gly-D-Tyr-<br />

Orn-Arg-NaI), 4-fluorobenzoylated and 2-fluoropropionylated<br />

versions showed IC 50 of 11±2 nM and 35±1<br />

nM ( 125 I-CXCL12 as competitor), and radioiodinated<br />

c(Gly-D-Tyr-Arg-Arg-NaI) exhibited an IC 50 of 4nM.<br />

We could demonstrate that N-methylated peptides<br />

showed an improved IC 50 when compared to the nonmethylated<br />

analogues. A variety of linkers were investigated<br />

allowing for conjugation of chelators without<br />

affecting the affinity of the entire molecule. The<br />

DOTA-Ahx-Asp (Ahx= aminohexanoic acid) conjugate<br />

of a cxclic D-Orn-peptide version showed an IC 50<br />

of 38nM (with free chelator) and 13.7nM after complexation<br />

with indium. A variety of dimers were synthesized,<br />

the linkers and linker length optimized and<br />

the generated constructs assessed. C4-C16 dicarbonic<br />

acid spacers showed a continuously increasing affinity<br />

in the series C4 (6.6nM) – C10 (2.3nM), whereas<br />

longer spacers were unsuitable. The first of a series of<br />

promising 68 Ga-labeled peptides is renally excreted<br />

leading to high tumor-to-background contrast and<br />

high contrasting small animal PET-imaging approx.<br />

1h post injection.<br />

Compared to the other CXCR4-probes described,<br />

the small cyclic peptides developed offer significant<br />

advantages and will allow for broadening state-ofthe-art<br />

high contrast peptide receptor imaging to<br />

another important class of GPCRs.<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

day2<br />

<strong>ESMI</strong> Plenary Lecture 4 by Hans-Jürgen Wester


88<br />

WarSaW, poland May 26 – 29, 2010<br />

Evaluation of the temporal window for drug delivery following ultrasound mediated<br />

membrane permeability enhancement<br />

Yudina A. (1) , Lepetit-Coiffé M. (1) , Moonen C. (1) .<br />

Laboratoire IMF CNRS UMR 5231 / Université Bordeaux 2, France<br />

anna@imf.u-bordeaux2.fr<br />

Introduction: Ultrasound (US)-mediated delivery<br />

is a new therapeutic option to locally facilitate the<br />

passage of drugs through cell plasma membrane<br />

by reversibly altering its permeability[1] possibly<br />

via the formation of pores that are known to spontaneously<br />

reseal within a short time – seconds to<br />

minutes[2]. However under certain conditions the<br />

effects of US for drug delivery last for hours after<br />

the exposure[3]. The present work is the first in<br />

vitro attempt to confirm and quantitatively assess<br />

the temporal window for the US-mediated intracellular<br />

drug delivery by live-cell imaging.<br />

Methods: Cell-impermeable optical chromophores<br />

with fluorescence intensity increasing 100-1000<br />

fold upon intercalation with nucleic acids served<br />

as smart agents for reporting cellular uptake. Opticell<br />

chambers with a monolayer of C6 cells were<br />

subjected to ultrasound in the presence of microbubbles<br />

followed by varying delays between<br />

0 and 24 hours before addition of Sytox Green<br />

optical contrast agent. Micro- and macroscopic<br />

fluorescence imaging was used for qualitative and<br />

quantitative analysis.<br />

Results: Strong enhancement of the fluorescence<br />

signal upon binding to nucleic acids allowed efficient<br />

visualization of the local effect of US on internalization<br />

of cell-impermeable intercalating dyes.<br />

Up to 25% of viable cells showed uptake of contrast<br />

agent with a half time of 8 hours, with cellular uptake<br />

persisting even at 24 hours (Figure 1). Only<br />

cells exposed to ultrasound showed the effect.<br />

imaging life<br />

Conclusions: Optical imaging showed that temporal<br />

window of increased membrane permeability is<br />

much longer than previously suggested. This may<br />

have important repercussions for in vivo studies in<br />

which membrane permeability may be temporally<br />

separated from drug administration to better adapt<br />

to pharmacokinetic / pharmacodynamic properties<br />

of the drug or drug carrier.<br />

Acknowledgement: This work is supported by the<br />

EC-project FP7-ICT-2007-1-213706 SonoDrugs and<br />

Foundation InNaBioSanté-project ULTRAFITT. Microscopy<br />

was performed in the Bordeaux Imaging<br />

Center of the Neurosciences Institute of the University<br />

of Bordeaux II.<br />

References:<br />

Figure 1. In the absence of US<br />

only occasional cells with the<br />

compromised membrane show<br />

the uptake of cell-impermeable<br />

Sytox green (A). However the<br />

effects of the US on membrane<br />

permeability can last up to<br />

24h (B), with the fluorescence<br />

intensity increasing as the time<br />

between US application and<br />

fluorophore administration<br />

shortens (C-F).<br />

1. Hernot S, Klibanov AL; Adv Drug Deliv Rev. 60(10):1153-<br />

66 (2008)<br />

2. van Wamel A et al.; J Control Release. 112(2):149-55<br />

(2006)<br />

3. Hancock HA et al. Ultrasound Med Biol. 35(10):1722-36<br />

(2009)


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

Integrisense: a novel near-infrared fluorescent probe for α v β 3 integrin and its applications in<br />

drug discovery<br />

Sur C. (1) , Lin S.A. (1) , Gleason A. (1) , Kossodo S. (1) , Pickarski M. (1) , Coleman P. (1) , Rajopadhye M. (2) , Duong L. T. (1) , Yared W. (2) ,<br />

Peterson J. (2) , Bednar B. (1) .<br />

(1) Merck Research Laboratories, Westpoint US<br />

(2) ViSen Medical.<br />

cyrille_sur@merck.com<br />

Introduction: The α v β 3 integrin belongs to the superfamilly<br />

of dimeric transmembrane cell adhesion<br />

receptors involved in critical biological processes<br />

such as cell-to-cell, and cell-to-extracellular<br />

matrix binding. As these activities are central to<br />

pathological conditions such as inflammation and<br />

tumor progression, the RGD-binding α v β 3 integrin<br />

has been considred as clinically-relevant biomarkers<br />

of these disease states. Although, RDG peptides<br />

have been developed for PET and SPECT imaging,<br />

the operating cost of these modalities limits their<br />

widespread deployment in drug discovery teams. In<br />

contrast optical molecular imaging allows the noninvasive<br />

monitoring and quantification of fluorescent<br />

probes in vivo with less expensive equipment<br />

that can be installed at multiple research sites. In<br />

order to take full advantage of the rich biology of integrins,<br />

a partnership has been established between<br />

Merck Research Laboratories and VisEn Medical to<br />

develop a novel near-infrared (NIR) fluorescent integrin<br />

probe, IntegriSense TM . The pharmacological<br />

and biological characterization of IntegriSense TM ,<br />

a peptidomimetic antagonist-based molecule with<br />

improved specificity and binding affinity will be<br />

presented together with research applications in the<br />

oncology and atherosclerosis research fields.<br />

Methods: The in vitro cell biology and pharmacological<br />

profiling of IntegriSense TM were conducted<br />

with recombinant HEK-293 cells expressing α v β 3<br />

and included confocal microscopy for cellular localization<br />

and flow cytometry for binding and kinetic<br />

constant determination. In vivo pharmacodynamics<br />

and tumor localization studies were performed<br />

in female NU/NU mice bearing different human<br />

tumor xenografts. IntegriSense TM NIR fluorescent<br />

signal was acquired with a Fluorescent Molecular<br />

Tomography imaging system (FMT2500). The potential<br />

application of IntegriSense TM in atherosclerosis<br />

was evaluated in apolipoprotein E-deficient<br />

(ApoE-/-) and human cholesteryl ester transfer<br />

protein (CETP) knockin/LDL receptor-deficient<br />

(C57BL/6-Tg(CETP)-Ldlrtm1) mice. Transgenic<br />

as well as control C57Bl/6 mice were fed a cholesterol<br />

enriched diet (9% fat, 0.15% cholesterol) to<br />

induce atherosclerosis. IntegriSense TM NIR signal<br />

was measured in vivo 24 and 48 hours after probe<br />

injection and its localization in inflamed aortas was<br />

further evaluated by ex-vivo imaging and histology.<br />

Results: Integrisense-680 has absorption/emission<br />

spectra centered at 674nm/692nm and an extinction<br />

coefficient of 2.02x10 5 M -1 cm -1 . Binding of IntegriSense<br />

TM to HEK-293 stably expressing recombinant<br />

human α v β 3 yielded a Kd of 4.2±0.6 nM and<br />

a dissociation constant koff of 1.08x10 -4 s -1 . Plasma<br />

pharmacokinetic analysis revealed a two compartmental<br />

profile with t1/2 of 6 min and 210 min corresponding<br />

to clearance of free and bound IntegriSense<br />

TM , respectively. Biodistribution studies in<br />

mice with xenograft tumors showed a specific uptake<br />

of IntegriSense TM by tumor cells and supported<br />

using IntegriSense TM to monitor tumor growth in<br />

animal models.<br />

In vivo measurements with FMT2500 in mice atherosclerosis<br />

models detected a longitudinal increase in<br />

IntegriSense TM fluorescence in the thorax area that<br />

was three to five fold higher when compared with<br />

control animals. This NIR signal originated from<br />

atherosclerotic lesions in the aortic arch, carotid,<br />

and subclavian arteries as confirmed by ex vivo imaging<br />

of the dissected vessels and histopathology.<br />

Conclusions: This report demonstrated that the α v β 3<br />

integrin optical reporter, IntegriSense TM developed<br />

by Merck and VisEn partnership can be used as a<br />

molecular imaging biomarker for in vivo imaging<br />

studies. For instance, IntegriSense TM can be used to<br />

monitor tumor growth as well as to detect inflammation<br />

in atherosclerosis plaques. The ability of<br />

this biomarker to report on several important cellular<br />

and physiological functions makes it a tool of<br />

choice to evaluate preclinically the therapeutic potential<br />

of novel anticancer and atherosclerosis drugs.<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

day2<br />

Plenary Session on current contribution of IMAGING TECHNOLOGIES to DRUG DEVELOPMENT


90<br />

WarSaW, poland May 26 – 29, 2010<br />

Harnessing the power of bioluminescence to cross the in vitro–in vivo divide<br />

Watson J. (1) , Klaubert D. (1) , Biserni A. (2) , Ciana P. (2) , Maggi A. (2) , Allard S. (1) .<br />

(1) Promega<br />

(2) TOP (Transgenic Operative Products)srl<br />

John.watson@promega.com<br />

Introduction: Bioluminescence imaging is accomplished<br />

by sensitive detection of light emitted following<br />

chemical reaction of the luciferase enzyme<br />

with substrate. The imaging process in animal<br />

models requires a reporter construct that leads to<br />

production of luciferase enzyme. The most commonly<br />

used reporter for this purpose is a construct<br />

that can express firefly luciferase (1). Data will be<br />

presented that shows how a number of biomarkers<br />

can be measured using bioluminescence in vitro,<br />

including caspase-3/7, ATP and transcriptional<br />

activation. We will also describe the in vivo characterization<br />

of VivoGlo Caspase-3/7 Substrate, a<br />

modified firefly luciferase substrate that in apoptotic<br />

cells is cleaved by caspase-3 to liberate aminoluciferin,<br />

which can be consumed by luciferase<br />

to generate a luminescent signal. Evidence will be<br />

shown illustrating the possibility to merge a line of<br />

reporter mice (repTOP) engineered for the ubiquitous<br />

expression of the luciferase reporter gene<br />

(2-4) and the modified luciferase substrate, Vivo-<br />

Glo Caspase-3/7 Substrate, to measure molecular<br />

apoptotic events in whole living animals. We will<br />

also demonstrate that this substrate can be used<br />

to non-invasively observe the apoptotic affects of<br />

chemotherapy several days before they are able to<br />

be detected by traditional methods (5).<br />

Methods: In this study, liver apoptosis was induced<br />

in luciferase reporter mice by a single i.p. injection<br />

of D-galactosamine (D-GalN; 800mg/kg) and<br />

Lipopolysaccharide (LPS; 100mg/kg) (6). Six hours<br />

after LPS/D-GalN or vehicle administration, mice<br />

were treated i.p. with increasing doses (17-150mg/<br />

kg) of the VivoGlo Caspase-3/7 substrate (Z-<br />

DEVD-Aminoluciferin, Sodium Salt) and subjected<br />

to the bioluminescence in vivo imaging procedure.<br />

Results: The in vivo imaging data obtained clearly<br />

showed a dose-dependent increase of photon<br />

emission in the hepatic area of mice treated with<br />

D-GalN/LPS. No photon emission was observed in<br />

organs not affected by the apoptotic treatment. A<br />

complete analysis of pro-apoptotic effects induced<br />

by the treatment was carried out by ex vivo imaging<br />

acquisition of photon emission in several dissected<br />

imaging life<br />

tissues. This investigation confirmed that light<br />

emission observed in vivo was produced selectively<br />

by liver and adipose tissues. Western blot analysis<br />

and the measure of Caspase-3/7 enzymatic activity<br />

fully supported in vivo imaging data.<br />

Conclusions: The work here shows the power of<br />

a modified luciferin substrate such as VivoGlo<br />

Caspase-3/7 Substrate to detect apoptotic cells in<br />

living animals, providing an important advancement<br />

over current imaging methodologies based<br />

on fluorescence, nuclear or magnetic resonance<br />

including: virtually no background, high sensitivity<br />

and simple instrumentation needed for the in vivo<br />

imaging measurement of the Caspase-3/7 activity.<br />

The present application carried out in reporter<br />

mice genetically engineered to develop specific<br />

cancers will open the way to novel, more predictive<br />

approaches for the study of anti-cancer treatments<br />

that will enable researchers to measure drug efficacy<br />

in space and time, providing relevant information<br />

to be rapidly translated to human therapy.<br />

References:<br />

1. Zinn, KR et al; ILAR Journal. 49:103-115 (2008)<br />

2. Ciana, P et al; Nat. Med. 9:82-86 (2003)<br />

3. Maggi, A and Ciana, P; Nat. Rev. Drug Discov. 4:249-255<br />

(2005)<br />

4. Maggi, A et al; Trends Pharmacol. Sci. 25:337-342<br />

(2004)<br />

5. Hickson, J et al; Cell Death Differ. doi:10.1038/<br />

cdd.2009.205 (2010)<br />

6. Nakama, T et al; Hepatology. 33:1441-1450 (2001)


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

In vivo imaging in drug discovery: the example of application in the development of novel<br />

estrogenic compounds.<br />

Biserni A. (1) , Rando G. (2) , Ciana P. (2) , Komm B. (3) , Maggi A. (2) .<br />

(1) TOP (Transgenic Operative Products)srl,<br />

(2) University of Milan,<br />

(3) Wyeth Research .<br />

andrea.biserni@top-mice.com<br />

Introduction: Intracellular Receptor (IR) signalling<br />

is temporally organized at different levels: at the<br />

cellular level, post-translational modification and<br />

proteolysis were shown to be responsible for IRdependent<br />

recruitment of co-regulators and proteins<br />

of the transcriptional apparatus necessary to<br />

modulate IR activity in time. At the whole organism<br />

level, circadian rhythms and pulsatility of hormone<br />

release may be responsible for maintaining<br />

a state of activity of IR that is critical for hormone<br />

action. With hormone therapy, the reinstatement<br />

of the receptor oscillatory activity may be a key element<br />

for achieving the desired beneficial effect. To<br />

date, the lack of appropriate models to study the<br />

response to pharmacological treatments in time<br />

has been a limiting factor in the study of spatiotemporal<br />

activity of drugs. The availability of the<br />

ERE-Luc reporter mouse model where the application<br />

of BLI-based imaging methodologies allows<br />

one to study estrogen receptor activity in living animals,<br />

opens the way to a more in-depth analysis of<br />

the effect of estrogen therapy (ET). The aim of this<br />

study was to set up a protocol for the study of ET<br />

and SERM (Selective Estrogen Receptor Modulator)<br />

therapies to verify their ability to efficaciously<br />

restore the effects of the natural endocrine state in<br />

reproductive and non reproductive organs.<br />

Methods: The study was carried out in the ERE-Luc<br />

reporter mouse model where the luciferase reporter<br />

gene is expressed under the transcriptional control<br />

of an ER responsive promoter. In this model, MARs<br />

(Matrix Attachment Regions) insulator sequences<br />

flanking the transgene guarantee a ubiquitous and<br />

tightly regulated expression of the biosensor making<br />

it suitable for pharmacological studies. In the present<br />

study, we treated ovariectomized, female mice with<br />

CE (3 mg/kg/day), raloxifene (RAL; 10 mg/kg/day)<br />

and bazedoxifene (BZA, 10 mg/kg/day) for 21 days.<br />

To measure ER state of activity, mice were subjected to<br />

in vivo imaging. Photon emission was measured by the<br />

use of a segmentation algorithm enabling the automatic<br />

quantification of photon emission in different body regions<br />

of mouse images. At the end of the study a further<br />

analysis was carried out on tissue extracts by luciferase<br />

enzymatic assay.<br />

Results: In the initial analysis, we evaluated the<br />

effect of each treatment in the genital, hepatic,<br />

abdominal area and limbs at 0, 3, 7, 14 and 21d.<br />

CE had a major effect in the genital and hepatic<br />

areas shown by a significant increase of ER activity.<br />

As expected the effect of SERMs was significant<br />

in the skeletal (limbs) where ER activity<br />

started to be significantly increased only after<br />

day 14. In the body areas affected by treatments,<br />

we observed time-dependent changes: i.e. in the<br />

liver CE activity decreased with time, while in<br />

the limbs ER activity was shown to increase with<br />

the time of exposure to SERMs. This observation<br />

prompted us to perform a more in-depth analysis<br />

of ER activity by measuring photon emission<br />

daily. This second analysis demonstrated that ER<br />

activity oscillates in time with a period and amplitude<br />

that is tissue specific and is differentially<br />

affected by the hormonal treatments. Furthermore,<br />

the analysis of ER oscillation revealed that<br />

ovariectomy does not fully disrupt the fluctuation<br />

observed in intact, cycling mice indicating<br />

the existence of mechanisms other than circulating<br />

hormone responsible for ER transcriptional<br />

activity. The treatments evaluated partially restored<br />

the physiological oscillatory behavior of<br />

the receptor.<br />

Conclusions: Our study: Provides evidence for a<br />

novel, long paced, oscillatory activity of ERs that is<br />

independent of ovarian function. This oscillatory<br />

ER activity may be relevant for the whole body homeostatic<br />

control and may be of relevance for the<br />

identification of safer and more efficacious therapies.<br />

Points to the importance of whole animal imaging<br />

for drug development and for the study spatio-temporally<br />

drug activity on targets.<br />

Novel protocols for a wider application of BLI to<br />

the study of molecular events in vivo should be<br />

developed.<br />

Acknowledgement: Grants from: NoE DIMI<br />

LSHB-CT-2005-512146; IP CRESCENDO LSHM-<br />

CT-2005-018652), National Institutes of Health<br />

(RO1AG027713), and Wyeth Pharmaceutical Co.<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

day2<br />

Plenary Session on current contribution of IMAGING TECHNOLOGIES to DRUG DEVELOPMENT


DAY<br />

3


• <strong>ESMI</strong> Plenary Lecture 5: Jagat Narula (Irvine, USA)<br />

Molecular Imaging of unstable coronary plaques<br />

Chairs: Uwe Haberkorn (Heidelberg, Germany), Helmut Maecke (Freiburg, Germany)<br />

• Parallel Session 9: Cardiovascular II<br />

Chairs: Klaas Nicolay (Eindhoven, The Netherlands), Michael Schäfers (Münster, Germany)<br />

• Parallel Session 10: Cancer II – together with COST action BM0607<br />

Chairs: Marion de Jong (Rotterdam, The Netherlands), Fabian Kiessling (Aachen, Germany)<br />

• Plenary Session and Closing Ceremony<br />

Chairs: John Clark (Edinburgh, UK), Bertrand Tavitian (Orsay, France)<br />

Day 3 - Friday May 29, 2010


94<br />

WarSaW, poland May 26 – 29, 2010<br />

imaging life<br />

Jagat Narula<br />

Jagat Narula MD, PhD, FACC, FRCP [Edin, Hon]<br />

Professor of Medicine and Chief, Division of Cardiology<br />

Director, Memorial Heart & Vascular Institute<br />

Medical Director, Edwards Lifesciences Center for Advanced Cardiovascular Technology<br />

University of California, Irvine School of Medicine<br />

Jagat Narula completed his cardiology training in India at the All India Institute of Medical Sciences, Delhi,<br />

and relocated to Massachusetts General Hospital and Harvard Medical School in 1989. After fellowships in<br />

heart failure, transplantation, and cardiovascular imaging, he joined the Massachusetts General cardiology<br />

faculty. In 1997, he moved to Hahnemann University School of Medicine in Philadelphia. At Hahnemann,<br />

he was the Thomas J. Vischer Professor of Medicine, Chief of Division of Cardiology, and Vice-Chairman<br />

of Medicine. He joined University of California, Irvine in 2003 as Professor of Medicine Chief, Division<br />

of Cardiology, and Associate Dean for Research. Dr. Narula is involved in clinical and basic research in<br />

the fields of heart failure and atherosclerosis, with major emphasis on development of novel noninvasive<br />

imaging techniques. He has made vital contributions to the imaging of apoptotic cell death in heart<br />

muscle, and to the imaging of atherosclerotic plaques that are vulnerable to rupture. His research is not<br />

limited by any one imaging modality; and uses integrated imaging approaches for better identification<br />

of cardiovascular pathology. His research is funded, in part, by grants from the National Institutes of<br />

Health. Dr. Narula has authored more than 700 research publications or presentations and edited 25 books<br />

and journal supplements. He has been awarded “Best Young Investigator” on several occasions by the<br />

Cardiovascular Council, the Society of Nuclear Medicine, and the American Society of Nuclear Cardiology.<br />

He serves on various committees of the American Heart Association. He is the editor-in-chief of the Journal<br />

of the American College of Cardiology- Imaging. He has also been an Associate Editor of the Journal of<br />

the American College of Cardiology and the Founder Editor of Heart Failure Clinics of North America.


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

Molecular imaging of unstable coronary plaques<br />

Narula J. .<br />

University of California, USA<br />

narula@uci.edu<br />

Sudden cardiac death and acute myocardial infarction<br />

often occur as the first manifestation of coronary<br />

artery disease. Otherwise asymptomatic individuals<br />

with subclinical atherosclerosis almost always<br />

have a classic risk-factor profile and it is essential<br />

that they are identified before the occurrence of an<br />

acute coronary event. The ability to recognize such<br />

individuals requires the development of noninvasive<br />

strategies that can localize unstable atherosclerotic<br />

lesions. Plaques that are vulnerable to rupture demonstrate<br />

distinct histological characteristics, including<br />

large plaque and necrotic core volumes covered<br />

by attenuated fibrous caps and extensive remodeling<br />

of the vessel at the lesion site. The morphologic features<br />

can be characterized by CT angiography and<br />

magnetic resonance imaging.<br />

In addition to characteristic morphological profile,<br />

monocyte-macrophage infiltration of the fibrous<br />

cap is one of the strongest determinants of plaque<br />

instability, and amenable to molecular inaging. After<br />

crossing the intimal border, infiltrating monocytes<br />

express receptors for chemoattractants (such as<br />

MCP-1 or adhesion molecules), and subsequently develop<br />

scavenger receptors in the subintimal layers for<br />

ingestion of oxidized LDL. Although multiple cellular<br />

processes can be targeted for the identification of<br />

plaque inflammation only a few molecular imaging<br />

strategies have been successfully employed for clinical<br />

imaging. The inflammatory cells in plaques have<br />

high metabolic activity. In contrast to granulocytes,<br />

which carry a supply of glycogen to provide energy<br />

for their phagocytic activities, the macrophages in<br />

an atheroma require exogenous glucose for their<br />

metabolism. Incidental uptake of a glucose analog,<br />

[18F]-FDG, commonly used to characterize malignant<br />

tumors, has been demonstrated in the aorta,<br />

carotid arteries and, rarely, the coronary vessels of<br />

patients with tumors. Carotid endarterectomy specimens<br />

have demonstrated that uptake correlates with<br />

macrophage content, and is reduced in patients<br />

treated with statins. A prospective study has demonstrated<br />

that FDG uptake can be seen in the coronary<br />

arteries if background FDG uptake in the myocardium<br />

is adequately suppressed. Following coronary<br />

interventions, intense FDG uptake has been seen at<br />

the sites of stent implantations in patients with acute<br />

coronary events; no uptake was observeat the stent<br />

sites in patients with stable disease. Concurrent CT<br />

angiography was performed in these patients for precise<br />

anatomical localization of FDG uptake on PET<br />

imaging. Another clinical approach with molecular<br />

imaging has utilized radiolabeled annexin A5 to<br />

target apoptotic macrophages in the atherosclerotic<br />

lesions. A large proportion of macrophages in the<br />

attenuated fibrous caps of vulnerable and ruptured<br />

plaques undergo apoptosis. The annexin uptake in<br />

the carotid vessels has been predominantly seen in<br />

symptomatic carotid disease. Annexin imaging of<br />

coronary vessels has not been attempted. Various other<br />

molecules targeting evolution of receptors such as<br />

MCP-1 and scavenger receptors, as well as those targeting<br />

the products of inflammation such as metalloproteinases<br />

are being successfully employed for the<br />

imaging of atherosclerosis in experimental models.<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

day3<br />

<strong>ESMI</strong> Plenary Lecture by Jagat Narula


96<br />

WarSaW, poland May 26 – 29, 2010<br />

Advances in contrast-enhanced MRI of the mouse heart<br />

Strijkers G. .<br />

Biomedical NMR Eindhoven, The Netherlands<br />

g.j.strijkers@tue.nl<br />

Introduction: In preclinical cardiovascular research<br />

using mouse models, MRI has proven to<br />

be the imaging modality of choice for studying<br />

cardiac pathology. This is mainly thanks to its<br />

high temporal and spatial resolution, enabling<br />

accurate determination of cardiac functional<br />

parameters.<br />

MRI contrast agents are used to differentiate between<br />

viable and infarct myocardium. Recently, it<br />

has become possible to use MR contrast agents for<br />

the evaluation of the mouse myocardial perfusion<br />

status. Molecular MR imaging techniques are being<br />

developed, allowing contrast agents to specifically<br />

target disease markers, providing additional<br />

information about the infarction. The use of contrast<br />

agents in the mouse myocardium requires innovative<br />

mouse cardiac MRI techniques to quantify<br />

contrast agent kinetics and concentration in the<br />

myocardium.<br />

Results: In this presentation I will present recent<br />

advances in the contrast-enhanced MR imaging of<br />

the mouse heart.<br />

First, I will discuss new MR technology, which enables<br />

the assessment of mouse myocardial perfusion.<br />

This methodology involves intravenous injection of<br />

an MRI contrast agent, after which the first pass of<br />

the contrast agent through the heart is monitored<br />

in a time-series of images. The combination of the<br />

high heart rate (400-600 bpm), small heart size (5-6<br />

mm left ventricle (LV) diameter) and fast systemic<br />

blood circulation time (4-5 s) require a very fast<br />

imaging protocol while preserving adequate spatial<br />

resolution to visualize the regional myocardial inflow<br />

of the contrast agent.<br />

Secondly, I will discuss recent advances in the T1<br />

quantification of the mouse myocardium, which<br />

enables quantification of contrast agent concentration.<br />

The T1 quantification method uses a combination<br />

of 3D IntraGate FLASH steady-state imaging<br />

together with DESPOT1 analysis. The 3D IntraGate<br />

FLASH makes use of a navigator echo that retrospectively<br />

triggers the acquisition to the heartbeat<br />

and respiration cycle.<br />

imaging life<br />

Acknowledgement: Grant sponsor: Dutch Technology<br />

Foundation STW, applied science division of<br />

NWO and the Technology Program of the Ministry<br />

of Economic Affairs; Grant numbers: 07952 and<br />

10191. Grant sponsor: European Union Network of<br />

Excellence Diagnostic Molecular Imaging; Grant<br />

number: 512146 (LSHB-CT-2005-512146).


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

Molecular imaging of α v β 3 integrin expression with 18 F-galacto-RGD after experimental<br />

myocardial infarction: comparison with left ventricular remodeling and function.<br />

Saraste A. (1) , Sherif H. (1) , Nekolla S. (1) , Weidl E. (1) , Higuchi T. (1) , Reder S. (1) , Tapfer A. (1) , Botnar R. (1) , Wester H. J. (1) ,<br />

Schwaiger M. (1) .<br />

Technische Universität München, Germany<br />

antti.saraste@utu.fi<br />

Introduction: Myocardial infarction (MI) and subsequent<br />

left ventricular (LV) remodeling are the<br />

most frequent cause for development of chronic<br />

heart failure. Expression of α v β 3 integrin receptors<br />

is increased early after MI as part of the healing<br />

process. Poor infarct healing can contribute to progressive<br />

LV remodeling. 18 F-galacto-RGD (RGD) is<br />

a PET tracer that binds to α v β 3 integrin receptors.<br />

We studied whether molecular imaging of myocardial<br />

RGD uptake early after MI can predict longterm<br />

LV remodeling in rat.<br />

Methods: Wistar rats underwent permanent left<br />

coronary artery (LCA) ligation to induce myocardial<br />

infarction (MI) or sham operation. One week<br />

post-MI, rats were imaged with a small animal PET<br />

scanner after injection of 13N ammonia to define<br />

perfusion defect area, and 90 minutes post injection<br />

of RGD in order to measure α v β 3 integrin expression.<br />

Myocardial RGD uptake (%ID/cc) in the<br />

infarcted (defect area) and remote myocardium<br />

were measured in co-registered polar maps. Cardiac<br />

magnetic resonance imaging (MRI) with 1.5T<br />

scanner, small animal coil and ECG triggering was<br />

used to measure LV remodeling and function repeatedly<br />

1 week and 12 weeks post-MI.<br />

Results: One week after LCA ligation, RGD uptake<br />

was significantly higher in the defect area than<br />

in the remote myocardium of MI rats and in controls<br />

(0.2±0.05 vs. 0.06±0.03 and 0.07±0.04 %ID/<br />

cc, respectively, p20% increase in EDV, early RGD uptake<br />

in the defect area was 29% lower than in rats with<br />

less than


98<br />

WarSaW, poland May 26 – 29, 2010<br />

Existing and emerging animal models mimicking cardiovascular disease and their relevance<br />

for molecular imaging<br />

Schäfers M. .<br />

European Institute for Molecular Imaging – EIMI, University of Münster<br />

schafmi@uni-muenster.de<br />

Appropriate animal models, mimicking the pathology<br />

of cardiovascular human diseases is essential<br />

for progress in the field of cardiovascular research<br />

and for the transfer of preclinical results into the<br />

clinical setting. In principle, a variety of animal<br />

species can be used in cardiovascular research. Although<br />

larger animal such as pigs (a classical model<br />

for myocardial infarction) are used in cardiovascular<br />

research, the focus has been shifted to small<br />

imaging life<br />

animal models such rats and mice recent years.<br />

Animal models of cardiovascular diseases can rely<br />

both on genetic manipulations or surgical interventions<br />

and do nowadays cover the wide spectrum<br />

of cardiovascular human diseases. In this talk, examples<br />

for relevant human cardiovascular diseases,<br />

myocardial infarction and atherosclerosis, already<br />

being studied by small animal imaging is given.


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

Imaging of Matrix Metalloproteinase Activity in Vulnerable Human Carotid Plaques with<br />

Multispectral Optoacoustic Tomography<br />

Razansky D. (1) , Harlaar N. J. (1) , Hillebrands J. L. (2) , Taruttis A. (1) , Herzog E. (1) , Zeebregts C.J. (2) , Van Dam G. M. (2) ,<br />

Ntziachristos V. (1) .<br />

(1) Technical University of Munich and Helmholtz Center Munich, Germany<br />

(2) University Medical Center Groningen, The Netherlands.<br />

tdr@tum.de<br />

Introduction: The indication for a carotid endarterectomy<br />

is nowadays mainly based on symptomatology<br />

or, alternatively, on the degree of stenosis in<br />

carotid arteries (>80%). Those indications however<br />

do not provide an accurate assessment of plaque<br />

vulnerability and therefore only a small percentage<br />

of patients do actually benefit from the surgical intervention<br />

by preventing a major cerebro-vascular<br />

event especially in the asymptomatic group [1].<br />

High activity levels of tissue biomarkers, such as<br />

cathepsins, integrins, and matrix metalloproteinases<br />

(MMPs), have been associated with atherosclerotic<br />

plaque instability, thus can potentially be<br />

used for highly specific diagnosis [2]. While fluorescent<br />

tagging of such molecules has been amply<br />

demonstrated, no imaging method has so far been<br />

shown capable of resolving inflammation-associated<br />

tags with high fidelity.<br />

Methods: We showcase herein the ability to resolve<br />

activation of common probes sensitive to matrix<br />

metallo-proteinases with unprecedented image<br />

quality and resolution utilizing multi-spectral optoacoustic<br />

tomography (MSOT) [3], thus revealing<br />

atherosclerotic activity. Human carotid plaque specimens<br />

from patients were incubated ex vivo with<br />

an MMP-sensitive activatable fluorescent probe<br />

(MMPsense 680TM) directly after endarterectomy.<br />

The specimen were subsequently imaged using an<br />

MSOT scanner capable of simultaneous high resolution<br />

visualization of morphology and molecular<br />

activity with better than 200 micron resolution.<br />

Results: MSOT analyses identified heterogeneous<br />

MMP activity throughout the plaques, revealing<br />

hot and cold spot regions indicative of relatively<br />

high and low MMP activity respectively. The results<br />

from intact specimen corresponded well with<br />

epi-fluorescence images made on thin cryosections.<br />

Elevated MMP activity in the hot spot regions resolved<br />

was further confirmed by in situ zymography,<br />

accompanied by increased macrophage influx.<br />

Results from both MSOT investigations and histological<br />

sections confirmed that most of the plaque<br />

formation activity occurs, as expected, close to the<br />

bifurcation area of the carotid artery.<br />

Conclusions: We show, for the first time to our<br />

knowledge, the ability of multispectral optoacoustic<br />

tomography to deliver volumetric images of activatable<br />

molecular probe distribution deep from optically<br />

opaque tissues. High resolution mapping of MMP<br />

activity in the vulnerable plaque of human carotid<br />

specimen was demonstrated. This ability directly relates<br />

to clinical potential as it can allow highly specific<br />

visualization and staging of plaque vulnerability<br />

in atherosclerosis during surgical intervention or by<br />

intravascular or potentially non-invasive imaging;<br />

thus impacting therapeutic clinical decision.<br />

Acknowledgement: This research was partially supported<br />

by the German Research Foundation (DFG)<br />

Research Grant (RA 1848/1), ERC Senior Investigator<br />

Award, and the Medizin Technik BMBF award for<br />

excellence in medical innovation.<br />

References:<br />

1. Chambers BR et al; Cochrane Database Syst Rev.<br />

2:CD001923 (2000).<br />

2. Libby P.; Inflammation in atherosclerosis. Nature.<br />

420:868-874 (2002).<br />

3. Razansky D et al.; Nature Phot. 3:3525-3529 (2009).<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

day3<br />

Parallel Session 9: CARDIOVASCULAR II


100<br />

WarSaW, poland May 26 – 29, 2010<br />

c-Jun N-terminal kinase promotes inflammation at atherosclerosis-prone sites by enhancing<br />

expression and activity of NF-κB transcription factors<br />

Evans P. (1) , Cuhlmann S. (1) , Van Der Heiden K. (1) , Haskard D. (1) , Krams R. (1) , Gsell W. (1) , Carlsen H. (2) , Jones H. (1) .<br />

(1) Imperial College London,<br />

(2) University of Oslo, Norway.<br />

paul.evans@imperial.ac.uk<br />

Introduction: Atherosclerosis is a chronic inflammatory<br />

disease of arteries that causes heart attack or<br />

stroke. Early lesions contain monocytes and T lymphocytes<br />

which are recruited from the circulation<br />

to activated vascular endothelial cells (ECs). This<br />

process relies on NF-κB transcription factors which<br />

induce pro-inflammatory molecules (e.g. VCAM-1)<br />

in ECs. Atherosclerosis develops predominantly at<br />

branches and bends of arteries that are exposed to<br />

low, oscillatory shear stress (frictional force exerted<br />

by flowing blood on ECs). We previously demonstrated<br />

that c-Jun N-terminal kinase (JNK), a MAP<br />

kinase, is activated in ECs at atherosusceptible but<br />

not atheroprotected sites (Chaudhury et al 2010).<br />

Here we combined in vitro studies with molecular<br />

imaging techniques in murine models to study the<br />

function of JNK in arterial EC.<br />

Methods: We studied the function of JNK by<br />

performing microarray analysis of cultured<br />

ECs treated with a pharmacological JNK inhibitor.<br />

En face immunostaining was used to determine<br />

the expression of particular proteins in<br />

ECs at atheroprotected and atherosusceptible<br />

sites of murine arteries. In addition, we assessed<br />

NF-κB transcriptional activity in murine<br />

arteries of transgenic NF-κB-luciferase reporter<br />

mice by measuring luminescence using an ultrasensitive<br />

camera (Xenogen). Inflammation<br />

was also assessed by F18-FDG PET/CT imaging<br />

of murine arteries and identification of<br />

CD68-positive macrophages by immunostaining.<br />

Results: Transcriptome profiling of cultured ECs<br />

treated with a pharmacological inhibitor revealed<br />

that JNK functions as a positive regulator of NFκB<br />

transcription factors. This observation was<br />

confirmed by silencing of JNK1 and ATF2 (a<br />

downstream transcription factor) which led to<br />

reduced NF-κB expression in cultured ECs. We<br />

validated our findings by studying ECs in the aorta<br />

of wild-type and JNK1-/- mice. En face immunostaining<br />

revealed that EC expression of NF-κB<br />

and VCAM-1 and accumulation of CD68-positive<br />

macrophages was enhanced at atherosusceptible<br />

imaging life<br />

compared to atheroprotected sites in wild-type<br />

mice. Genetic deletion of JNK1 suppressed NF-κB<br />

and VCAM-1 expression at the atherosusceptible<br />

site, indicating that JNK1 positively regulates<br />

NF-κB expression and inflammation. Similarly,<br />

studies of transgenic NF-κB-luciferase mice revealed<br />

that NF-κB activity in ECs was enhanced<br />

at atherosusceptible compared to atheroprotected<br />

sites. To determine whether a causal relationship<br />

exists between shear stress and vascular inflammation<br />

we altered blood flow in the murine carotid<br />

artery by placing a flow altering cuff. We<br />

observed that low, oscillatory shear stress enhanced<br />

JNK activity, increased the expression of<br />

NF-κB and VCAM-1, promoted the accumulation<br />

of macrophages and enhanced arterial uptake of<br />

F18-FDG.<br />

Conclusions: We conclude that JNK1-ATF2 signalling<br />

promotes EC activation and inflammation<br />

at atherosusceptible sites exposed to low, oscillatory<br />

shear stress by enhancing NF-κB expression.<br />

Our findings illuminate a novel level of cross-talk<br />

between the NF-κB and JNK signalling pathways<br />

that may influence the spatial distribution of atherosclerotic<br />

lesions.<br />

Acknowledgement: Funded by the European Union<br />

FP6 NoE DiMI (LSHB-CT-2005-512146) and British<br />

Heart Foundation.<br />

References:<br />

1. Chaudhury et al. 2010 Arterioscler. Thromb. Vasc. Biol.<br />

30(3):546-53.


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

Absolute Quantification in Small Animal Pinhole Gated Myocardial Perfusion SPECT<br />

Goethals L. (1) , Devos H. (1) , Vanhove C. (1) , De Geeter F. (1) , Lahoutte T. (1) .<br />

(1) VUB Brussels, Belgium<br />

lode.goethals@vub.ac.be<br />

Introduction: Previously, our group has demonstrated<br />

the feasibility of absolute quantification using SPECT<br />

[1] imaging by incorporating a CT derived non-uniform<br />

attenuation correction (AC) map and a triple energy<br />

scatter window correction in the reconstruction<br />

algorithm of SPECT images. In this study we apply<br />

these techniques to the rat myocardium, which has a<br />

size below the resolution of our imaging system, thus<br />

suffering from a partial volume effect, which results in<br />

an underestimation of the activity concentration in the<br />

target tissues. To compensate for this underestimation<br />

we performed ultrasound imaging prior to SPECT imaging<br />

to determine myocardial wall thickness. We then<br />

derived a recovery coefficient (RC) from phantom studies<br />

to correct for the partial volume effect.<br />

Methods: SPECT/CT scans were performed in 9<br />

healthy Wistar rats 30 min after injection of 99mTc-<br />

Tetrofosmin. Prior to SPECT/CT imaging, 2D cardiac<br />

ultrasound images were acquired in the midventricular<br />

segments to determine diastolic and systolic<br />

myocardial wall thickness. After SPECT/CT imaging,<br />

animals were sacrificed and 6 midventricular segments(anterior-anterolateral-inferolateral-inferiorinferoseptal-anteroseptal)<br />

of the left ventricle were<br />

excised and counted in a gamma well counter. Using<br />

6 midventricular ROI’s in AMIDE, the effect of scatter<br />

correction (SC) and attenuation correction (AC) was<br />

determined in every midventricular segment.<br />

The activity in every ROI was expressed in Curie per<br />

gram (taking into account the density of myocardial tissue<br />

of 1,055 Kg/L). These image-derived activities were<br />

compared to the ex vivo activity concentrations<br />

counted in the gamma counter. To correct for<br />

the partial volume effect, a RC was determined from<br />

a phantom study. All values obtained after AC and SC<br />

were corrected by this RC and compared to the ex vivo<br />

counting data.<br />

Results: AC leads to a significant increase in counts in<br />

every midventricular segment, whilst SC results in a<br />

decrease in counts in every midventricular segment.<br />

The combination of both S and AC leads to a significant<br />

increase in counts in the inferior midventricular segment<br />

(paired Student’s T test, p < 0,05).<br />

After combined AC and SC, there remains a vast underestimation<br />

of activity in the myocardium, compared<br />

to the ex vivo data. Incorparation of a recovery<br />

coefficient derived from the phantom study, alleviates<br />

this problem: in the sub 5mm region, a RC of Y<br />

= 17,252 . X (with X = myocardial size in mm, Y = RC<br />

in %) was derived from the phantom studies. Dividing<br />

the combined AC and SC values by a RC derived<br />

from the average wall thickness (diastolic + systolic<br />

/ 2) leads to an adequate estimation of activity, correlating<br />

well with the ex vivo data. Use of the diastolic<br />

thickness as source of the RC leads to an overestimation<br />

of activity, use of the systolic thickness<br />

as a source of the RC leads to an underestimation<br />

of activity on the non-gated images. Use of the appropriate<br />

RC on the corresponding gated images also<br />

leads to an adequate estimation of activity on images.<br />

Conclusions: Combining attenuation, scatter and<br />

partial volume effect corrections in small animal<br />

myocardial perfusion SPECT allows for non invasive<br />

absolute quantification of myocardial perfusion.<br />

Acknowledgement: Researh at ICMI is funded by the<br />

Interuniversity Attraction Poles Program - Belgian<br />

State - Belgian Science Policy. Tony Lahoutte is a Senior<br />

Clinical Investigator of the Research Foundation - Flanders<br />

(Belgium) FWO. Lode Goethals is a PhD fellow of<br />

the research foundation – Flanders (Belgium) -FWO.<br />

References:<br />

1. Improved quantification in single-pinhole and<br />

multiple-pinhole SPECT using micro-CT information.<br />

Vanhove C, Defrise M, Bossuyt A, Lahoutte T. Eur J Nucl<br />

Med Mol Imaging. 2009 Jul;36(7):1049-63.<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

YIA applicant<br />

day3<br />

Parallel Session 9: CARDIOVASCULAR II


102<br />

WarSaW, poland May 26 – 29, 2010<br />

Cancer imaging<br />

Haberkorn U. .<br />

University Heidelberg, Germany<br />

Uwe.Haberkorn@med.uni-heidelberg.de<br />

Increased metabolism has been found to be one<br />

of the most prominent features of malignant<br />

tumors. This property led to the development of<br />

tracers for the assessment of glucose metabolism<br />

and amino acid transport and their application for<br />

tumor diagnosis and staging. Prominent examples<br />

are fluorodeoxyglucose, methionine and tyrosine<br />

analogs which have found broad clinical application.<br />

Since quantitative procedures are available these<br />

imaging life<br />

techniques can also be used for therapy monitoring.<br />

Another approach may be based on the noninvasive<br />

detection of apoptosis with tracers for<br />

phosphytidyl-serine presentation and/or caspase<br />

activation as surrogate marker for therapeutic<br />

efficacy. Finally, the use and development of<br />

specific ligands to target structures overexpressed<br />

in tumor tissue may be a valuable tool for diagnosis<br />

and novel therapeutic interventions.


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

Controlled drug delivery under image guidance<br />

Gruell H. (1) , Langereis S. (1) , De Smet M. (2) , Sanches P. (2) , Hijnen N. (2) , Rossin R. (1) , Bohmer M. (1) , Keupp J. (1) , Tiemann K. (3) ,<br />

Heijman E. (1) .<br />

(1) Philips Research,<br />

(2) Eindhoven University of Technology,<br />

(3) University Hospital Münster.<br />

holger.gruell@philips.com<br />

Introduction: Focused ultrasound (FU) is an excellent<br />

method for local triggered drug delivery<br />

using either pressure or temperature sensitive delivery<br />

systems. Temperature sensitive liposomes<br />

(TSLs) as drug carriers are promising to maximize<br />

delivery of drugs to tumors that are heated<br />

slightly above body temperature using FU. Besides<br />

heating, the pressure pulses of ultrasound<br />

waves can be exploited to disrupt microbubbles,<br />

which can lead to formation of transient pores in<br />

the endothelial wall. The latter is termed sonoporation<br />

and was shown to mediate cellular uptake<br />

of therapeutic agents. Either way, imaging based<br />

method to visualize, follow, and control drug delivery<br />

in-vivo are of importance to establish more<br />

quantitative treatment protocols.<br />

Methods: TSLs of different lipid compositions<br />

and a release temperature slightly above body<br />

temperature were loaded with T1, CEST or fluorinated<br />

MRI contrast agents and the drug doxorubicin.<br />

MRI was used to assess the signal change<br />

upon heating the TSLs from T=310 K to T=315 K<br />

using FU. For pressure induced drug delivery, microbubbles<br />

were injected into tumor bearing mice.<br />

Tumor and muscle tissues were treated with FU.<br />

Subsequently, radiolabeled albumin was injected<br />

and the extravasation in tumor and muscle tissue<br />

was followed and quantified using single photon<br />

computed tomography (SPECT).<br />

Results: Focused ultrasound is used to locally<br />

heat tissue, while MRI provides spatial and temperature<br />

control of the process. Co-encapsulation<br />

of drugs and MRI contrast agents allows to image<br />

the drug carrier system as well as to quantify<br />

release of drugs under hyperthermia conditions<br />

with MRI, making this modality the perfect<br />

choice for temperature induced drug delivery<br />

under image guidance. The temperature of drug<br />

release but also contrast properties can be tuned<br />

by nature and composition of the phospholipid<br />

bilayer. The rapid release of drugs and contrast<br />

agents can be explained by the formation of transient<br />

pores in the lipid membrane upon heating.<br />

For pressure induced drug delivery, the cavitation<br />

of microbubbles by ultrasound creates transient<br />

pores in the endothelial layer allowing macromolecules<br />

to efficiently extravasate. This effect<br />

is found to be more pronounced in muscle tissue<br />

than in tumors. In healthy muscle tissue the<br />

intact endothelial layer prevents extravasation of<br />

macromolecules while the leaky and premature<br />

vasculature in tumors is more permeable for macroscopic<br />

compounds.<br />

Conclusions: Focused Ultrasound allows to noninvasively<br />

trigger drug delivery in vivo, either using<br />

a local heating or focused pressure pulses in<br />

combination with suited drug delivery vehicles.<br />

The drug delivery process can be planned, controlled<br />

and followed using diagnostic imaging<br />

modalities such as MRI or ultrasound in combination<br />

with smart drug delivery vehicles. Above<br />

approach allows local non-invasive image guided<br />

treatment of a tissue that potentially leads to an<br />

improvement of the therapeutic window or even<br />

enables delivery of completely different classes of<br />

drugs such as siRNA or plasmid DNA.<br />

Acknowledgement: This project was funded in<br />

part by the EU Project Sonodrugs (NMP4-LA-<br />

2008-213706)<br />

References:<br />

1. Langereis et al., J. Am. Chem. Soc. 131, 1380 (2009).<br />

2. De Smet et al. J. Contr. Release 143, 120 (2010).<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

day3<br />

Parallel Session 10: CANCER II - together with COST


104<br />

WarSaW, poland May 26 – 29, 2010<br />

Comparative biodistribution of twelve gastrin/CCK2 receptor targeting peptides<br />

De Jong M. .<br />

Erasmus MC Rotterdam, The Netherlands<br />

m.hendriks-dejong@erasmusmc.nl<br />

Introduction: Molecular targeted radionuclide cancer<br />

therapy is becoming of increasing importance,<br />

especially for disseminated diseases. Systemic chemotherapies<br />

often lack selectivity; targeted radionuclide<br />

therapy has important advantages as the<br />

radioactive cytotoxic unit of the targeting vector is<br />

specifically directed to the cancer, sparing normal<br />

tissues. The basis of COST Action BM0607 is the<br />

great potential of targeted radionuclide therapy<br />

using a variety of vectors and radionuclides. This<br />

Action brings together the different disciplines involved<br />

and provides a reliable and rapid means for<br />

developing new (fundamental) knowledge, method<br />

standardization and products.<br />

Minigastrin and CCK analogs have been developed<br />

for radionuclide imaging and therapy of CCK2R<br />

expressing tumors. One project in COST Action<br />

BM0607 aims to select the optimal gastrin/CCK<br />

analogs for targeting MTC and SCLC expressing<br />

the CCK2R. In the present study, twelve DOTA-<br />

conjugated analogues were labeled with In-111<br />

and their in vitro and in vivo CCK2R targeting<br />

properties were studied in nude mice with CCK2R<br />

expressing A431 tumors.<br />

Methods: All peptides were labeled with In-111<br />

at high specific activity. Receptor affinity and internalization<br />

were tested in vitro using CCK2Rtransfected<br />

A431 tumor cells. Mice with CCK2Rtransfected<br />

A431 tumors in the left flank and with<br />

mock-transfected A431 tumors in the right flank<br />

received radiolabeled peptide intravenously for<br />

imaging and biodistribution studies.<br />

Results: All peptides demonstrated high receptor affinity<br />

and receptor-specific internalization in vitro<br />

and tumor targeting in the CCK2R expressing tumor<br />

in vivo. Tumor uptake at 1 h p.i. ranged from<br />

2.53 ± 0.47 %ID/g for CP07 to 13.3 ± 4.9 %ID/g for<br />

CP10, whereas uptake at 4 h p.i. ranged from 1.88 ±<br />

1.12 %ID/g to 9.90 ± 2.0 %ID/g, respectively. Uptake<br />

in non-target tissues was low for all peptides, except<br />

for high kidney uptake for CP10 and CP11. Highest<br />

tumor-to-kidney ratios (3-5) were obtained with<br />

the gastrin analogues CP01, CP02, CP03 and CP09.<br />

imaging life<br />

Conclusions: All peptides showed good CCK2Rmediated<br />

tumor-targeting in vitro and in vivo,<br />

with low uptake in non-target organs except for<br />

the kidneys in some cases. CP01, CP04 and CP05<br />

displayed favorable in vivo characteristics, combing<br />

high tumor uptake with low kidney retention.<br />

Acknowledgement: This study is part of COST Action<br />

BM0607


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

Targeting cancer stem cells using radiolabeled Sonic Hedgehog<br />

Tworowska I. (1) , Delpassand E. S. (1) , Sims-Mourtada J. (1) .<br />

(1) RadioMedix Housten, USA<br />

itworowska@radiomedix.com<br />

Introduction: Sonic Hedgehog (SHH) is an extracellular<br />

signalling protein involved in embryo<br />

development and morphogenesis [1]. Constitutive<br />

activation of the hedgehog pathway is observed in<br />

several types of cancers [2], especially in the most<br />

aggressive cancer stem cells and it is associated<br />

with their chemo/radiation resistance [3]. This<br />

work reports results on characterization of the SHH<br />

conjugates and their application to the cancer imaging<br />

using positron emission tomography (PET).<br />

Methods: 6xHis-tagged recombinant SHH<br />

(19.5kDa) with terminal NH2 group was conjugated<br />

to the DOTA (1,4,7,10-Tetraazacyclododecane-N,N’,N’’,N’’’-tetraacetic<br />

acid) in phosphate<br />

buffer and purified by dialysis. Alternatively<br />

SHH ligand was coupled to the maleimido-monoamide-DOTA<br />

chelator through thiol group of<br />

cysteine. Protein conjugates were further characterized<br />

using MALDI-MS and HPLC. Radiolabeling<br />

of the DOTA-SHH was performed in 0.1M<br />

NH4OAc (pH=4.4) using 68GaCl3 eluted from<br />

68Ge/68Ga generator (iThemba). In vitro bioactivity<br />

was evaluated by cellular uptake studies of<br />

the radiolabeled ligand in the several cancer cell<br />

lines and enriched stem cancer-cells population<br />

(mammospheres).<br />

Results: DOTA-SHH was obtained with total<br />

yield of 74%-89%. Reaction proceeded with radiochemical<br />

purity of >98% and specific activity<br />

of 3.38E+05 MBq/g, (theoretical specific activity<br />

= 3.64E+11 MBq/gram) as assessed by radio-TLC<br />

and radio-HPLC. 68Ga-DOTA-SHH have shown<br />

high affinity to Patched receptors (PTCH) in cancer<br />

cells (BT-474, MDA-MB-231, MCF-7) and its<br />

cellular uptake correlates with PTCH receptor expression<br />

levels. Cellular uptake of the SHH conjugates<br />

in the presence of the hedgehog pathway inhibitor,<br />

cyclopamine, was reduced by more than<br />

50%. Competitive uptake studies in the presence<br />

of the cold SHH, have also shown reduced cellular<br />

uptake by approximately 90%. In vitro uptake<br />

of 68Ga-DOTA-SHH was approximately 12-fold<br />

higher in cancer stem-cells (MCF-7 cultures)<br />

compared to the total cell population.<br />

Conclusions: Our studies have shown that 68Ga<br />

DOTA-SHH conjugates have potential application<br />

to non-invasive imaging of cancer, especially<br />

the most aggressive cancers. This is<br />

the first report on application of radiolabeled<br />

SHH conjugates for identification of the cancer<br />

stem-cell from the total tumor population.<br />

SHH conjugates may be also useful to evaluate<br />

new therapies targeting cancer stem-cells.<br />

References:<br />

1. Hammerschmidt M., Brook A., McMahon A.P., Trends<br />

Genet., (1997), 13, 14-2.<br />

2. Beachy P.A., Karhadkar S.S., Berman D.M., Nature (2004),<br />

432, 324-331; Mullor JL, Sánchez P, Altaba A.R., Trends<br />

Cell Biol.,(2003), 12 (12): 562–9.<br />

3. Al-Hajj M., Becker M.W., Wicha M., Weissmann I., Clarke<br />

M.F., Curr. Opin. Genet. Dev., (2004), 14(1), 43-47<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

day3<br />

Parallel Session 10: CANCER II - together with COST


YIA applicant<br />

106<br />

WarSaW, poland May 26 – 29, 2010<br />

Evaluation of the photosensitizer Bremachlorin for photodynamic treatment of breast<br />

cancer bone metastasis.<br />

Van Driel P. (1) , Que I. (1) , Snoeks T. (1) , Mol I. (1) , Xie B. (1) , Keereweer S. (1) , Kaijzel E. (1) , Löwik C.W.G.M. (1) .<br />

Leiden University Medical Center<br />

tpieter_v_driel@hotmail.com<br />

Introduction: Photodynamic therapy (PDT) is a<br />

promising treatment for tumors in which harmless<br />

light is used for the irradiation of nontoxic photosensitizers<br />

to create highly toxic reactive oxygen<br />

species for the destruction of certain tumors. In<br />

this study, the photosensitizer Bremachlorin TM<br />

(Brema Pharma Int.), derived from the seaweed<br />

spirulina, consisting of the compounds Chlorin e6,<br />

Purpurin 5 and Chlorin p6, was evaluated for PDT<br />

of breast cancer derived bone metastasis.<br />

Methods: The uptake of Bremachlorin in the human<br />

breast cancer cell line MDA-MB231 luc<br />

D3H2LN was quantitatively measured with flow<br />

cytometry and visualized with confocal laser scan<br />

microscopy. Cell viability after photodynamic<br />

treatment was measured with a luciferase assay<br />

and colorimetric MTS cell proliferation assay to<br />

identify the cytotoxic potential of Bremachlorin<br />

on these cells.The tumor cells were injected in the<br />

bone marrow cavity in femurs of immunodeficient<br />

mice to form osteolytic tumors. In vivo effects of<br />

PDT with Bremachlorin were followed by whole<br />

body bioluminescent imaging (BLI).<br />

Results: Flow cytometry analysis showed<br />

Bremachlorin uptake by the tumor cells was (close<br />

to) 100%. Also, fluorescent signal increased with<br />

an increasing dose of Bremachlorin. Complete<br />

cell death in vitro was already achieved after an<br />

incubation time of 5 hours with a concentration<br />

of 2 mg/l Bremachlorin. In vivo BLI experiments<br />

showed the partial or total eradication of the osteolytic<br />

tumors after a single pulse treatment of<br />

imaging life<br />

Bremachlorin-treated animals. In case of partial<br />

cure, the tumors were successfully eradicated by a<br />

second PDT treatment<br />

Conclusions: Our in vitro results show the tumor<br />

selective uptake of the photosensitizer Bremachlorin<br />

and its cytotoxic effects on the human<br />

breast cancer cells. Furthermore, with whole body<br />

BLI we show that PDT treatment of Bremachlorin-treated<br />

animals is an interesting therapy to<br />

eradicate aggressive tumors deep down in bone<br />

marrow. These results are very promising and<br />

interesting for future clinical use of photodynamic<br />

treatment of tumors with the photosensitizer<br />

Bremachlorin.<br />

Acknowledgement: Dhr. H. Vink and Andrei Reshetnickov<br />

from Brema Pharma


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

In vivo targeting of HEK-hsst 2/3/5 Xenografts by 111 in-labeled [(DOTA)Ser 1 ,Leu 8 ,trp 22 ,Tyr 25 ]-SS-<br />

28 in SCID mice<br />

Maina T. (1) , Tatsi A. (1) , Marsouvanidis I. P. (1) , Krenning E. P. (2) , De Jong M. (2) , Nock B.A. (1) .<br />

(1) IRRP, NCSR “Demokritos”,<br />

(2) Dept. of Nuclear Medicine, EMC Rotterdam.<br />

maina_thea@hotmail.com<br />

Introduction: Radiolabeled pansomatostatin-like<br />

analogs are expected to enhance the diagnostic<br />

sensitivity and to expand the clinical indications<br />

of currently applied sst 2 -specific radioligands<br />

(like OctreoScan ® ) [1,2]. We have previously reported<br />

on [(DOTA)Ser 1 ,Leu 8 ,trp 22 ,Tyr 25 ]SS-28<br />

displaying a high affinity for all five hsst 1-5 . In<br />

mice, the 111 In-radioligand showed specific uptake<br />

in AR4-2J tumors which express the rat sst 2<br />

[3]. We were further interested to investigate the<br />

ability of [( 111 In-DOTA)Ser 1 ,Leu 8 ,trp 22 ,Tyr 25 ]SS-<br />

28 to target human sst x expressing tumors in vivo.<br />

Methods: HEK-293 cells stably transfected with<br />

one of the hsst 2A , the hsst 3 or the hsst 5 were cultured<br />

in a humidified-5% CO 2 atmosphere at 37 0 C<br />

in DMEM GLUTAMAX-I supplemented with<br />

10%FBS, 100 U/mL penicillin / 100 µg/ml µg/mL<br />

streptomycin and 400 µg/mL G418. Cells were<br />

cropped, suspended in PBS and cell-suspension<br />

was equally divided in Eppendorf tubes (1 – 2 x<br />

10 7 cells, 150 mL). Inoculation was performed<br />

by subcutaneous injection of a cell suspension<br />

bolus in the left flank of young SCID mice. After<br />

10-18 days well-palpable tumor masses developed<br />

at the inoculation site and biodistribution<br />

experiments were performed. On the day of<br />

the experiment the radioligand was injected as<br />

a bolus (100 µL, 2 µCi) in the tail vein, alone or<br />

together with a high excess sst x -binding peptide<br />

(blocker). Animals were sacrificed in groups of<br />

four at 4 h postinjection (pi) and tissues were<br />

excised, weighed and counted in a gamma-counter.<br />

Values were calculated as percent injected<br />

dose per gram (%ID/g) tissue and are expressed<br />

as mean ± SD.<br />

Results: [( 111In-DOTA)Ser 1 ,Leu8 ,trp22 ,Tyr 25 ]SS-28<br />

showed significant and specific uptake in the hsst - 2A<br />

positive HEK implants of 4.43 ± 1.5%ID/g vs. 0.49 ±<br />

0.0%ID/g block (50 µg Tate) at 4 h pi (the previously<br />

+ reported values in the rsst -AR4-2J tumor, were<br />

2<br />

9.35 ± 1.31%ID/g and 0.35 ± 0.0%ID/g, respectively<br />

[3]). Equally high and specific uptake was displayed<br />

+ in the hsst -HEK xenografts with a 4.88 ± 1.1%ID/g<br />

3<br />

vs. 0.52 ± 0.05%ID/g block at 4 h pi (100 µg Demo-<br />

+ Pan 2 [2]). Preliminary values in the hsst -HEK<br />

5<br />

xenografts were slightly inferior (


poster


• MI in Cancer Biology – Visualisation of Extra- and Intracellular Processes 114<br />

Poster number P001 – P012<br />

• Imaging in Drug Development 126<br />

Poster number P013 – P022<br />

• Cancer from Bench to Bedside – Translational Research in Oncology 136<br />

Poster number P023 – P030<br />

• Imaging in Cardiovascular Disease: from Bench to Bedside 144<br />

Poster number P031 – P033<br />

• Molecular Neuroimaging: from Bench to Bedside 147<br />

Poster number P034 – P047<br />

• Imaging-guided Gene and Cell based Therapies 161<br />

Poster number P048 – P057<br />

• Probe Design – Innovative Approaches to Smart Contrast Agents 171<br />

Poster number P058 – P069<br />

• Technology – Technical Advances in MI Instrumentation 183<br />

Poster number P070 – P078<br />

• Imaging in Endocrine Diseases 192<br />

Poster number P079 – P083<br />

• MI of Infection and Inflammation 197<br />

Poster number P084 – P091<br />

• Imaging for Targeted Therapy 205<br />

Poster number P092 – P096<br />

• MI Data Analysis Methods 210<br />

Poster number P097 – P104<br />

• Late Breaking Abstracts 218<br />

Poster number P105 – P107<br />

Poster Session


110<br />

WarSaW, poland May 26 – 29, 2010<br />

Guided Poster Sessions – Poster Walks 1 to 7<br />

Guided Poster Session 1, Day 1: Thursday May 27, 2010 from 14:30 to 16:00<br />

Poster Walk 1: Imaging Cancer Biology – P001 to P012 & P105 to P106<br />

Co-Chairs: Fabian Kiessling (Aachen, Germany), Markus Rudin (Zürich, Switzerland)<br />

Poster Walk 2: Technology & Data Analysis Methods – P070 to P078 & P097 to P104<br />

Co-Chairs: Serge Maitrejean (Paris, France), Adriaan Lammertsma (Amsterdam, The Netherlands)<br />

Poster Walk 3: Molecular Neuroimaging – P034 to P047 & P107<br />

Co-Chairs: Sabina Pappata (Naples, Italy), Andreas Jacobs (Muenster, Germany)<br />

Poster Walk 4: Probe Design – P058 to P069<br />

Co-Chairs: Frédéric Dollé (Orsay, France), Helmut Maecke (Freiburg, Germany)<br />

Guided Poster Session 2, Day 2: Friday May 28, 2010 from 14:30 to 16:00<br />

Poster Walk 5: Imaging in Cancer & Drug Development – P013 to P022 & P023 to P030<br />

Co-Chairs: Peter Brader (Graz, Austria), NN<br />

Poster Walk 6: Imaging in Other Diseases – P031 to P033 & P079 to P083 & P084 to P091<br />

Co-Chairs: Bertrand Tavitian (Orsay, France), Nicolas Grenier (Bordeaux, France)<br />

Poster Walk 7: Imaging-guided Gene and Cell based and Targeted Therapies –<br />

P048 to P057 & P092 to P096<br />

Co-Chairs: Ludwig Aigner (Salzburg, Austria), NN<br />

imaging life


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

overview poster presentations – per title<br />

MI in CANCER BIOLGY<br />

In vivo MRI multicontrast kinetic analysis of intracellular trafficking of liposomes 114<br />

Diffusion-weighted MRI for differentiation of breast lesions at 3.0 Tesla: a new biomarker for breast imaging 115<br />

Imaging tumour apoptosis with 68Ga-labelled AnnexinA5 derivatives early after cancer therapy 116<br />

68 Ga-RGD-based PET tracers for imaging αν β 3 expression: a comparative study 117<br />

Influence of anaesthetics on tumor tracer uptake in radiopeptide receptor imaging (PRI) and radionuclide therapy (PRRT) 118<br />

Optical imaging of oral squamous cell carcinoma and cervical lymph node metastasis using near-infrared fluorescent<br />

probes in a mouse model – a pilot study 119<br />

Spectral unmixing of red and green luciferases for in vivo bioluminescence imaging 120<br />

High resolution redox imaging of intact cells by rxYFP, a ratiometric oxidation- sensitive fluorescent protein<br />

Assessment of antiangiogenic therapy effects in preclinical tumour models: Implementation of a novel contrast -<br />

121<br />

enhanced 3 D scanning technique and comparison to established ultrasound imaging protocols<br />

Influence of the BMP pathway on cell cycle regulation and its differentiation inducing potential on brain<br />

122<br />

tumor initiating cells 123<br />

A near infrared fluorescent-based method for imaging breast cancer induced osteolysis 124<br />

Near infrared fluorescent probes for whole body optical imaging of 4T1-luc2 mouse breast cancer development<br />

and metastasis 125<br />

IMAGING in DRUG DEVELOPMENT<br />

Improved animal models to study tumor growth and spontaneous metastases: bioluminescence imaging<br />

characterization of the HT-29 human colorectal cancer cell line<br />

Development of dorsal skin-fold window chamber for the analysis of blood vessel modifications induced by<br />

126<br />

electropermeabilization<br />

Characterization and evaluation of a tumor specific RGD optical probe for time-domain near-infrared<br />

127<br />

fluorescence imaging 128<br />

18F labeling of insulin via click chemistry<br />

Whole-body distribution, pharmacokinetics and dosimetry of radioiodinated fully humanized anti-VAP-1<br />

129<br />

antibody – a PET imaging study of rabbits 130<br />

Pharmacological characterization of iodine labeled adenosine kinase inhibitors<br />

Determining the nasal residence time of protein-polymer conjugates for nasal vaccination using a novel<br />

131<br />

imaging technique 132<br />

Nanotubes as mutli-modality vehicles for imaging and therapy of cancer 133<br />

Early assessment of temozolomide treatment efficacy in glioblastoma using [ 18F]FLT PET imaging 134<br />

Sensitive time-gated FRET microscopy of G-Protein coupled receptors using suicide enzymes, lanthanide cryptates and<br />

fluorescent ligands 135<br />

CANCER from BENCH to BEDSIDE<br />

Selection of a nanobody scaffold with low renal retention 136<br />

In vitro assessment of androgen mediated uptake of 18F-FDG, 11C-choline and 11C-acetate in prostate cancer 137<br />

Comparison of two 68Gallium-labeled octreotide analogues for molecular PET(CT) imaging of neuroendocrine tumours 138<br />

Real time per operative optical imaging for the improvement of tumour surgery in an in vivo micro-metastases model 139<br />

Fluorescence imaging modalities for imaging gastrointestinal tumor models<br />

Assessment of effectiveness and toxicity of the therapy with somatostatin analogue labelled 90Y-DOTATATE in<br />

140<br />

patients with non-functional pancreatic neuroendocrine tumours (PNT)<br />

Intra operative near-infrared fluorescent imaging of colorectal liver metastases using clinically available Indocyanine<br />

141<br />

Green in a syngene rat model 142<br />

Molecular imaging of resistance to EGFR tyrosine kinase inhibitors by 18F-FLT PET/CT and its reversal in non small cell<br />

lung cancer 143<br />

MI in CARDIOVASCULAR DISEASE<br />

Synchronised Cardiac and Lung CT in Rodents Using the Mobile CT Scanner LaTheta 144<br />

Molecular imaging of neurovascular inflammation in a mouse model of focal cerebral ischemia using Ultra small<br />

Superparamagnetic Particles of Iron Oxide (USPIOs) targeted to vascular cell adhesion molecule-1 (VCAM-1) 145<br />

Scintigraphy with the use of 123I-IL-2: a new promising tool for cardiovascular risk assessment in patients with high<br />

cardiovascular risk 146<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

poStEr


112<br />

WarSaW, poland May 26 – 29, 2010<br />

imaging life<br />

NEUROIMAGING from BENCH to BEDSIDE<br />

MEMRI-DTI study of focal transient ischemia in immature rat brain 147<br />

A clinically relevant model of in situ embolic stroke in the anesthetized monkey (macaca mulatta): long-term<br />

electrophysiological and mri analyses 148<br />

Automated radiosynthesis of [ 18F]MPPF derivatives for imaging 5-HT receptors 1A<br />

The vitamine E analogue CR6 protects against the long-term microstructure damage induced by MCA occlusion: a<br />

149<br />

longitudinal Diffusion Tensor Imaging study 150<br />

Design and synthesis of fluorescent probes for serotonin 5-HT1A receptors 151<br />

Comparative evaluation of cerebral blood flow in a rat model of cerebral ischemia using 15O-H 0 positron emission<br />

2<br />

tomography and 99mTc-HMPAO single-photon emission tomography<br />

Comparison of dopamine transporter density in Parkinson’s disease patients with and without autonomic dysfunction<br />

152<br />

using F-18 FP-CIT PET/CT 153<br />

Synthesis of tosylate and mesylate precursors for one-step radiosynthesis of [ 18F]FECNT 154<br />

Sex differences inDopamine D Receptor Occupancy in the Amygdala using AMPT with PET and [ 2 18F]fallypride 155<br />

Manual versus automatic delineation of VOIs for analysis of nuclear medicine images 156<br />

PET Amyloid and Tau Ligand [18F]FDDNP uptake in early Alzheimer disease<br />

Cryogenic brain injury as a model of brain trauma: Use of GFAP-luc mice to assess GFAP expression as an indication<br />

157<br />

of neural injury 158<br />

Plaque burden in the APPPS1 mouse picked up with diffusion kurtosis magnetic resonance imaging 159<br />

In vivo Imaging of Rat Glioma using the TSPO-ligand [ 18F]DPA-714 160<br />

GENE and CELL based THERAPIES<br />

A novel 19F MRI-based migration assay: application to primary human dendritic cells<br />

In vivo magnetic resonance imaging reveals altered migration of endogenous neural progenitor cells following<br />

161<br />

cuprizone-induced central nervous system demyelination 162<br />

Evolution pathways of compartmentalization and distribution of labeling iron-oxide particles in tumor tissue 163<br />

Acupuncture Works on Endorphins via Activating Stretch-Activated Cation Channels 164<br />

Labeling protocols for MRI and optical imaging of human muscle cells precursors 165<br />

Studying molecular processes in-vivo: A framework for quantifying variability in molecular MRI 166<br />

Clinically applicable cell tracking by MRI in cartilage repair using Superparamagnetic Iron Oxide (SPIO)<br />

Labeling of HUVEC with different iron oxide particles: An in vitro study about incorporation, distribution,<br />

167<br />

retention and toxicity<br />

Visualization of aberrant migration in the YAC 128 mouse model for Huntington’s disease by in situ labelling of<br />

168<br />

neural progenitor cells with iron oxide particles 169<br />

Optimization of in vitro radiolabeling of mesenchymal stem cells with 18F-fluorodeoxyglucose 170<br />

PROBE DESIGN<br />

Responsive MRI contrast agent for specific cell imaging of inhibitory, GABAergic neurons 171<br />

Novel ultrasound contrast agents for drug delivery 172<br />

Methods to study the interaction between aptamer probes and cell surface biomarkers 173<br />

MRI intelligent contrast agents as enzyme responsive nanosystems 174<br />

Standardization of molecular PBCA-microbubbles for routine use 175<br />

Novel Gd(III)-based probes for MR molecular imaging of matrix metalloproteinases 176<br />

Functionalization of nanoparticles for molecular imaging; a covalent approach<br />

Comparison of different chelating systems for the synthesis of Ga-68 labelled peptides for molecular imaging using<br />

177<br />

RGD-peptides as model compound 178<br />

GdDOTA-PIB: a potential MRI marker for Alzheimer’s disease<br />

A comparative study of the self-elimination of para-aminobenzyl alcohol and hemithioaminal-based linkers.<br />

179<br />

Application to the design of Caspase-3 sensitive pro-fluorescent probes 180<br />

Olefin Metathesis for the functionalization of superparamagnetic nanoparticles<br />

Pyridine-based lanthanide complexes : towards bimodal agents operating as near infrared luminescent and<br />

181<br />

MRI reporters 182<br />

TECHNOLOGY<br />

Delivery of multiple F-18 tracers from a single automated platform (FASTlab TM synthesizer) 183<br />

Monte-carlo modelling of a silicon detector insert combined with a PET scanner 184<br />

Autofluorescence corrected multispectral red-shifted fluorescent protein tomography 185


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

Acquiring the sample surface from co-registered FMT/MR measurements of a murine tumor model 186<br />

fDOT/ PET/CT imaging of biological processes in tumors 187<br />

Deep tissue molecular imaging with fluorescent biomarkers using multispectral optoacoustic tomography.<br />

A simulation study 188<br />

Microfluidic [ 11C]-carbonylation reactions for the rapid synthesis of radiolabelled compounds for PET 189<br />

Boosting image quality in low-dose RC-gated 5D cone-beam micro-CT 190<br />

Methodological approach using microscopy for quantitative evaluation of neo-angiogenesis and tumour<br />

progression in pre-clinical cancer models 191<br />

ENDOCRINE DISEASES<br />

Changes of heart stroke volume index established by 99mTc MIBI GSPECT scintigraphy after radioiodine treatment<br />

of patients with subclinical hyperthyroidism 192<br />

PET/CT investigations with 68Ga-DOATATE in neuroendocrine tumors - first clinical experience 193<br />

Influence of number of subsets and iterations of Ordered Subsets Expectation Maximization (OSEM 3D Flash)<br />

reconstruction on quantitative assessment of small and medium detected lesions in SPECT study with used in<br />

99mTc[EDDA/HYNIC]Octreotate for patients with GEP-NET 194<br />

The precise localization of metastatic lesion with used SPECT/CT with CoDe system after 131I – MIBG therapy in patients<br />

with disseminated pheochromocytoma 195<br />

Multimodal in vivo imaging of pancreatic beta-cells via antibody mediated targeting of beta-cell tumors 196<br />

INFECTION and INFLAMMATION<br />

6-[ 18F]Fluoro-PBR28, a novel TSPO 18 kDa radioligand for imaging neuroinflammation with PET 197<br />

Neuroinflammation is increased in the brain of ageing corpulent (JCR:LA-cp) rats: a positron emission tomography study 198<br />

Detection of inflammatory diseases by NIRF imaging with specific probes targeting leukotriene receptor CysLT R 1 199<br />

PET imaging of Hypoxia by 18F-Fluoromisonidazole ([ 18F]FMISO) to detect early stages of experimental arthritis<br />

Intracellular [<br />

200<br />

64Cu]PTSM and extracellular [ 64Cu]DOTA-antibody labelling of ovalbumin-specific Th1 cells for in vivo PET<br />

investigations of Th1 cell trafficking in OVA-specific lung inflammation 201<br />

[ 18F]DPA-714, [ 18F]PBR111 and [ 18F]FEDAA1106: Radiosyntheses on a TRACERLAb FX-FN synthesizer<br />

In vivo near-infrared fluorescence imaging of lung matrix metalloproteinases in an acute cigarette smoke-induced<br />

202<br />

airway inflammation model in different mice strains<br />

Development and pre-clinical evaluation of a novel class of<br />

203<br />

18F labelled PET ligands for evaluation of PBR/TSPO<br />

in the brain 204<br />

Local administration of adeno-associated virus into the mammary gland ductules 205<br />

TARGETED THERAPY<br />

Copper-64- and Gallium-68- NODAGA-conjugated bombesin antagonists as new PET tracers 206<br />

A novel indocyanine green nanoparticle probe for non invasive fluorescence imaging in vivo<br />

Evaluation and optimization of the concept of an antibody directed enzyme-prodrug therapy using noninvasive<br />

207<br />

imaging technologies 208<br />

A novel [ 18F] PET imaging agent for the epidermal growth factor receptor 209<br />

MI DATA ANALYSIS METHODS<br />

Structural methods in subcellular image analysis 210<br />

In vivo-post mortem multimodal image registration in a rat glioma model 211<br />

Feasibility and success of Independent Component Analysis of resting state fmri data from the rat 212<br />

A fast and robust acquisition scheme for CEST experiments<br />

Binding potential estimation in 11C-PE2I PET brain striatal images: impact of partial volume correction under<br />

213<br />

segmentation errors<br />

Automated quantification scheme based on an adapted probabilistic atlas based segmentation of the brain basal<br />

214<br />

nuclei using hierarchical structure-wise registration 215<br />

VHISTdiff - comparing workflow histories (VHIST/VINCI) 216<br />

Fast matrix-free method for fluorescence imaging 217<br />

Late Breaking<br />

PET and MRI studies applied on characterization of Fisher/F98 rat glioma model 218<br />

Non-invasive “E2F sensing” system for monitoring DNA damage alteration induced by BCNU<br />

Multi-tracer PET imaging of a mouse model of Alzheimer´s disease to assess microglial activation related to ageing<br />

219<br />

and anti-inflammatory treatment 220<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

poStEr<br />

Poster Session


P-001<br />

114<br />

WarSaW, poland May 26 – 29, 2010<br />

In vivo MRI multicontrast kinetic analysis of intracellular trafficking of liposomes<br />

Aime S. , Delli Castelli D. , Dastrù W. , Terreno E. , Cittadino E. , Torres E. , Mainini F. , Spadaro M. .<br />

University of Torino, Italy<br />

silvio.aime@unito.it<br />

Introduction: Liposomes are mainly used in the<br />

pharmaceutical field as drug delivery systems.<br />

Despite their large use, there is still a lack of<br />

information about the interaction between the<br />

nanovesicles and the cells in tumor environments<br />

and their intracellular fate after the cellular uptake.<br />

Most of the information on this topic comes out from<br />

in vitro cellular studies that are not always reliable<br />

models for mimicking the in vivo system. The aim<br />

of this work is the visualisation of the metabolic<br />

pathway of these vesicles directly in vivo by means<br />

of MRI. Since MRI does not provide enough spatial<br />

resolution to directly observe events at subcellular<br />

level, we have developed a multicontrast analysis<br />

that provides indirect evidence about the uptake<br />

and the intracellular trafficking of the nanovesicles.<br />

The method relies on the peculiarity of nanovesicles<br />

encapsulating paramagnetic Ln(III)-based complexes<br />

that may act as T1, T2, and CEST agents. In order to<br />

account for the observed MRI data, a kinetic model<br />

able to describe the underlying biological processes<br />

has been developed. The fit of the data provides a<br />

rough estimate of the kinetic constants for each<br />

process considered in the model.<br />

Methods: Non targeted, stealth or pH-sensitive,<br />

liposomes encapsulating paramagnetic<br />

lanthanide(III) complexes were prepared and in vitro<br />

characterized. The liposomes were locally injected in<br />

B16 melanoma tumor xenografted on C57 mice. The<br />

temporal evolution of T1, T2 and CEST MR contrast<br />

was followed at 7 T until 48 h post-injection.<br />

Results. The evolution over time is different among<br />

the three contrast modes (T1,T2, CEST). The process<br />

taking place (cellular uptake, intracellular release,<br />

esocytosis, and wash out) have been modeled and<br />

rough estimates for the kinetic constants have been<br />

determined upon the simultaneous interpolation of<br />

all the data. Moreover, the intracellular trafficking of<br />

stealth vs pH sensitive liposomes has been compared.<br />

The comparison among the evolution of the three<br />

contrast modalities for the two different liposomes<br />

is quite different. In particular, the maximum T1<br />

contrast was observed at 5hrs post injection while for<br />

the stealth one it occurs at 24hrs post injection.<br />

imaging life<br />

Conclusions: The MRI multicontrast analysis<br />

developed in this work represents an innovative way<br />

to get deeper insight into the in vivo detection of<br />

sub-cellular process. In particular, the intracellular<br />

trafficking of two liposomal formulations (pHsensitive<br />

and conventional stealth) of great relevance<br />

in the field of drug delivery have been compared. The<br />

kinetic analysis revealed that both liposomes are taken<br />

up quite fast from Tumor Associated Macrophages<br />

but the intracellular release of the imaging<br />

reporters is much faster for the pH sensitive one.


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

Diffusion-weighted MRI for differentiation of breast lesions at 3.0 Tesla: a new biomarker for<br />

breast imaging<br />

Bogner W. , Gruber S. , Pinker K. , Grabner G. , Moser E. , Trattnig S. , Stadelbauer A. , Helbich T. .<br />

Department of Radiology, Vienna, Austria<br />

thomas.helbich@akhwien.at<br />

Introduction: To compare the diagnostic quality of<br />

different diffusion weighting schemes with regard<br />

to apparent diffusion coefficient (ADC) accuracy,<br />

ADC precision, and diffusion-weighted image<br />

(DWI) contrast-to-noise ratio (CNR) for different<br />

types of lesions and breast tissue. Based on these<br />

assessments a new biomarker for breast imaging<br />

will be defined.<br />

Materials and Methods: Institutional Review Board<br />

approval and written, informed consent were<br />

obtained. Fifty-one patients with histopathologic<br />

correlation or follow-up were included in this study<br />

on a 3.0 Tesla MR scanner. There were 112 regions<br />

of interest (ROIs) drawn in 24 malignant, 17 benign,<br />

20 cystic, and 51 normal tissue regions. ADC maps<br />

were calculated for combinations of ten different<br />

diffusion-weightings (b-values), ranging from 0<br />

to 1250 s/mm². Differences in ADC among tissue<br />

types were evaluated. The CNR of lesions on DWI<br />

was compared for all b-values. A repeated measure<br />

ANOVA was used to assess lesion differentiation.<br />

Results: ADC (mean±SD x10-3mm²/s) values<br />

calculated from b=50 and 850 s/mm² were<br />

0.99±0.18, 1.47±0.21, 1.85±0.22, and 2.64±0.30<br />

for malignant, benign, normal, and cystic tissue,<br />

respectively. An ADC threshold of 1.25 x10-3mm²/s<br />

allowed discrimination of malignant from benign<br />

lesions with a diagnostic accuracy of 95% (p


P-003<br />

116<br />

WarSaW, poland May 26 – 29, 2010<br />

Imaging tumour apoptosis with 68Ga-labelled AnnexinA5 derivatives early after cancer<br />

therapy<br />

De Saint-Hubert M. (1) , Bauwens M. (1) , Devos E. (1) , Deckers N. (2) , Reutelingsperger C. (2) , Verbruggen A. (1) , Mortelmans L. (1) ,<br />

Mottaghy F. (3) .<br />

(1) KULeuven, Heverlee, Belgium<br />

(2) University Maastricht, The Netherlands<br />

(3) RWTH Aachen, Germany<br />

marijke.desainthubert@med.kuleuven.be<br />

Introduction: Molecular imaging of apoptosis offers<br />

a direct and early measurement of response to<br />

cancer therapy which allows a fast decision making<br />

in cancer treatment. One of the early characteristics<br />

of apoptosis is externalization of phosphatidylserine<br />

(PS) on cell membranes. Annexin V<br />

(AnxA5) binds with a high affinity to membranebound<br />

PS. Due to suboptimal imaging quality of<br />

99m Tc labelled AnxA5 we aimed to specifically label<br />

AnxA5 with an emerging radioisotope, 68 Ga, allowing<br />

Positron Emission Tomography (PET).<br />

Methods: We radiolabelled AnxA5 with 68 Ga using<br />

two mutated forms of AnxA5 with a single cysteine<br />

residue at position 2 or 165, respectively Cys2-<br />

AnxA5 and Cys165-AnxA5, allowing site-specific<br />

coupling of 68 Ga-Dota-maleimide. In vitro cell<br />

binding of both radiotracers was studied in healthy<br />

and anti-Fas treated Jurkat cells. Biodistribution<br />

and pharmacokinetics were studied with µPET in<br />

healthy mice and in a hepatic apoptosis model (anti-Fas<br />

Ab treated) up to 60 min p.i.. Daudi (Burkitt<br />

lymphoma) tumour bearing mice were scanned<br />

before and after treatment with combined chemotherapy<br />

(125 mg/g Endoxan) and radiotherapy (10<br />

Gy/tumour) using µPET and µMRI. Tracer uptake<br />

was measured and imaged ex vivo using autoradiography<br />

and correlated to histological evidence of<br />

apoptosis (TUNEL).<br />

Results: 68 Ga-Dota-maleimide labelling yield was at<br />

least 98% and coupling yield of 68 Ga-Dota-maleimide<br />

to Cys2-AnxA5 and Cys165-AnxA5 was ~70%.<br />

Labelling and purification took about 60 min, with<br />

a final radiochemical purity of at least 98%. In vitro<br />

binding of 68 Ga-Cys2-AnxA5 and 68 Ga-Cys165-<br />

AnxA5 to anti-Fas treated tumour cells was 5 times<br />

higher compared to normal cells.<br />

Dynamic PET images in normal mice revealed that<br />

both tracers showed a fast clearance from the blood<br />

towards the kidneys, with no significant change in<br />

biodistribution from 30 min p.i. on. Dissection data<br />

confirmed clearance was mainly via the urinary tract.<br />

imaging life<br />

Dynamic PET images of anti-Fas treated animals<br />

revealed a major shift of radioactivity from the<br />

kidneys to the (apoptotic) liver for both 68 Ga-<br />

Cys2-AnxA5 and 68 Ga-Cys165-AnxA5. Compared<br />

to normal mice anti-Fas treated animals showed a<br />

7 to 9 times higher liver uptake (for respectively<br />

68 Ga-Cys2-AnxA5 and 68 Ga-Cys165-AnxA5) as<br />

compared to healthy animals. Autoradiography<br />

images confirm the higher uptake in anti-Fas<br />

treated livers, corresponding to TUNEL positive<br />

cells.<br />

MRI-PET fusion images allowed clear delineation<br />

of each tumor. Using this technique, we noted that<br />

the post-therapy SUV values significantly exceeded<br />

those prior to therapy. The absolute tumour uptake<br />

of 68 Ga-Cys2-AnxA5 and 68 Ga-Cys165-AnxA5<br />

was respectively only 0.5 ± 0.1 % ID/g and 1.0 ±<br />

0.3 % ID/g and significantly increased to 1.5 ± 0.2<br />

% ID/g and 1.6 ± 0.1 % ID/g after therapy. Autoradiography<br />

confirms an increased activity in<br />

treated tumors with several regions showing 10-20<br />

times higher uptake compared to the tumor uptake<br />

prior to treatment while other regions were<br />

found relatively unaffected. The same heterogeneous<br />

distribution of apoptosis was observed with<br />

TUNEL stainings.<br />

Conclusions: 68 Ga-Cys2-AnxA5 and 68 Ga-<br />

Cys165-AnxA5 can be prepared with a high yield<br />

and within a reasonable time period. Apoptosis<br />

targeting capacity was clearly demonstrated in<br />

a model of hepatic apoptosis. PET-MRI fusion<br />

images demonstrated a higher tumor uptake after<br />

cancer therapy, indicating that 68 Ga-AnxA5<br />

may be useful for the early evaluation of tumor<br />

therapy.<br />

Acknowledgement: This work was financially<br />

supported by the European Union through the<br />

grant Euregional PACT II by the Interreg IV program<br />

of Grensregio Vlaanderen-Nederland (IVA-<br />

VLANED-1.20).


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

68 Ga-RGD-based PET tracers for imaging αν β 3 expression: a comparative study<br />

Dumont R. (1) , Maecke H. (1) , Haubner R. (2) , Behe M. (1) , Weber W. (1) , Fani M. (1) .<br />

(1) University Clinic Freiburg, Germany<br />

(2) Medical University Innsbruck, Austria<br />

rebecca.dumont@uniklinik-freiburg.de<br />

Introduction: The integrin α ν β 3 is a cellular adhesion<br />

molecule that is frequently expressed on<br />

malignant tumors and acts as an important receptor<br />

affecting tumor angiogenesis, local invasiveness,<br />

and metastatic potential. For imaging of the<br />

α ν β 3 integrin, cyclic pentapeptides containing the<br />

tripeptide sequence arginine-glycine-aspartate<br />

(RGD) have been developed. These peptides specifically<br />

bind to α ν β 3 in its activated state and recent<br />

clinical trials have shown that PET imaging<br />

with radiolabelled RGD peptides allows quantitative<br />

determination of activated α ν β 3 integrin in<br />

patients with various malignant tumors [1,2]. Development<br />

of a 68 Ga-RGD-based tracer would be<br />

of great clinical utility given the increasing clinical<br />

importance of oncologic PET imaging and the excellent<br />

imaging properties and availabilty of 68 Ga.<br />

Thus, we developed [ 68 Ga]-DOTA-c(RGDfK) [3]<br />

and most recently [ 68 Ga]-NODAGA-c(RGDfK)<br />

[4]. The aim of this study was to compare the<br />

tracer properties in a U87MG human glioblastoma<br />

xenograft model.<br />

Methods: The conjugate DOTA-c(RGDfK) was<br />

synthesised according to the literature [3]. An<br />

alternative head-to-tail cyclization protocol of<br />

the linear RGDfK was followed for the conjugate<br />

NODAGA-c(RGDfK) using a 50% solution<br />

of 1-propanephosphonic acid cyclic anhydride<br />

(T3P), triethylamine, and 4-di(methylamino)pyridine<br />

(DMAP). The two conjugates NODAGAc(RGDfK)<br />

and DOTA-c(RGDfK) were labelled<br />

with 68 Ga in sodium acetate buffer 0.2 N, pH 4.0<br />

using the Modular-Lab PharmaTracer module by<br />

Eckert & Ziegler. Biodistribution studies were performed<br />

in balb-c nude mice bearing subcutaneous<br />

U87MG human glioblastoma xenografts 1 h postinjection<br />

of the radiotracers (600 pmol / 4 MBq /<br />

mouse). To evaluate specificity of the compounds,<br />

blocking studies were done with an excess of the<br />

commercially available c(RGDfV). Static images<br />

were concurrently accquired with a microPET Focus<br />

small animal scanner and all results were evaluated<br />

comparatively.<br />

Results: Both conjugates were labelled with 68 Ga<br />

with radiochemical purity > 97% and specific activity<br />

of 8-10 MBq/nmol at room temperature<br />

(NODAGA-c(RGDfK)) or elevated temperatures<br />

(DOTA-c(RGDfK)). The biodistribution profile<br />

of the two radio-conjugates was similar. However,<br />

[ 68 Ga]-NODAGA-c(RGDfK) showed faster blood<br />

clearance and higher tumor uptake compared to<br />

[ 68 Ga]-DOTA-c(RGDfK) (5.2 ± 1.4 %IA/g vs 3.5 ±<br />

0.8 %IA/g) and improved tumor-to-blood and tumor-to-kidney<br />

ratios (27.7 ± 7.0 vs 9.2 ± 1.1 and 2.6<br />

± 0.3 vs 1.6 ± 0.1, respectively). Specificity of both<br />

tracers was demonstrated by successful blocking of<br />

tumor uptake in the presence of excess c(RGDfV),<br />

present on both biodistribtion and imaging studies.<br />

PET imaging quantitatively confirmed tumor targeting<br />

and biodistribution results of both conjugates.<br />

Conclusions: Compared to [ 68 Ga]-DOTA-c(RGDfK),<br />

the superior labelling conditions and in vivo profile<br />

of [ 68 Ga]-NODAGA-c(RGDfK) make this<br />

compound a compelling radiotracer candidate<br />

for use in PET imaging of α ν β 3 -expressing tumors.<br />

References:<br />

1. Haubner R et al., PLoS Med. 2:e70 (2005)<br />

2. Beer AJ et al., Clin Cancer Res. 12:3942-9 (2006)<br />

3. Decristoforo C et al., Eur J Nucl Med Mol Imaging.<br />

35:1507-1515 (2008)<br />

4. Knetsch P et al., J Label Compd Radiopharm. 52: S413<br />

(2009)<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

P-004<br />

poStEr<br />

MI in CANCER BIOLGY


P-005<br />

118<br />

WarSaW, poland May 26 – 29, 2010<br />

Influence of anaesthetics on tumor tracer uptake in radiopeptide receptor imaging (PRI) and<br />

radionuclide therapy (PRRT)<br />

Haeck J. , De Poel M. , Bijster-Marchand M. , Verwijnen S. , De Swart J. , Bernsen M. , De Jong M. .<br />

Erasmus Medical Centre, Oegstgeest, The Netherlands<br />

j.haeck@erasmusmc.nl<br />

Introduction: Neuro-endocrine tumors originating<br />

from various organs are known to over express somatostatin<br />

receptors (SSR) on the cell membrane (1).<br />

PRI and PRRT enable selective imaging and treatment<br />

of tumor cells over expressing SSR using radiolabelled<br />

somatostatin analogues, e.g. the SPECT<br />

tracer 111In-DTPA-octreotide. In experimental studies,<br />

in vivo imaging and measurement of tracer uptake<br />

in tumors and in normal organs is generally<br />

performed in anaesthetised laboratory animals. Different<br />

methods of anaesthesia are used according to<br />

the possibilities, experience and common practice<br />

in a laboratory. However, in many in vivo experiments<br />

the influence of changes in physiology due to<br />

anaesthetics is not taken into account. The aim of<br />

the current study was to determine the influence of<br />

different anaesthetics on bio-distribution of radioactive<br />

111In-DTPA-octreotide, a somatostatin analogue,<br />

5<br />

in rats bearing SSR positive tumors.<br />

Methods: Lewis rats (n=8) were inoculated with 0.5<br />

million CA20948 (SSR-positive) pancreatic tumor<br />

cells in the flank. The tumors were grown to approximately<br />

1.5 cm2 3<br />

2<br />

before imaging. Prior to i.v. injection<br />

of the radiolabelled peptide (50 MBq in 0.4 1 µg) the<br />

animals were anaesthetized with either inhalant-an-<br />

0<br />

aesthetic isoflurane (n=4) or a mixture of injectable<br />

anaesthetics consisting of sufentanil (300µg/kg) and<br />

medetomidine (300µg/kg) injected intra-peritoneally<br />

(n=4). The rats were kept under anaesthesia during 1h<br />

tracer circulation and subsequent SPECT/CT imaging.<br />

After imaging the rats were euthanized and tumor and<br />

organs were removed to study the peptide biodistribution<br />

ex-vivo. Biodistribution was performed at 3h p.i.<br />

Results: A significantly higher tumor uptake of<br />

111 In-DTPA-octreoscan was found when the rats<br />

were anaesthetized with isoflurane in comparison<br />

to medetomidine/sufentanil (figure 1), p


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

Optical imaging of oral squamous cell carcinoma and cervical lymph node metastasis using<br />

near-infrared fluorescent probes in a mouse model – a pilot study<br />

Keereweer S. (1) , Mol I. (2) , Snoeks T. (2) , Kerrebijn J. (1) , Van Driel P. (2) , Xie B. (2) , Kaijzel E. (2) , Baatenburg De Jong R. J. (1) ,<br />

Löwik C.W.G.M. (2) .<br />

(1) Erasmus Medical Center, Rotterdam, The Netherlands<br />

(2) Leiden University Medical Center, The Netherlands<br />

s.keereweer@erasmusmc.nl<br />

Introduction: In head and neck cancer surgery, intra-operative<br />

assessment of the tumor-free margin<br />

is critical to completely remove the tumor, thereby<br />

improving the prognosis of the patient. Currently,<br />

this mostly relies on visual appearance and palpation<br />

of the tumor. However, optical imaging techniques<br />

provide real-time visualization of the tumor,<br />

warranting intra-operative, image-guided<br />

surgery. The use of the near-infrared (NIR) light<br />

spectrum offers two additional advantages: increased<br />

tissue penetration of light and an increased<br />

signal-to-background-ratio of contrast agents. We<br />

performed a pilot study to assess the possibilities<br />

of optical imaging of oral squamous cell carcinoma<br />

and cervical lymph node metastasis using<br />

near-infrared fluorescent probes in a mouse model.<br />

A<br />

B<br />

Fig1. A. Bioluminescence image of oral carcinoma and cervical<br />

lymph node metastasis. B. Fluorescence image showing increased<br />

signal of Prosense680TM in the tongue and cervical lymph nodes.<br />

Methods: An oral tumor model was developed using<br />

luciferase-bearing OSC19 (human oral squamous<br />

cell carcinoma) cells that were injected directly into<br />

the tongue of 8 Balb/C nu/nu female mice. Tumor<br />

progression was followed by bioluminescence imaging<br />

(BLI) using the IVIS-100 (Caliper LS) and by<br />

inspection of the tongue. At day 21, different NIR<br />

fluorescent probes were systemically injected, targeting<br />

cathepsins (Prosense680TM, VisEn Medical),<br />

matrix metalloproteinases (MMPsense 680TM,<br />

VisEn Medical), increased glucose uptake (800CW<br />

2-DG, LI-COR) and increased epidermal growth<br />

factor expression (800CW EGF, LI-COR) of the tumor.<br />

Fluorescence imaging of the mouse and organs<br />

(after surgical removal) was performed using the<br />

Maestro (CRi) and Odyssey (LI-COR). A control<br />

group of 8 Balb/C nu/nu mice without tumor was<br />

used to compare light intensity between healthy and<br />

cancer tissue.<br />

Results: After 7 days, oral squamous cell carcinoma<br />

was established in all 8 mice. After 11 days, all<br />

mice had developed cervical lymph node metastasis,<br />

which was confirmed by a BLI signal (figure 1,A).<br />

The primary tumor and cervical lymph node metastases<br />

were successfully detected by the use of all NIR<br />

fluorescent probes, with increased tumor-to-control<br />

ratios that varied per probe (figure 1,B).<br />

Conclusions: This preliminary study shows the establishment<br />

of an oral squamous cell carcinoma<br />

mouse model, which progress could be followed by<br />

bioluminescence. Moreover, real-time visualization<br />

of the primary tumor and cervical lymph node metastases<br />

was achieved by fluorescence imaging using<br />

various NIR fluorescent probes. This technique<br />

can be used for intra-operative, image-guided surgery,<br />

which could improve complete removal of oral<br />

squamous cell carcinoma.<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

P-006<br />

poStEr<br />

MI in CANCER BIOLGY


WarSaW, poland May 26 – 29, 2010<br />

P-007 Spectral unmixing of red and green luciferases for in vivo bioluminescence imaging<br />

120<br />

Mezzanotte L. (1) , Kaijzel E. (2) , Michelini E. (1) , Que I. (2) , Calotti F. (2) , Hoeben R. (2) , Roda A. (1) , Löwik C. W.G.M. (2) .<br />

(1) University of Bologna, Bologna, Italy<br />

(2) Leiden University Medical Center, Leiden, The Netherlands<br />

laura.mezzanotte@libero.it<br />

Introduction: Amongst the numerous luciferase reporter<br />

genes cloned from different animal species<br />

and mutated to achieve different emission properties<br />

only a few have been employed for in vivo bioluminescence<br />

imaging (BLI). Good thermostability, high<br />

and stable photon emission and codon optimization<br />

of the luciferase gene are required (1,2). Moreover,<br />

spectral overlap prevents them from being measured<br />

simultaneously. Recently, spectral unmixing<br />

methodologies have been employed to unmix and<br />

quantify signals from images obtained from the<br />

collection of light emitted from proteins with different<br />

emission spectra (3,4). Here we investigated<br />

the combined use of green click beetle luciferase<br />

(CBG99, max. emission 537nm) and a red codonoptimized<br />

mutant of P.pyralis (Ppy-RE8, max emission<br />

618nm) for in vivo BLI purposes.<br />

Methods: Both in vitro and in vivo studies were carried<br />

out to analyse the different luciferases. Lentiviruses<br />

expressing Red Ppy-RE8 and Green CBG99<br />

luciferase reportergenes were generated and human<br />

embryonic kidney (Hek293) cells were subsequently<br />

Fig1.: CBG99 and PpyRE8 expressing cells were mixed in different<br />

ratios and measured in vitro (a) and in vivo at 540nm (c) and<br />

620nm (d).The graph (b) shows the max. emission peaks of the<br />

different luciferases.<br />

imaging life<br />

transduced to express the different variants of luciferase.<br />

Cell population of red and green emitting<br />

were mixed in different proportions and BLI measurements<br />

were carried out in a plate luminometer<br />

with appropriate filters.<br />

In addition, Hek293 cells co-expressing the mentioned<br />

luciferases were imaged using the IVIS Spectrum system<br />

(Caliper LS) by collecting BLI signals at different<br />

wavelengths. Finally, the cells expressing Ppy-RE8 or<br />

Green CBG99 were injected subcutaneously into immunodeficient<br />

mice and BLI imaging was performed<br />

in vivo using the same system. Signals from the two<br />

luciferases were unmixed using Living Image 3.2<br />

software.<br />

Results: Both in vitro as in vivo results demonstrated<br />

the feasibility to use Red Ppy-RE8 and Green CBG99<br />

luciferase reporter genes, simultaneously. These luciferase<br />

variants demonstrated to be a good couple for<br />

dual luciferase applications employing the same substrate<br />

luciferin. Using a pair of band pass filter (535nm<br />

and 628nm) contributions of red- and green-emitting<br />

luciferases can be calculated using a luminometer.<br />

Confirmative results were obtained using the Living<br />

image software to spectrally unmix the images composed<br />

by the signals of the two different luciferases and<br />

obtained from both in vitro and in vivo experiments.<br />

Conclusions: The possibilities of a combined use of<br />

the selected red and green luciferases have been demonstrated.<br />

Preliminary in vivo data envisage the future<br />

application of these couple of luciferases for monitoring<br />

of multiple events simultaneously by means of bioluminescence<br />

imaging.<br />

References:<br />

Branchini BR et al. Anal Biochem. 2010 Jan 15;396(2):290-7.<br />

Mezzanotte L, et al. Mol Imaging Biol. 2009,Nov 25.<br />

Gammon ST, et al. Anal Chem. 2006 Mar 1;78(5):1520-7.<br />

Michelini E, et al. Anal Chem. 2008 Jan 1;80(1):260-7.


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

High resolution redox imaging of intact cells by rxYFP, a ratiometric oxidation- sensitive<br />

fluorescent protein<br />

Pani G. , Maulucci G. , Mele M. , Labate V. , Panieri E. , De Spirito M. .<br />

Catholic University Medical School, Rome, Italy<br />

gpani@rm.unicatt.it<br />

Introduction: Plasmid encoded redox sensitive<br />

fluorescent probes promise to pave new avenues<br />

for realt time imaging of oxidative signaling in live<br />

cells. We have recently shown that rxYFP, a redoxsensitive<br />

variant of the Yellow Fluorescent Protein,<br />

can be used ratiometrically to construct high resolution<br />

redox maps of live cells. Confocal analysis of<br />

cell fluorescence at two excitation wavelenghts allows<br />

in fact to monitor, voxel by voxel, the distribution<br />

of the probe between its reduced and oxidised<br />

states, while normalizing fror probe concentration<br />

and photobleaching.<br />

Methods: rxYFP was transiently transfected in 293T<br />

human kidney carcinoma and B16F10 murine melanoma<br />

cells plated on glass bottom dishes (IbiDi).<br />

Cell fluorescence was analysed by confocal microscopy<br />

at two different excitation wavelengths (450<br />

and 488nm) and fluorescence ratios calculated by a<br />

dedicated software and converted into pseudocolors<br />

to construct redox-based cell maps.<br />

Results: By imaging cells based on rxYFP distribution<br />

between reduced and oxidised states, we were<br />

able to confirm that, in human and murine malignant<br />

cells, the nucleus is significantly more reduced than<br />

the cytosol; additionally, simple deconvolution of redox<br />

images constructed with untargeted rxYFPallows<br />

to identify hypereduced perinuclear spots that correspond<br />

to mitochondria, as further confirmed by the<br />

use of a mitochondrially targeted form of the probe.<br />

In 2D scans, the cell border of adherent cells consistently<br />

appears to be significantly more oxidised than<br />

the inner cytosol, while 3D cell reconstruction oxidation<br />

is polarized towards the bottom of the cells; finally,<br />

in a spontaneously migrating cell the leading edge<br />

appears to me more oxidised than the trailing one.<br />

Conclusions: This set of obserbations suggests, in<br />

keeping with our previous biochemical studies, that<br />

integrin signaling and cuytoskeleton rearrangement<br />

are associated with increased prooxidant activity, with<br />

relevant implications for cell invasion and metastasis.<br />

Acknowledgement: Work partially supported by DiMI<br />

FP6 European NoE DiMI (LSHB-CT-2005-512146)<br />

References:<br />

1. Maulucci G, Labate V, Mele M, Panieri E, Arcovito G,<br />

Galeotti T, Østergaard H, Winther JR, De Spirito M and<br />

Pani G High resolution imaging of redox signaling<br />

in live cells through an oxidation-sensitive yellow<br />

fluorescent protein Sci. Signal. 1, pl3 (2008).<br />

2. Maulucci G, Pani G, Labate V, Mele M, Panieri E, Papi M,<br />

Arcovito G, Galeotti T, De Spirito M. 2009 Investigation<br />

of the spatial distribution of glutathione redox-balance<br />

in live cells by using Fluorescence Ratio Imaging<br />

Microscopy. Biosens Bioelectron. 25:682-687.<br />

3. Maulucci G, Pani G, Fusco S, Papi M, Arcovito G, Galeotti<br />

T, Fraziano M, De Spirito M. 2009 Compartmentalization<br />

of the redox environment in PC-12 neuronal cells. Eur<br />

Biophys J. in press<br />

4. P. Chiarugi, G. Pani , E. Giannoni, L. Taddei, R. Colavitti,<br />

G. Raugei, M. Symons, S. Borrello, T. Galeotti, and G.<br />

Ramponi 2003 Reactive oxygen species as essential<br />

mediators of cell adhesion:The oxidative inhibition of a<br />

FAK tyrosine phosphatase is required for cell adhesion<br />

J. Cell. Biol. 161, 933-944<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

P-008<br />

poStEr<br />

MI in CANCER BIOLGY


122<br />

WarSaW, poland May 26 – 29, 2010<br />

P-009 Assessment of antiangiogenic therapy effects in preclinical tumour models: Implementation<br />

of a novel contrast - enhanced 3 D scanning technique and comparison to established<br />

ultrasound imaging protocols<br />

Rix A. , Lederle W. , Bzyl J. , Gaetjens J. , Fokong S. , Grouls C. , Kiessling F. , Palmowski M. .<br />

Helmholtz Institute for biomedical engineering , Aachen, Germany<br />

arix@ukaachen.de<br />

Introduction: Assessment of antiangiogenic therapy<br />

effects in rodent tumours plays an important role<br />

in drug development. Different ultrasound imaging<br />

protocols are described, predominantly using 2 D<br />

contrast-enhanced techniques. However, a limitation<br />

of 2 D scans is the reduced reproducibility of<br />

the data. Even if the acquired slice might be representative<br />

for the entire tumour, it will be impossible<br />

to find the identical slice position in longitudinal<br />

studies. In our study, we implemented a novel contrast-enhanced<br />

3 D scanning technique, evaluated<br />

its sensitivity and compared it to established 2 D<br />

and 3 D ultrasound imaging protocols.<br />

Methods: S.c epidermoid carcinoma xenografts in<br />

nude mice were scanned using a small animal ultrasound<br />

system (40 MHz). B-mode images with a<br />

low mechanical index and a slice thickness of 300<br />

µm were acquired and merged into a 3 D dataset.<br />

Prior to administration of the contrast agent, only<br />

one image was acquired per slice. During stable<br />

blood levels of polybutylcyanoacrylate-microbubbles,<br />

5 frames per slice were acquired (frame rate: 8<br />

Hz). The maximum intensity of each voxel per slice<br />

was recorded and compared to the pre-contrast<br />

dataset. To compare the potential role of this technique,<br />

a non contrast-enhanced 3 D Power Doppler<br />

scan (HF-VPDU: High frequency - volumetric<br />

power Doppler ultrasound [1]) and a 2 D dynamic<br />

contrast-enhanced scan (analyzed using the MIOT<br />

post-processing technique [2]) were performed.<br />

The sensitivity of all methods for assessing the antiangiogenic<br />

effects of SU11248 were examined at<br />

day 0, 1, 2 and 4 after treatment start. Tumour vascularisation<br />

was validated by determining the vessel<br />

density on corresponding histological sections.<br />

Mann-Whitney test was used for statistical analysis.<br />

Results: The novel 3 D contrast-enhanced imaging<br />

protocol could be applied to all animals successfully.<br />

The vascularisation of untreated control tumours<br />

slowly increased during the study, whereas<br />

a reduction was observed in tumours of treated<br />

animals. Differences in vascularisation between<br />

imaging life<br />

both groups were significant at day 1 (p


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

Influence of the BMP pathway on cell cycle regulation and its differentiation inducing<br />

potential on brain tumor initiating cells<br />

Rudan D. (1) , Monfared P. (1) , Viel T. (1) , Hadamitzky M. (1) , Euskirchen P. (1) , Knödgen E. (1) ,tSchneider G. (1) , Jacobs A.H. (1,2) .<br />

(1) Max Planck Institute for Neurological Research, Cologne, Germany<br />

(2) European Institute for Molecular Imaging, , Germany<br />

daniel.rudan@nf.mpg.de<br />

Introduction: Glioblastoma multiforme is the<br />

most common type of brain tumor, and several alterations<br />

of the cell cycle and DNA repair mechanisms<br />

have led to increasing resistance against<br />

chemotherapy. Latest findings suggest that a subset<br />

of tumor cells, the brain tumor initiating cells<br />

(BTIC) with stem cell like properties, are responsible<br />

for initiation and maintenance of the disease.<br />

Approximately 1-1000 out of 1.000.000 cells<br />

within a glioma is a BTIC. These cells are thought<br />

to bring about relapse and metastasis through<br />

their tumorigenic potential and chemotherapeutic<br />

resistance1,2. Design of a specific therapeutic<br />

strategy against BTICs may improve overall survival<br />

and quality of life for patients with gliomas.<br />

The aim is to examine a novel, experimental therapy<br />

against malignant gliomas by differentiating<br />

their BTIC population. Building on our previous<br />

work3, where we demonstrated that BMP-7 treatment<br />

decreases the proliferation of Gli36ΔEGFR-<br />

LITG glioma cells up to 50% via a cell cycle arrest<br />

in G1 phase, we will further characterize and<br />

compare the effect of the BMP-pathway on cell<br />

cycle regulation of different glioma cell lines and<br />

“glioma stem cells”.<br />

Methods: We want to determine (i) whether BMP<br />

treatment, alone or in combination with state-ofthe-art<br />

chemotherapeutic agents, could be applied<br />

in a disease-tailored therapy against gliomas by<br />

depleting their BTIC pool and (ii) whether our<br />

reporter construct (LITG) can quantify the responses<br />

to such a treatment in culture and in vivo.<br />

Glioma cell lines, primary glioma cell cultures<br />

and BTICs with different genetic profiles are being<br />

utilized to analyze the influence of BMP-7<br />

and BMP pathway inhibitors on cell cycle regulating<br />

protein expression, cell viability and caspase<br />

activation as well as its differentiation-induction<br />

potential in culture. BTIC will be transduced<br />

with lentiviral reporter vectors to image changes<br />

in cell cycle regulation, differentiation status<br />

and BMP pathway activity non-invasively in vivo<br />

upon treatment.<br />

Results: Western blot data indicates that BMP-7<br />

treatment causes an arrest in cell cycle progression<br />

in the established glioma cell line A172 by influencing<br />

the expression of the key cell cycle regulators<br />

(p53�, p21�E2F-1�). Furthermore we have shown<br />

the differentiation-induction potential of BMP-7 on<br />

BTICs in culture optically and on the protein level.<br />

Upon BMP-7 treatment and respective growth-factor<br />

withdrawal the expression of the neural stem cell<br />

marker Mshi1 is clearly downregulated. Currently<br />

we focus on the design of a lentiviral reporter vector<br />

for imaging the observed BMP-Pathway reponses.<br />

Conclusions: Antagonizing the proliferative potential<br />

of BTICs by targeting the BMP pathway, thereby<br />

triggering cellular differentiation of malignant stem<br />

cells, could provide a promising means of improving<br />

the prognosis for patients with gliomas.<br />

Acknowledgement: This work is supported in part by<br />

the FP6 European NoE DiMI (LSHB-CT-2005-512146).<br />

References:<br />

1. Singh SK, Hawkins C,Clarke ID, Squire JA, Bayani J, Hide<br />

T, Henkelman RM, Cusimano MD, Dirks PB, Nature 432,<br />

396 (2004)<br />

2. Piccirillo SG, Reynolds BA, Zanetti N, Lamorte G, Binda<br />

E, Broggi G, Brem H, Olivi A, Dimeco F, Vescovi AL,<br />

Bone morphogenetic proteins inhibit the tumorigenic<br />

potential of human brain tumour-initiating cells,<br />

Nature 444, 761 (2006)<br />

3. Klose A, Waerzeggers Y, Klein M, Monfared P, Vukicevic<br />

S, Kaijzel EL, Winkeler A, Löwik CW, Jacobs AH, Imaging<br />

bone morphogenetic protein induced cell cycle arrest<br />

in experimental gliomas, In Submission<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

P-010<br />

poStEr<br />

MI in CANCER BIOLGY


124<br />

WarSaW, poland May 26 – 29, 2010<br />

P-011 A near infrared fluorescent-based method for imaging breast cancer induced osteolysis<br />

Snoeks T. , Que I. , Mol I. , Kaijzel E. , Löwik C.W.G.M. .<br />

Leiden University Medical Center, The Netherlands<br />

t.j.a.snoeks@lumc.nl<br />

Introduction. In vivo bioluminescence imaging<br />

(BLI) of luciferase (Luc) expressing tumor cell lines<br />

has become a well accepted method to quantitatively<br />

follow tumor growth over time. Fluorescence<br />

imaging (FLI) can be used to visualize and quantify<br />

specific structures, molecules and even enzymatic<br />

activity using targeted dyes and enzyme activated<br />

smart probes. Two of such targeted fluorescent dyes<br />

are OsteoSense-680TM (VisEn Medical) and Bone-<br />

Tag-680TM (LI-COR Biosciences) both of which are<br />

directed specifically to bone.<br />

Traditionally, osteolytic lesions and subsequent bone<br />

loss are quantified using x-ray radiographs and μ-CT<br />

scans. These procedures are often time consuming<br />

and both methods possibly irradiate the animal limiting<br />

the number of repeated measurements.<br />

The aim of this study was to assess the possibility<br />

to quantify osteolysis by a loss of fluorescent signal<br />

after pre-labelling the skeleton with a bone-specific<br />

fluorescent probe. In this setup, we compared the<br />

performance of the two aforementioned commercially<br />

available bone specific fluorescent probes;<br />

OsteoSense-680TM and BoneTag-680TM.<br />

Fig1.: Decrease of fluorescence signal on osteolysis.<br />

Representative mouse labelled with BoneTag-680, imaged with<br />

the Pearl imager. A) Background FLI image before labelling<br />

with BoneTag-680. B-E) FLI images at day 2, 8, 19 and 42 after<br />

tumor cell inoculation in the right leg (*). F) Quantification of<br />

the fluorescent signal of the tumor bearing leg compared to<br />

the healthy leg.<br />

imaging life<br />

Methods. Immunodeficient mice received an intra<br />

venous injection with either OsteoSense-680 (2<br />

nmol, n=5) or BoneTag-680 (8 nmol, n=5). Three<br />

days after injection of the fluorescent probes the<br />

human breast cancer cell line MDA-BO2-Luc was<br />

inoculated in the femur bone marrow cavity of the<br />

right leg to form an osteolytic tumor. Tumor growth<br />

was followed by weekly BLI measurements using the<br />

IVIS Spectrum (Caliper LifeSciences) over a period<br />

of 6 weeks. Fluorescent data was obtained weekly using<br />

the IVIS Spectrum and at day 2, 8, 19 and 42 using<br />

the Pearl Imager (LI-COR Biosciences). In addition,<br />

μCT scans of each animal were made at day 21<br />

and 42 using the SkyScan-1076 μCT (SKYSCAN).<br />

Results. All inoculated mice developed a tumor in<br />

the right leg confirmed with BLI. Both bone specific<br />

probes remained detectable throughout the experiment,<br />

45 days after injection, in the healthy leg. The<br />

fluorescence signal decreased faster in the tumor<br />

bearing legs than in the healthy legs, this loss of signal<br />

could be quantified (Fig 1). The analysis of the<br />

x-ray and μCT data and the subsequent correlation<br />

between oslteolytic lesion size and the loss of fluorescence<br />

signal is still ongoing.<br />

Conclusions. Preliminary results show the possibility<br />

to follow osteolysis over time using an animal<br />

pre-labelled with a bone specific probe.<br />

Acknowledgement: Supported by the Dutch Cancer<br />

Society (Grant UL2007-3801)


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

Near infrared fluorescent probes for whole body optical imaging of 4T1-luc2 mouse breast<br />

cancer development and metastasis<br />

Xie B. , Snoeks T. , Mol I. , Van Driel P. , Keereweer S. , Kaijzel E. , Löwik C.W.G.M. .<br />

Leiden University Medical Center , The Netherlands<br />

b.xie@lumc.nl<br />

Introduction: The rapid development of molecular<br />

imaging has improved early diagnosis and treatments<br />

for various diseases including cancer. The<br />

imaging modalities vary broadly in their sensitivitie,<br />

and the high sensitivity of optical imaging, especially<br />

near infrared fluorescent (NIRF) imaging,<br />

makes it an excellent experimental tool for imaging<br />

small animals like mice. NIRF light has advantages<br />

such as considerably low tissue absorption coefficient<br />

in the NIR region (700-900 nm) and low tissue<br />

autofluorescence so that deeper light penetration<br />

can be achieved. These advantages make NIR light<br />

ideal for whole body optical imaging. Furthermore,<br />

NIRF probes are becoming available for clinical applications,<br />

e.g. imaging-guided surgery. In the current<br />

work, we have implanted mouse mammary<br />

gland cancer cell line 4T1-luc2 in nude mice, and<br />

then assessed 2 commercially available NIRF probes<br />

in their ability and specificity of detecting tumor<br />

progression and metastasis by whole body FLI in<br />

comparison with BLI.<br />

Methods: The following NIRF probes were tested<br />

and validated on detecting the mouse mammary<br />

gland cancer cell line 4T1-luc2 both in vitro and in<br />

vivo: Prosense680 TM (VisEn Medical) and 800CW<br />

Figure: Upper panel shows BLI and FLI Prosense680TM imaging.<br />

Lower panel shows BLI and FLI Prosense680TM imaging of LNs.<br />

2-DG(LI-COR Biosciences). In vitro, studies included<br />

cell-based fluorescent assay by Odyssey measurements,<br />

flow cytometry analysis, and visualization<br />

of probe uptake by confocal laser scan microscopy<br />

analysis. In vivo, 20.000 4T1-luc2 cells were implanted<br />

into the upper mammary fat pad (MFP) of<br />

immunodeficient mice. BLI was used as an internal<br />

control to evaluate the luciferase expression level in<br />

tumor areas, and also for co-locolization of the NIRF<br />

probes. After time-dependent BLI and FLI measurements,<br />

thoracic cavities of mice bearing tumors<br />

were surgically opened and reimaged to reveal the<br />

metastasis of surrounding tissues and axial lymph<br />

nodes (LNs). Organs were quickly removed, and ex<br />

vivo BLI and FLI were performed. Tissues with positive<br />

signals were further analyzed by histochemistry.<br />

Results: Both tested probes could successfully visualize<br />

4T1-luc2 mouse breast cancer cells, both in<br />

vitro and in vivo. Our in vivo data showed that there<br />

was already a nice increase in the BLI signal 3 days<br />

after implanting 4T1-luc2 cells in the MFP of nude<br />

mice. We could also detect the tumor progression<br />

using the NIRF probes in intact animals. At the end<br />

of the experiment (18 days) the animals were surgically<br />

opened up, and we could clearly detect lung and<br />

axial LNs metastases, both by BLI and FLI. This was<br />

confirmed by ex vivo imaging and histochemistry.<br />

Conclusions: This preliminary study of 4T1-luc2<br />

mouse breast cancer indicates that the development<br />

and metastasis of cancer can not only be detected<br />

by BLI but also by FLI, using commercially available<br />

NIRF probes, especially after surgically opening<br />

up the animals. This should pave the way for realtime<br />

visualization of tumor tissues during operation,<br />

making radically removal of all tumor tissues and<br />

local metastases possible.<br />

Acknowledgement: This study is supported by the<br />

Dutch CTMM Project MUSIS<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

P-012<br />

poStEr<br />

MI in CANCER BIOLGY


126<br />

WarSaW, poland May 26 – 29, 2010<br />

P-013 Improved animal models to study tumor growth and spontaneous metastases:<br />

bioluminescence imaging characterization of the HT-29 human colorectal cancer cell line<br />

Fernández Y. , Miranda S. , López M.E. , Suárez L. , Céspedes M.V. , Mangues R. , Herance J. R. , Rojas S. , Abasolo I. ,<br />

Schwartz Jr S. .<br />

CIBBIM-Nanomedicina. Edifici Hospital General. Bioquímica, Barcelona, Spain<br />

yofernan@ir.vhebron.net<br />

Introduction: The main problem in the treatment of<br />

colorectal cancer (CRC) is not so much the eradication<br />

of the primary tumor, but rather the formation<br />

of incurable metastases. To improve survival of<br />

these patients, there is an urgent need for new treatment<br />

strategies. For this purpose, the development<br />

of reliable, reproducible, clinically-relevant and<br />

inexpensive mouse models that fulfill tumor progression,<br />

invasion and metastasis of human CRC is<br />

needed. A new era of modeling cancer metastasis<br />

involves the use of optical imaging technologies to<br />

monitor tumor growth and colonization after introduction<br />

of cancer cells into the animals. The purpose<br />

of this study was to characterize the behavior<br />

of the human colon cancer cell line HT-29 in a subcutaneous,<br />

experimental metastases and orthotopic<br />

mouse models using non-invasive bioluminescence<br />

imaging (BLI) technologies, thereby providing an<br />

additional method to study CRC disease.<br />

Methods: Luciferase expressing HT-29 cells were injected<br />

subcutaneously, into the left ventricle and orthotopically<br />

into the cecal wall of immunodeficient<br />

nude mice. The tumor growth and metastatic dissemination<br />

patterns were quantitatively and continuously<br />

followed up via BLI. Non-invasive monitoring<br />

of tumorigenicity and metastasis was also compared<br />

to the traditional assays of tumor volume, weight or<br />

histology. Ex vivo BLI and histological analyses were<br />

performed to further identify the exact nature and location<br />

of lesions. BLI results were also validated using<br />

the positron emission tomography (PET) imaging.<br />

Results: We show that BLI has been successfully<br />

used to monitor colorectal tumor growth and metastases<br />

in vivo, and even helps to identify new<br />

metastatic sites. In the subcutaneous mouse model,<br />

BLI production and traditional tumor volume<br />

measurements show a strong correlation, demonstrating<br />

that BLI is an appropriate method to noninvasively<br />

quantify tumor burden. The left cardiac<br />

ventricle injection resulted in colonic tumor<br />

colonies in most organs, including the skeletal<br />

system of mice. In orthotopic implants, locoregional<br />

tumor growth and distant metastases occur<br />

spontaneously and rapidly, closely resembling<br />

imaging life<br />

clinical human disease progression that includes<br />

lymphatic, hematogenous and celomic dissemination.<br />

Ex vivo BLI confirmed the localization of<br />

metastases and dictated which tissues were going<br />

to be analyzed by histopathology reducing the<br />

number of histology rounds required to eventually<br />

detect all the small micrometastases identified<br />

by BLI.<br />

Conclusions: Our findings show that BLI improves<br />

upon and refines traditional animal cancer<br />

models by using fewer animals, offering a rapid,<br />

sensitive and less invasive monitoring of early<br />

neoplastic growth and metastases. Moreover, it<br />

provides an accurate and temporal assessment in<br />

the same animal over time increasing the statistical<br />

power of the model. In conclusion, BLI is a<br />

powerful tool for high-throughput longitudinal<br />

monitoring of tumor load in small animals and<br />

allows the implementation of more advanced orthotopic<br />

tumor models in therapy intervention<br />

studies with almost the same simplicity as when<br />

measuring traditional ectopic models. The availability<br />

of these bioluminescent models are powerful<br />

and reliable tools with which to investigate<br />

metastatic human CRC and novel therapeutic<br />

strategies directed against it.<br />

Acknowledgement: We are grateful to the Spanish<br />

Ministry of Science and Innovation for supporting<br />

laboratory technician and pre-doctoral personal,<br />

and CIBER-BBN for funding the project.<br />

References:<br />

1. Taketo, M. M. & Edelmann, W.; Gastroenterology 136,<br />

780-98 (2009)<br />

2. Cespedes, M. V.; et al. Am J Pathol 170, 1077-85 (2007)<br />

3. Weissleder, R. & Pittet, M.; J. Nature 452, 580-9 (2008)<br />

4. O’Neill, et al.; J Pathol 220, 317-327 (2009)


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

Development of dorsal skin-fold window chamber for the analysis of blood vessel<br />

modifications induced by electropermeabilization<br />

Golzio M. (1) , Bellard E. (1) , Markelc B. (2) , Cemazar M. (2) , Sersa G. (2) , Teissie J. (1) .<br />

(1) IPBS-CNRS, Toulouse, France<br />

(2) Institute of Oncology, Slovenia<br />

muriel.golzio@ipbs.fr<br />

Introduction: Recent developments in intravital microscopy<br />

(IVM) enable studies of tumour angiogenesis<br />

and microenvironment at the cellular level after<br />

different therapies. Preparation of skin fold chamber<br />

enables to follow fluorescent events on live animal.<br />

Electroporation/electropermeabilization, i.e. application<br />

of electric pulses to tissues, is a physical method<br />

for delivery of exogenous molecules. It is already used<br />

in clinical therapies of cancer, for electrochemotherapy<br />

of tumors (ECT). Its use was recently developed<br />

in electrogene therapy (EGT). In vivo, “electroporation”<br />

is associated with a blood -flow modifying effect<br />

resulting in decreased blood flow.<br />

The aim of our study was to observe directly on the<br />

living animal the effects of “electropermeabilization”<br />

on subcutaneous normal blood vessels by monitoring<br />

changes in morphology (diameter) and dynamics<br />

(vasomotricity, permeability and recovery).<br />

Methods: These parameters were measured using<br />

fluorescently labelled dextrans injected in the<br />

blood vessels observed via a dorsal skin fold window<br />

chamber, intravital digitized stereomicroscope,<br />

in vivo intravital biphoton microscopy and custom<br />

image analysis. A mathematical modelling gave access<br />

to the changes in permeability from the time<br />

lapse observation. Delivery of electric pulses was<br />

operated on the microscope stage directly on the<br />

animal under anaesthesia.<br />

Results: It resulted in immediate constriction of<br />

blood vessels that was more pronounced for arterioles<br />

(up to ~65%) compared to venules (up to ~20%).<br />

A rapid increase in vascular permeability was present<br />

that gradually decreased to basal (control) levels at<br />

1 h post-treatment. The decay of the high increase<br />

in vascular permeability was biphasic with an initial<br />

fast decrease, but was still present at 1h post-treatment.<br />

Furthermore, vasoconstriction of arterioles after<br />

“electropermeabilization” resulted in a “vascular<br />

lock” that remained for at least 6 minutes. This correlated<br />

approximately with the duration of decreased<br />

diameters of arterioles that lasted for 8 minutes.<br />

Conclusions: the results of our study provided<br />

direct in vivo monitoring of a vascular effect of<br />

electric pulses on normal vessels. The observed<br />

increase in permeability of vessels associated<br />

with delayed perfusion induced by electric pulses<br />

explains the improved delivery of molecules into<br />

tissues induced by this method after systemic<br />

delivery.<br />

Acknowledgement: CNRS, Region Midi Pyrenees,<br />

ARC, canceropole GSO, ANR “Cemirbio”, Slovenian<br />

French Proteus<br />

References:<br />

1. Cemažar M, Golzio M, et al. Current Pharmaceutical<br />

Design 12: 3817-3825. (2006).<br />

2. Marty M, et al. EJC 4(11): 3-13. (2006).<br />

3. Sersa G, et al. Br J Cancer; 98: 388-398 (2008).<br />

4. Golzio M., et al. Gene Therapy 11, S85-S91 (2004).<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

P-014<br />

poStEr<br />

IMAGING in DRUG DEVELOPMENT


128<br />

WarSaW, poland May 26 – 29, 2010<br />

P-015 Characterization and evaluation of a tumor specific RGD optical probe for time-domain<br />

near-infrared fluorescence imaging<br />

Mathejczyk J.E. (1) , Resch-Genger U. (2) , Pauli J. (2) , Dullin C. (3) , Napp J. (1) , Tietze L. F. (4) , Kessler H. (5) , Alves F. (1) .<br />

(1) Max-Planck Institute for Experimental Medicine, Göttingen, Germany<br />

(2) BAM Federal Institute for Materials Research and Testing, , Germany<br />

(3) University Medical Center Göttingen, Germany<br />

(4) University of Göttingen, Germany<br />

(5) Technical University of Munich, Germany<br />

jmathej@gwdg.de<br />

Introduction: RGD peptides provide useful tools<br />

for specific targeting of tumors overexpressing<br />

α ν β 3 integrins. We used a cyclic RGDfK peptide,<br />

coupled to the near-infrared fluorophore, Cy5.5,<br />

via an aminohexaonic spacer (RGD-Cy5.5) for<br />

functional imaging of α ν β 3 integrins on tumors in<br />

vivo. To assess the suitability and the achievable<br />

sensitivity of RGD-Cy5.5 for tumor imaging<br />

we characterized the spectroscopic properties<br />

of the optical probe and applied time-domain<br />

near-infrared fluorescence (NIRF) imaging for<br />

noninvasive and specific tumor targeting in vivo.<br />

Methods: Application-relevant properties like the<br />

fluorescence quantum yield, lifetime and the thermal<br />

stability of the cyclic RGDfK peptide coupled<br />

to Cy5.5 via an aminohexanoic acid spacer and its<br />

parent fluorophore, Cy5.5 were analyzed spectroscopically.<br />

For in vivo imaging, human α ν β 3 integrin-expressing<br />

glioblastoma cells, U87MG, were<br />

subcutaneously implanted into nude mice. Imaging<br />

was performed using the time-domain fluorescence<br />

imager, Optix MX2 (ART, Montreal, Canada).<br />

Results: RGD-Cy5.5 shows excellent spectroscopic<br />

properties making it a useful tool for in vivo NIRF<br />

imaging. Remarkable is the enhancement in fluorescence<br />

quantum yield of Cy5.5 in RGD-Cy5.5 increasing<br />

from 0.29 to 0.34. In vivo, the specificity<br />

of the RGD-Cy5.5-derived signals was confirmed<br />

by fluorescence lifetime measurements which were<br />

used to distinguish the probe signals from unspecific<br />

fluorescence. RGD-Cy5.5 binds selectively to<br />

glioblastoma with a maximum fluorescence intensity<br />

resulting 5 h after probe injection. The binding<br />

specificity was further confirmed by reduction<br />

of this fluorescence after application of an excess<br />

of unlabeled RGD peptides.<br />

Conclusions: Knowledge of the spectroscopic properties<br />

of fluorescent conjugates represents an important<br />

prerequisite for the successful application<br />

and sensitive detection of fluorescent probes in vivo.<br />

With RGD-Cy5.5, we developed a strongly emissive<br />

and stable optical probe for targeting α ν β 3 integrin<br />

receptors. Fluorescence lifetime measurements<br />

imaging life<br />

contributed to a further enhancement in detection<br />

sensitivity as compared to conventional steady state<br />

NIRF imaging in the intensity domain. Since the<br />

RGD probe contains an aminohexanoic acid spacer,<br />

that allows an easy and effective coupling with<br />

anti-cancer agents, this probe might be applied in<br />

oncology for therapeutic and diagnostic purposes.


18 F labeling of insulin via click chemistry<br />

<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

Paris J. , Mercier F. , Thonon D. , Kaisin G. , Lemaire C. , Goblet D. , Luxen A. .<br />

University of Liege, Belgium<br />

J.Paris@ulg.ac.be<br />

Introduction: As a positron emission tomography<br />

probe, a new 18 F-bearing insulin derivative was prepared<br />

by an original labeling method. This tracer was<br />

required as a radiolabelled active principle model<br />

compound to perform biodistribution imaging studies<br />

of new pulmonary administrable formulations.<br />

Methods: A 1,3-dipolar azide/alkyne cycloaddition<br />

(click chemistry) approach was developed for the<br />

mild and efficient linking of the radioactive probe<br />

onto insulin (Fig1).<br />

Gly A1 Insulin<br />

H2N CO2H H2N Phe B1<br />

NH 2<br />

As an initial step, native insulin had to be derivatized<br />

in order to present an accessible alkyne<br />

group at an appropriate position. For this, two of<br />

the three amino functions available on the molecule<br />

(Glycine A1 and Lysine B29) were first protected<br />

as N-Boc derivatives. An alkyne bearing<br />

prosthetic group could then be selectively grafted<br />

on the phenylanine B1 residue which does not<br />

interfere in the insulin-receptor binding process<br />

[1]. The final [ 18 F] fluorine insertion step was carried<br />

out by reacting this insulin derivative with<br />

the radioactive azide synthon 1-(azidomethyl)-<br />

4-[ 18 F]-fluorobenzene[2]under Cu(I) catalysis<br />

conditions.<br />

Results: The clickable 18 F azide 1-(azidomethyl)-4-<br />

-[ 18 F]-fluorobenzene was obtained in 65 minutes<br />

with good radiochemical yield (40-45% decay-corrected)<br />

and radiochemical purity (>90%) thanks to<br />

a fully automated preparation method developped<br />

on remote-controlled commercial radiosynthesis<br />

CO 2H<br />

BocHN CO 2H<br />

H 2N<br />

Boc 2 O<br />

Lys B29<br />

CO2H NHBoc<br />

O<br />

O<br />

O<br />

N<br />

O<br />

BocHN CO 2H<br />

HN<br />

O<br />

unit. Subsequent copper catalyzed click reaction on<br />

the alkyne bearing insulin derivative was performed<br />

at room temperature in less than 20 minutes with<br />

current radiochemical yields of 70-80% (decaycorrected).<br />

Conclusions: A mild and efficient radiolabelling<br />

strategy of insulin was succesfully developped using<br />

click chemistry. This radiotracer can now be incorporated<br />

in new inhalable pharmaceutical formulations<br />

for biodistribution imaging studies.<br />

CO2H NHBoc<br />

Acknowledgement: NeoFor and Keymarker research<br />

platforms from the BioWin projects of the Walloon<br />

Region are acknowledged for their financial support.<br />

References:<br />

Click<br />

[ 18 F]AzidoFB<br />

18F + TFA (deprotection)<br />

1. Guenther K.J. ; Yoganathan, S.; Garofalo, R.; Kawabata,<br />

T.; Strack, T.; Labiris, R.; Dolovich, M.; Chirakal, R. and<br />

Valliant, J.F., J.Med.Chem., 49: 1466-1474 (2006)<br />

2. Thonon, D. ; Kech, C. ; Paris, J. ; Lemaire, C. and Luxen,<br />

A.; Bioconjugate Chem., 20(4):817-823, (2009)<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

18 F<br />

N 3<br />

N<br />

N N<br />

HN<br />

O<br />

Insulin<br />

Fig1<br />

P-016<br />

poStEr<br />

IMAGING in DRUG DEVELOPMENT


130<br />

WarSaW, poland May 26 – 29, 2010<br />

P-017 Whole-body distribution, pharmacokinetics and dosimetry of radioiodinated fully<br />

humanized anti-VAP-1 antibody – a PET imaging study of rabbits<br />

Roivainen A. (1) , Autio A. (2) , Suilamo S. (2) , Mali A. (2) , Vainio J. (2) , Saanijoki T. (2) , Oikonen V. (2) , Luoto P. (2) , Teräs M. (2) , Karhi T. (2) ,<br />

Vainio P. (2) .<br />

(1) Academy of Finland/University of Turku, Finland<br />

(2) Turku PET Centre, Finland<br />

anne.roivainen@utu.fi<br />

Introduction: Vascular adhesion protein-1 (VAP-1)<br />

is an inflammation inducible endothelial glycoprotein<br />

[1]. It plays a key role in leukocyte trafficking<br />

and is a potential target for anti-inflammatory therapy.<br />

The antibody BTT-1023 is first-in-class, fully<br />

human monoclonal antibody to VAP-1. BTT-1023<br />

is potentially useful for the treatment of inflammatory<br />

diseases and in vivo imaging of inflammation.<br />

Positron emission tomography (PET) is powerful,<br />

non-invasive method particularly suitable for drug<br />

development because of its high sensitivity and ability<br />

to provide quantitative and kinetic data without<br />

sacrificing the animal. We assessed the usefulness<br />

of PET in evaluation of iodine-124 labeled antibody<br />

pharmacokinetics and distribution in rabbits.<br />

Methods: BTT-1023 was labeled with [ 124 I] using<br />

Chloramine-T method. Immunoreactivity of [ 124 I]<br />

BTT-1023 was verified in human VAP-1 transfected<br />

Chinese Hamster Ovary cells and by time-resolved<br />

immunofluorometric assay using human recombinant<br />

VAP-1. Ten rabbits were intravenously injected<br />

with [ 124 I]BTT-1023. PET/CT was obtained over the<br />

first 2 h after dosing and at 24, 48 and 72 h postinjection.<br />

Blood samples were collected during PET<br />

study to clarify in vivo stability and pharmacokinetics.<br />

As a final point, the human radiation dose estimates<br />

were extrapolated from rabbit information.<br />

Results: Binding of [ 124 I]BTT-1023 to hVAP-1<br />

transfected cells was app. 600-fold higher than<br />

in mock transfected controls. In rabbits, the radioactivity<br />

was distributed especially to liver and<br />

thyroid. Also heart and lungs showed some uptake<br />

whereas brain uptake was very low. Liver uptake<br />

is likely mediated, at least in a large part, by<br />

VAP-1, since the antigen is found on sinusoidal<br />

endothelia in the liver. Thyroid gland radioactivity<br />

is due to the de-iodination of [ 124 I] from the<br />

antibody. The plasma half-life of [ 124 I]BTT-1023<br />

was 58.3 h and clearance 7.8 mL/h/kg. [ 124 I]BTT-<br />

1023 showed good in vivo stability (80% of signal<br />

from intact antibody at 72 h after injection).<br />

The estimated human radiation dose resulting<br />

from [ 124 I]BTT-1023 was 3.66 mSv/MBq. Provided<br />

that thyroid gland uptake is blocked, the effective<br />

dose in a human adult of about 70 kg would<br />

imaging life<br />

decrease to 0.55 mSv/MBq, which is equivalent<br />

to 21 mSv from 35 MBq and 30 mSv from 50 MBq<br />

of [ 124 I]BTT-1023 PET.<br />

Conclusions: [ 124 I]BTT-1023 retained its biological<br />

activity to bind hVAP-1 also after radioiodination.<br />

This PET study of [ 124 I]-labeled VAP-1 targeting<br />

BTT-1023 antibody further elucidated whole-body<br />

distribution and pharmacokinetics of the therapeutical<br />

antibody, and supports clinical trials with [ 124 I]<br />

BTT-1023 at doses of 50 MBq or less.<br />

Acknowledgement: The study was conducted within<br />

the Finnish CoE in Molecular Imaging in Cardiovascular<br />

and Metabolic Research supported by the<br />

Academy of Finland, University of Turku, Turku<br />

University Hospital and Åbo Akademi University.<br />

Anu Autio is a PhD student supported by Drug Discovery<br />

Graduate School.<br />

References:<br />

1. Salmi M, Jalkanen S. Science. 257:1407-1409 (1992)


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

Pharmacological characterization of iodine labeled adenosine kinase inhibitors<br />

Sihver W. (1) , Schulze A. (1) , Kaufholz P. (1) , Meyer A. (1) , Grote M. (2) .<br />

(1) Research Center Jülich, Germany<br />

(2) Hannover Medical School, Germany<br />

w.sihver@fz-juelich.de<br />

Introduction: The intracellular enzyme adenosine<br />

kinase (AK) catalyzes the phosphorylation of adenosine<br />

to adenosine monophosphate thus controlling<br />

the adenosine concentration. The inhibition of<br />

AK enhances the amount of adenosine, which is suggested<br />

to have neuroprotective, anticonvulsant, and<br />

antinociceptive effects [1,2] . Years ago, the non-nucleoside<br />

AK inhibitor ABT-702 has been investigated in<br />

vivo and in vitro [3,4] . In the present study the distribution<br />

of the radioiodine labeled adenosine kinase<br />

inhibitors [ 131 I]iodotubercidine ([ 131 I]IT) and [ 131 I]<br />

ABT-702 as well as the pharmacological behaviour<br />

of other known AK inhibitors (e.g. A134974) versus<br />

[ 131 I]IT was compared in rat brain. Furthermore the<br />

AK inhibiting strength of other halogenated ABT-<br />

702 derivatives was determined.<br />

Methods: The syntheses of [ 131 I]IT and [ 131 I]ABT-702,<br />

as well as the different halogenated ABT-702 derivatives<br />

were performed in house. Autoradiography was<br />

conducted using frozen rat brain sections. Binding<br />

competition was done with IT and ABT-702 vs. [ 131 I]<br />

IT. AK inhibition assays were performed with [ 3 H]<br />

adenosine and ATP using pig hippocampus cytosol.<br />

Results: Autoradiography showed generally high<br />

binding all over the brain, but more distinct in the<br />

hippocampus, outer layers of the cortex, and gray<br />

matter of cerebellum using [ 131 I]IT (10nM) compared<br />

to [ 131 I]ABT-702 (7 nM). [ 131 I]IT Binding could<br />

be blocked more than 90% with 5-IT and A134974,<br />

about 70% with tubercidine and 60% with ABT-702.<br />

[ 131 I]ABT-702 Binding could be blocked not even 50<br />

% with ABT-702, and even less with the other AK inhibitors.<br />

K i s of IT and ABT-702 were 20 nM and 850<br />

nM, respectively, in rat hippocampus vs. [ 131 I]IT.<br />

In the AK assays, A134974 and IT had the highest<br />

AK inhibition strength in hippocampal cytosol, followed<br />

by ABT-702 > I- ABT-702 > Cl- ABT-702 ><br />

F- ABT-702 with 78%, 62%, 42%, 30% and 21% inhibition,<br />

respectively.<br />

Conclusions: As a result of this study it is suggested<br />

that [ 131 I]IT and [ 131 I]ABT-702 binding represent<br />

AK distribution in rat brain. [ 131 I]IT appears to be<br />

the preferred ligand for in vitro binding compared<br />

H 2N<br />

N<br />

NH 2<br />

N<br />

I<br />

N N<br />

N<br />

X = Br: ABT-702<br />

X = I: I-ABT-701<br />

X = Cl: Cl-ABT-702<br />

X = F: F-ABT-702<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

N<br />

HO<br />

X<br />

O<br />

N N<br />

OH<br />

OH<br />

O<br />

5-Iodotubercidine (IT)<br />

to [ 131 I]ABT-702. ABT-702 Has recently shown<br />

beneficial therapeutic potential [3, 5] , but radioiodo-ABT-702<br />

seems to be unsuited as radiotracer.<br />

The AK inhibition strength was high both for IT<br />

and ABT-702, but low for the halogenated ABT-702<br />

derivatives. Since IT shows good in vitro behaviour<br />

as a radioligand as well as AK inhibitor, further experiments<br />

with [ 131 I]IT in vivo might be interesting.<br />

References:<br />

1. Kowaluk EA et al.; Expert Opin Investig Drugs.9:551-64<br />

(2000)<br />

2. Boison D; Drug News Perspect. 20:607-11 (2007)<br />

3. Jarvis MF et al.; J Pharmacol Exp Ther. 295:1156-64<br />

(2000)<br />

4. Kowaluk EA et al.; J Pharmacol Exp Ther. 295:1165-74<br />

(2000)<br />

5. McGaraughty S et al.; Curr Top Med Chem. 5:43-58 (2005)<br />

P-018<br />

poStEr<br />

IMAGING in DRUG DEVELOPMENT


WarSaW, poland May 26 – 29, 2010<br />

P-019 Determining the nasal residence time of protein-polymer conjugates for nasal vaccination<br />

using a novel imaging technique<br />

132<br />

Slütter B. (1) , Que I. (2) , Soema P. (1) , Hennink W. (3) , Kaijzel E. (2) , Löwik C.W.G.M. (2) , Jiskoot W. (1) .<br />

(1) Amsterdam Center for Drug Research (LACDR), The Netherlands<br />

(2) Leiden University Medical Center, The Netherlands<br />

(3) Utrecht Institute for Pharmaceutical Sciences (UIPS), The Netherlands<br />

bslutter@lacdr.leidenuniv.nl<br />

Introduction: Nasal administration of vaccines<br />

holds great promise as a painless alternative for the<br />

use of needles. Nonetheless, only one nasal vaccine<br />

is currently on the market (Flumist®). A major hurdle<br />

for successful nasal vaccination is the relatively<br />

short residence time of the vaccine in the nasal cavity,<br />

which hampers efficient uptake of the antigen<br />

through the nasal epithelium. Increasing the nasal<br />

residence time of the antigen is therefore an interesting<br />

approach to improve the efficacy of nasal<br />

vaccines. Here we investigated the possibility of extending<br />

the nasal residence time of a small antigen<br />

(ovalbumin, OVA) with a mucoadhesive polymer,<br />

trimethyl chitosan (TMC), using a novel live imaging<br />

technique1.<br />

Methods: A near-IR fluorescent probe (IR dye<br />

CW800) was covalently linked to OVA. TMC was<br />

either mixed with or conjugated to OVA using the<br />

SPDP method2. After nasal administration of the<br />

antigen to hairless mice, the fluorescence intensity<br />

in the nasal cavity was assessed using an IVIS Spectrum®<br />

and followed in time (Fig1).<br />

Results: We observed an exponential decay of fluorescence<br />

intensity after administration of OVA alone,<br />

whereas after co-administration of OVA with TMC<br />

or TMC-OVA conjugate fluorescence decay was delayed<br />

substantially.<br />

Conclusions: Using a live imaging technique we successfully<br />

studied the nasal residence time of a model<br />

subunit antigen. TMC prolongs the nasal residence<br />

time of OVA, either by mixing it with or conjugating<br />

it to the antigen.<br />

imaging life<br />

Fig1: Nasal residence time of OVA determined using<br />

fluorescent detection of OVA-IRdye CW 800. Intensity of<br />

fluorescence signal from the nasal cavity was measured<br />

in time and normalized for time point 0. n=3+/- SEM.<br />

References:<br />

1. Hagenaars et al. Role of trimethylated chitosan (TMC)<br />

in nasal residence time, local distribution and toxicity<br />

of an intranasal influenza vaccine. J Control Release.<br />

2010, in press.<br />

2. Slütter et al. Conjugation of ovalbumin to trimethyl<br />

chitosan improves immunogenicity of the antigen. J<br />

Control Release. 2010, in press.


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

Nanotubes as mutli-modality vehicles for imaging and therapy of cancer<br />

Tworowska I. (1) , Mackeyev Y. (2) , Sims-Mourtada J. (1) , Wilson L. J. (2) .<br />

(1) RadioMedix, Houston, USA<br />

(2) Rice University, Houston, USA<br />

itworowska@radiomedix.com<br />

Introduction: There is a growing interest in development<br />

of new radiolabeled nanocarriers for targeted<br />

delivery of isotopes. Nanotubes have been actively<br />

explored as new systems for delivery of anticancer<br />

drugs [1], fluorescent probes [2], genes [3], and peptides<br />

[4] to the target tissues. Previously reported<br />

synthesis of Gd-loaded US-tubes [5] and I 2 -modified<br />

US-tubes [6] have opened new possibilities in<br />

designing of contracts agents for MRI and CT. Here,<br />

we report on the synthesis of radiolabeled SWNT<br />

that can serve as vehicles for PET/SPECT imaging<br />

and cancer therapy.<br />

Methods: HiPco single-wall nanotubes (SWNTs)<br />

were chemically cut into US-tubes, using the established<br />

procedure [7], sonicated at RT in 0.5M<br />

HNO for 30min-2h. US-tubes were loaded with<br />

3<br />

radioisotopes: 177LuCl , 3 99mTcCl , 3 68GaCl , and cold<br />

3<br />

Re compounds (ReCl 5 , NH 4 ReO 4 , [NBu 4 ] + ReClO 4<br />

manually or using automated module for labelling,<br />

SmarTrace TM . The radiochemical yield was determined<br />

by radio-TLC and ICP-OES (Inductively<br />

coupled plasma optical emission spectrometry). Stability<br />

tests of labelled US-tubes were performed at<br />

RT for 1- 24h using 1M PBS, 0.1% FBS and transchelator<br />

(0.1M EDTA) to determine the desorption<br />

half-life of the labelled US-tubes.<br />

Results: Loading of US-tubes with 68 Ga was performed<br />

in 0.5M NH 4 OAc buffer at 90°C for 10<br />

min using 68 GaCl 3 eluted from 68 Ge/ 68 Ga generator<br />

(iThemba). Yield of the synthesis was found to be<br />

pH dependent with over 70% loading at pH=3.6- 4.1<br />

and to decrease drastically to 1% at pH51%. Yield of the reaction decreased<br />

to 1% in the absence of SnCl 2 , or when reduction<br />

proceeded after completion of labelling.<br />

Best 177 Lu loading of US-tubes was performed in<br />

0.1 M NH 4 OAc at pH=5.1 at 90°C for 20 min. The<br />

final yield of this synthesis was found to be >55%<br />

after repeated dialysis with 0.1M EDTA, pH=4.7.<br />

- )<br />

Loading of US-tubes with cold ReCl proceeded<br />

5<br />

in 0.6M NaOH at 90oC for 1h with final yield<br />

> 20%. Application of other Re compounds<br />

([NBu ] 4 + - ReClO and NH4ReO ) for modification<br />

4<br />

4<br />

of nanotubes gave products with yield higher than<br />

17% and approximately 0.4%, respectively.<br />

Conclusions: Our preliminary studies have shown<br />

that loading of US-tubes with imaging and therapeutic<br />

isotopes is feasible. Application of nanotubes<br />

in nuclear medicine may expand possibilities for<br />

development of new multi-modality agents for early<br />

detection and therapy of cancer.<br />

References:<br />

1. Tripisciano C., Kraemer K., Taylor A., Borowiak-Palen E.,<br />

Chem. Phys. Lett., 2009, 478, 200-205<br />

2. Wong Shi Kam N., Liu Z., Dai H., Angew. Chem. Int. Ed.,<br />

2006,45, 577-581<br />

3. Ramathan T, Fisher F.T., Ruoff R.S., Brinson L.C., Chem.<br />

Mater., 2005, 17, 1290-1295<br />

4. Kam N.W., Dai H., J. Am. Chem. Soc., 2005, 102, 6021-<br />

6026<br />

5. Mackeyev Y., Hartman K.B, Ananta J.S., Lee A.V., Wilson<br />

L.J., J. Am. Chem. Soc., 2009; 131(24): 8342–8343<br />

6. Ashcroft J.M, Hartman K.B., Kissel K.R., Mackeyev Y.,<br />

Pheasant S., Young S., Van der Heide P.A., Mikos A.,<br />

Wilson L.J., Advanced Materials, 2007, 19, 573-576<br />

7. Mickelson E.T., Huffman C.B., Rinzler A.G., Smalley R.E.,<br />

Hauge R.E., Margrave J.L., Chem Phys Lett, 1998, 296,<br />

188-194.<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

P-020<br />

poStEr<br />

IMAGING in DRUG DEVELOPMENT


WarSaW, poland May 26 – 29, 2010<br />

P-021 Early assessment of temozolomide treatment efficacy in glioblastoma using [ 18 F]FLT PET<br />

imaging<br />

134<br />

Viel T. (1) , Backes H. (1) , Hadamitzky M. (1) , Monfared P. (1) , Rapic S. (1) , Rudan D. (1) , Schneider G. (1) , Neumaier B. (1) , Jacobs A.H. (1,2) .<br />

(1) MPI for Neurological Research, Köln, Germany<br />

(1,2) European Institute for Molecular Imaging – EIMI, University Muenster, Germany<br />

thomas.viel@nf.mpg.de<br />

Introduction: Temozolomide chemotherapy to radiation<br />

therapy is now the standard therapy for<br />

glioblastomas [1] . However there is considerable uncertainty<br />

with regard to the indication for and the<br />

chances of success of chemotherapy in affected patients.<br />

Standard Temozolomide treatment in clinic<br />

consists of 5 days treatment with 150-200 mg/m2 per<br />

day, repeated every 28 days. Treatment efficacy assessment<br />

is performed using MRI Imaging before treatment<br />

and after 3 cycles of treatment (meaning after<br />

3 months). Due to the rapid evolution of the disease<br />

(median survival of patient bearing glioblastoma is<br />

around 15 months), methods to assess TMZ efficacy<br />

early during treatment is needed. In several clinical<br />

studies [ 18 F]FLT PET imaging has been validated to<br />

assess proliferation of different types of tumors in<br />

vivo [2] , and could therefore be of interest for evaluation<br />

of TMZ during treatment of glioblastoma.<br />

The purpose of this study was to monitor the metabolic<br />

effects of temozolomide (TMZ) chemotherapy<br />

in malignant gliomas by means of repeated Positron<br />

Emission Tomography (PET) with [ 18 F]FLT.<br />

Methods: A glioma cell line, displaying a low resistance<br />

to TMZ (Gli36) was treated with two low<br />

doses of TMZ (25 and 50 µM). Two stable cell lines<br />

were obtained and characterized regarding their<br />

resistance to TMZ by growth and clonogenic assay.<br />

Resistant and sensitive cells were xenografted into<br />

nude mice. Mice were treated during five days with<br />

daily intra-peritoneal injection of 50 mg/kg of TMZ.<br />

Efficacy of TMZ treatment is followed using FLT<br />

imaging, before treatment as well as two and seven<br />

days after beginning of treatment.<br />

Results: Two stable cell lines resistant to TMZ were<br />

established (Gli36-25TMZ and Gli36-50TMZ). The<br />

EC50 for the induction of cytotoxicity cell death (as<br />

determined by growth assay) were 10 µM (Gli36), 110<br />

µM (Gli36-25TMZ) and 460 µM (Gli36-50TMZ). The<br />

EC50 for the prevention of clonogenic growth (as determined<br />

by clonogenic assay) were 10 µM (Gli36), 50<br />

µM (Gli36-25TMZ) and 325 µM (Gli36-50TMZ). In<br />

vivo, the TMZ treatment induced a strong reduction<br />

of Gli36 tumor volume, while Gli36-50TMZ continued<br />

to grow. Correlation between diminution of SUV in<br />

imaging life<br />

the tumor after two days of treatment (with regards to<br />

SUV before treatment) and diminution of tumor size<br />

seems to be observed so far (n=3 for each tumor type).<br />

Conclusions: Our results so far indicate that [ 18 F]FLT<br />

PET scan could be appropriate for an early evaluation<br />

of the response of Glioblastoma to TMZ chemotherapy.<br />

Acknowledgement: This work is supported in part<br />

by DiMI (LSHB-CT-2005-512146).<br />

References:<br />

1. Norden AD, Drappatz J, et al; Lancet Neurol. 7(12):1152-<br />

60 (2008).<br />

2. Ullrich RT, Zander T, et al; PLoS One. 3(12):e3908<br />

(2008).


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

Sensitive time-gated FRET microscopy of G-Protein coupled receptors using suicide enzymes,<br />

lanthanide cryptates and fluorescent ligands<br />

Zwier J. (1) , Laget M. (1) , Cottet M. (2) , Durroux T. (2) , Mathis G. (1) , Trinquet E. (1) .<br />

(1) Cisbio Bioassays, Bagnols sur Cèze, France<br />

(2) Insitut de Génomique fonctionelle, France<br />

jzwier@cisbio.com<br />

Introduction: Donor crosstalk and non-specific fluorescence<br />

in classical biological FRET experiments<br />

using fluorescent proteins remains a major concern<br />

in high content screening. Using a time-gated microscope<br />

equipped with an intensified CCD camera<br />

[1], lanthanide cryptates and fluorescent-ligands it<br />

is possible to visualize G-protein coupled receptors<br />

(GPCR) labeled with the SNAP-tag® suicide enzyme<br />

at the cell membrane. Oligomerization of GPCR’s can<br />

be studied using time gated FRET without significant<br />

background. Using fluorescent ligands, binding<br />

and receptor trafficking can be imaged without ratiometric<br />

treatment of data.<br />

Methods: A bright terbium-cryptate (lumi4®-Tb)<br />

coupled to 0 6 -Benzylguanine is used to covalently<br />

label GPCR’s tagged with the SNAP-tag suicide enzyme<br />

developed by the laboratory of Kai Johnsson<br />

[2]. The luminescence of this lanthanide compound,<br />

which has a lifetime of about 2 ms, can be time-gated<br />

in such a way that all background fluorescence from<br />

the cells has decayed and only specific lanthanide or<br />

sensitized acceptor emission to either red or green<br />

emitting dyes can be detected. An in-house developed<br />

microscope equipped with an intensified CCD<br />

camera can monitor this luminescence. HEK/COS/<br />

CHO cells were either stably or transiently transfected<br />

with plasmids containing the SNAP-tagged GPCR<br />

under study and labeled with the benzylguanines of<br />

choice after which they are fixed.<br />

Results: Due to the significant Förster radius of 58<br />

A of the lumi4-Tb/red donor-acceptor couple, homodimerization<br />

of a GPCR can be monitored using<br />

a mix of the Tag-lite® reagent SNAP-Lumi4-Tb and<br />

SNAP-red wich results in a bright time gated sensitized<br />

emission image at 665 nm. These results confim<br />

the results obtained by dimerization assays [3]<br />

in standard plate readers.<br />

Due to the advantages of this technique it is also<br />

possible to visualize specific labeling of fluorescent<br />

ligands on the GPCR of interest. Using fluorescent<br />

ligands without the use of time gating gives significant<br />

contributions of non-specific binding. Since<br />

sensitized emission only occurs when donor and<br />

acceptor are in close proximity ligand receptor<br />

interactions were monitored for several GPCR’sligand<br />

couples including chemokine receptors.<br />

Conclusions: Time gated microscopy with terbiumcryptate<br />

is a convenient and novel way to avoid<br />

non-specific emission signals and to monitor FRET<br />

intensities showing genuine interactions between<br />

proteins and their ligands. This might become a<br />

powerful method to develop high content assays for<br />

drug evaluation.<br />

Acknowledgement: Tag-lite is a registered trademark<br />

of Cisbio bioassays. Lumi4 is a regis tered<br />

trademark of Lumiphore Inc. SNAP-tag is a registered<br />

trademark of New England Biolabs.<br />

References:<br />

1. Ghose S et al, J. Alloys and cmpds 451:35-37 (2008)<br />

2. Keppler A et al, Nature Biotechnology 21:86-89 (2003)<br />

3. Maurel D, Comps-Agrar L et al, Nature Methods 5:561-<br />

567 (2008)<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

P-022<br />

poStEr<br />

IMAGING in DRUG DEVELOPMENT


136<br />

WarSaW, poland May 26 – 29, 2010<br />

P-023 Selection of a nanobody scaffold with low renal retention<br />

D’huyvetter M. , Vaneycken I. , Tchouate Gainkam O. , Hernot S. , Caveliers V. , Xavier C. , Devoogdt N. , Lahoutte T. .<br />

Vrije Universiteit Brussel, Jette, Belgium<br />

mdhuyvet@vub.ac.be<br />

Introduction: Nanobodies are small (15kDa) antibody<br />

fragments, derived from heavy chain-only<br />

antibodies present in Camelidae. The structure<br />

consists of a scaffold and three CDR loops. The<br />

CDR loops determine the specificity of the nanobody.<br />

Nanobodies, labelled with a therapeutic<br />

radionuclide, could be used for the treatment of<br />

cancer. Unfortunately, the in vivo biodistribution<br />

shows high renal retention of radio-labeled nanobodies<br />

in the kidney cortex and this could result<br />

in an important kidney toxicity. However, during<br />

the in vivo screening of a large set of nanobodies<br />

we noticed that there is a wide variety in this kidney<br />

retention. This variability could be partially<br />

related to the amino acid sequence of the nanobody<br />

scaffold. In this study we aim to identify the<br />

nanobody scaffold with the lowest renal retention.<br />

Methods: The renal retention of 8 Nanobodies was<br />

evaluated in healthy Wistar rats. 99mTc labeling<br />

was performed using 99mTc-tricarbonyl (Isolink,<br />

Covedien). Dynamic planar imaging with a gamma<br />

camera was performed immediately after injection<br />

(100 frames of 30s). Three hours post injection an<br />

additional static image was acquired. Time activity<br />

curves of the kidney were generated using AMIDE.<br />

imaging life<br />

Results: We measured two distinct kinetic profiles:<br />

one showing a steadily increase in function of time<br />

and the other showing an initial peak (5 min p.i.)<br />

followed by a decrease. The highest renal activity<br />

measured at 1h p.i. was 46.8 % IA (range 41.6-53.3<br />

%IA), while the lowest was 11.3 % IA (range 10.1-<br />

13.2 %IA). At 3h p.i. we measured upto 69.3 %IA<br />

in the kidney for the group with a steadily increasing<br />

profile, while for the other group it remained<br />

low at 14.8 %IA.<br />

Conclusions: We identified a scaffold with low<br />

renal retention. The use of this scaffold for nanobody<br />

based targeted radionuclide therapy could<br />

significantly reduce kidney toxicity.<br />

Acknowledgement: The research at ICMI is funded<br />

by the Interuniversity Attraction Poles Program –<br />

Belgian State – Belgian Science Policy. Matthias<br />

D’huyvetter is funded by SCK-CEN/VUB.


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

In vitro assessment of androgen mediated uptake of 18 F-FDG, 11 C-choline and 11 C-acetate in<br />

prostate cancer<br />

Emonds K. (1) , Swinnen J. (1) , Van Weerden W. (2) , Nuyts J. (1) , Mortelmans L. (1) , Mottaghy F. (3) .<br />

(1) KULeuven, Belgium<br />

(2) Josephine Nefkens Institute, The Netherlands<br />

(3) University Hospital Aachen, Germany<br />

Kimy.Emonds@med.kuleuven.be<br />

Introduction: Androgen deprivation is one of the<br />

first line palliative treatment approaches in recurrent<br />

prostate cancer. Although 18 F-FDG is mostly applied<br />

in oncological PET imaging, a low diagnostic performance<br />

has been shown for prostate cancer detection.<br />

Otherwise, both 11 C-choline and 11 C-acetate offer a<br />

higher diagnostic efficiency for PET detection of relapsing<br />

patients. With this study we aimed to evaluate the<br />

androgen dependency of the uptake of metabolic PET<br />

tracers ( 18 F-FDG, 11 C-choline and 11 C-acetate) in five<br />

different human prostate cancer cell lines, distinctive<br />

considering their androgen responsiveness, in order to<br />

define the most reliable tracer for treatment response<br />

assessment.<br />

Methods: Cell uptake experiments were performed with<br />

the prostate cancer cell lines LNCaP, PC346C, 22Rv1,<br />

PC346DCC and PC3, and the benign prostatic hyperplasia<br />

cell line BPH-1. Prior to the uptake experiments,<br />

cells were cultured in the presence of charcoal treated<br />

serum (native). In parallel cell cultures 10 -8 M R1881, 10 -<br />

10 M R1881, 10 -6 M bicalutamide or the combination of<br />

10 -10 M R1881 and 10 -6 M bicalutamide was added to the<br />

medium. The significant influence of androgens on the<br />

uptake of 18 F-FDG, 11 C-choline and 11 C-acetate in each<br />

prostate cancer cell line was evaluated using ANOVA<br />

and Tukey-HSD post-hoc analysis. The relative tracer<br />

uptake was evaluated in all prostate cancer cell lines<br />

with respect to the tracer uptake in BPH-1.<br />

Results: A significant increased 11 C-choline uptake is<br />

observed in the androgen responsive and unresponsive<br />

cell line, respectively PC346C and 22Rv1, grown in the<br />

presence of androgens (10 -8 M R1881). The same androgen<br />

concentration also caused a significant increase<br />

in 18 F-FDG uptake in LNCaP (androgen dependent)<br />

and 22Rv1. In both androgen unresponsive cell lines<br />

(PC3, PC346DCC) androgens did not significantly affect<br />

the uptake of these metabolic PET tracers. Unlike<br />

11 C-choline and 18 F-FDG, 11 C-acetate uptake was not<br />

influenced by androgen supplementation in any prostate<br />

cancer cell line.<br />

Compared to the tracer uptake in BPH-1, all prostate<br />

cancer cell lines showed a significant higher 11 C-acetate<br />

and 11 C-choline uptake. In contrast, a relative increased<br />

18 F-FDG uptake was only observed in PC346C and -DCC.<br />

Conclusions: 11 C-acetate uptake is androgen<br />

independent in every prostate cancer cell line,<br />

whereas a significant higher 11 C-choline and<br />

18 F-FDG uptake was established by androgens<br />

in respectively two (PC346C, 22Rv1) and three<br />

(LNCaP, PC346C, 22Rv1) human prostate cancer<br />

cell lines. Also, 11 C-acetate has the most<br />

clearly elevated uptake in all prostate cancer<br />

cell lines with respect to the benign prostatic<br />

hyperplasia cell line.<br />

The absent influence of androgens on 11 C-acetate<br />

uptake and the better differentiation of<br />

cancer from benign prostatic hyperplasia with<br />

this PET tracer compared to 11 C-choline and<br />

18 F-FDG, suggests a higher efficiency of 11 C-acetate<br />

PET for evaluation of treatment response.<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

P-024<br />

poStEr<br />

CANCER from BENCH to BEDSIDE


138<br />

WarSaW, poland May 26 – 29, 2010<br />

P-025 Comparison of two 68 Gallium-labeled octreotide analogues for molecular PET(CT) imaging<br />

of neuroendocrine tumours<br />

Garcia C. , Woff E. , Muylle K. , Ghanem G. , Van der Linden B. , Vandormael S. , Rutten E. , Flamen P. .<br />

Institut Jules Bordet. Université Libre de Bruxelles, Brussels, Belgium<br />

camilo.garcia@bordet.be<br />

Introduction: PET(CT) using 68 Ga-labeled octreotide<br />

analogues is able to show the expression of<br />

somatostatin receptors (SSR) on gastroenteropancreatic<br />

neuro-endocrine tumours (GEP NET). The<br />

study aim was to compare the biodistribution and<br />

the tumour uptake intensity of two radiolabeled<br />

octreotide analogues, 68 Ga-DOTA-TOC ([DOTA-<br />

DPhe1,Tyr3]-octreotide) and 68 Ga-DOTA-TATE<br />

([DOTA-DPhe1,Tyr3]-octreotate).<br />

Methods: Octreo-PET(CT) was performed on 58<br />

patients: 29 patients using 68Ga-DOTA-TOC and<br />

29 patients using 68 Ga-DOTA-TATE. Standard Uptake<br />

Values (SUV) was measured in normal organs<br />

and in the lesions (SUVmax). Patient preparation,<br />

acquisition and image processing was standardized<br />

and identical in both subgroups. None of the<br />

patients had received any peptide therapy or radiolabeled<br />

peptide therapy before the study. The<br />

time delay between tracer injection and PET(CT)<br />

acquisition for respectively DOTA-TOC vs. DOTA-<br />

TATE patients was 95 min ± 0.01 vs. 100 min ± 0.01<br />

(p=NS); the mean injected tracer activity was 90.28<br />

MBq ± 46.62 vs. 88.8 MBq ± 21.09 (2.44 mCi ± 1.26<br />

vs. 2.40 mCi ± 0.57) (p=NS), and BMI 25.7 ± 3.71<br />

vs. 25.2 ± 3.6 (p=NS).<br />

imaging life<br />

Results: PET(CT) identified 107 tumor lesions: 52<br />

(49%) in the DOTA-TOC, and 55 lesions (51%) in<br />

the DOTA-TATE group. No significant differences<br />

of SUV(max) values were found in GEP NET lesions<br />

between DOTA-TATE and DOTA-TOC groups.<br />

Physiologic uptake in organs did not significantly<br />

differ between the 2 radiopharmaceuticals, except<br />

for the uptake of 68 Ga DOTA-TOC which was significantly<br />

lower in the head of pancreas than the<br />

uptake of 68 Ga DOTA-TATE (3.4 ± 1.5 vs. 4.6 ± 1.6)<br />

(P


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

Real time per operative optical imaging for the improvement of tumour surgery in an in vivo<br />

micro-metastases model<br />

Keramidas M. , Josserand V. , Righini C. , Coll J. L.<br />

INSERM U823, Grenoble, France<br />

michelle.keramidas@ujf-grenoble.fr<br />

Introduction: In a wide range of cancer cases,<br />

surgery is the first therapeutic indication before<br />

radiotherapy and chemotherapy. In that<br />

way the prognostic strongly depends on the tumour<br />

removal exhaustiveness and in particular<br />

on metastases elimination.<br />

We developed a couple near-infrared fluorescent<br />

tracer / per operative detection system in<br />

order to improve tumour surgery efficacy.<br />

RAFT-c(RGD)4-Alexa 700 (Angiostamp®, Fluoptics)<br />

specifically bind to integrin αvß3, a<br />

receptor strongly expressed in angiogenesis<br />

and in many tumour types. The per-operative<br />

detection system (Fluobeam 700, Fluoptics) is<br />

a portative 2D fluorescent imager that can be<br />

used in white light environment and so, could<br />

be used directly during surgery to help the surgeon<br />

for tumour and metastases excision.<br />

We already demonstrated in animal models<br />

the very significant improvement in primary<br />

tumours resection: higher number of tumour<br />

nodules removed, sane margins on the removed<br />

fragments and surgery time divided by 2 (Keramidas<br />

M., British Journal of Surgery 2010).<br />

In the clinical situation, recurrence of cancer<br />

by metastases invasion after primary tumour<br />

surgery is a critical point. The aim of the present<br />

study is to evaluate the metastases resection<br />

impact on the survey. In order to proceed we<br />

first need to establish a suitable animal model<br />

of micro-metastases following primary tumour<br />

surgery. After calibration of the metastatic<br />

in vivo model, we will evaluate the survey of<br />

the twice-operated animals (metastases resection<br />

after primary tumour removal) versus the<br />

once-operated animals (only primary tumour<br />

removal).<br />

Methods: Luciferase positive tumour cells (TS/<br />

Apc-luc) are injected in the kidney capsule of<br />

nude mice and the primary tumour growth is<br />

followed by in vivo bioluminescence imaging.<br />

7 days after tumour cells implantation, the tumoral<br />

kidney is removed and the metastases<br />

development is followed by in vivo bioluminescence<br />

imaging. Then RAFT-c(RGD)4-Alexa<br />

700 is injected intravenously and twenty-four<br />

hours later the portative fluorescence detection<br />

system is used to assist the metastases excision.<br />

The possible recurrence of cancer is followed<br />

by in vivo bioluminescence imaging. Mice are<br />

sacrificed when they loose 10% of their weight.<br />

The mice survey is compared with the one of<br />

two control groups: in one group the mice don’t<br />

undergo the second surgery for metastases resection,<br />

and in a second group the mice don’t<br />

undergo any surgery and keep the primary kidney<br />

tumour.<br />

Results: Micro-metastases appear about 7 days<br />

after the primary tumour removal and soar up<br />

to 20 days after the first surgery. The metastases<br />

removal assisted by the RAFT-c(RGD)4-Alexa<br />

700 and the per operative fluorescence detection<br />

system significantly improved the mice<br />

survey (36 days).<br />

Conclusions: We developed an in vivo model of<br />

micro-metastases invasion following primary<br />

tumour surgery which is very relevant regarding<br />

to the clinical situation of head and neck<br />

or prostate cancer. The use of RAFT-c(RGD)4-<br />

Alexa 700 coupled with the portative device allows<br />

micro-metastases detection, significantly<br />

improve their resection and increase the mice<br />

survey.<br />

References:<br />

1. Intraoperative near-infrared image guided surgery for<br />

peritoneal carcinomatosis in a preclinical experimental<br />

model. M. KERAMDAS, V. JOSSERAND, RIGHINI C.A., WENK<br />

C., FAURE C. And COLL J.L. British Journal of Surgery,2010.<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

P-026<br />

poStEr<br />

CANCER from BENCH to BEDSIDE


140<br />

WarSaW, poland May 26 – 29, 2010<br />

P-027 Fluorescence imaging modalities for imaging gastrointestinal tumor models<br />

Schulz P. , Cordula D. , Rexin A. , Wiedenmann B. , Groetzinger C. .<br />

Charite, Universitätsmedizin Berlin, Germany<br />

petra.schulz@charite.de<br />

Introduction: A number of peptide receptors have<br />

been found to be overexpressed in tumors of the GI<br />

tract. The overexpression of somatostatin receptors<br />

in neuroendocrine gastroenteropancreatic tumors<br />

has long been utilized for molecular imaging using<br />

radiolabelled somatostatin analogs in scintigrafic<br />

procedures. Other peptide GPCRs may therefore<br />

represent attractive new targets. The ongoing project<br />

is occupied in particular with the design of GPCR<br />

peptide ligands, which bind with high specificity to<br />

surface receptors of tumor cells. Pharmacologically<br />

optimized peptides could be used as vehicles for<br />

transport of contrast agents as well as therapeutics.<br />

Methods: The ongoing project follows four main<br />

goals: (1) The validation of GPCR peptides as tumor<br />

targets for the gastrointestinal tract (2) The identification<br />

of new peptide binding targets (3) The Optimization<br />

of peptide structures for imaging and targeted<br />

therapy and (4) the development of NIRF-probes<br />

and –techniques for a better diagnosis of tumors.<br />

NIRF probes are validated in established in vivo tumor<br />

models, including subcutaneous tumor models<br />

and in a number of orthotopic tumor models using<br />

planar and tomografic fluorescent imaging, micro<br />

CT and endoscopy for small animals. For monitoring<br />

and characterization of tumor growth of human cancer<br />

cells in nu/nu mice the bioluminescence imaging<br />

approach is utilized. For this purpose several stable<br />

cancer lines (colorectal, pancreatic, neuroendocrine),<br />

expressing the luciferase gene were established.<br />

Results: Currently, a number of peptide, antibody<br />

and small-molecule contrast agents are validated.<br />

Each tracer is characterized for its potency as a contrast<br />

agent by imaging with a 2-D fluorescence imaging<br />

system, which reveals target-to background<br />

ratios of the contrast agents, characteristic for target<br />

tissue specificity. Limitations of planar imaging<br />

systems are still the lack of depth resolution and<br />

difficult quantification. Currently we are therefore<br />

evaluating a fluorescence tomographic system with<br />

regard to deep tissue imaging and signal quantification.<br />

Tomograpic images also allow fusion with<br />

other imaging mmodalities such as CT to confirm<br />

the anatomical localization of the fluorescent signal.<br />

imaging life<br />

Conclusions: Although near-infrared radiation has<br />

a higher penetration depth than the visual light,<br />

scattering and absorption still prevent whole-body<br />

imaging. One potential application for the use of<br />

near-infrared optical agents in humans is therefore<br />

fluorescence-guided endoscopy. We are currently establishing<br />

a protocol for endoscopic imaging in mice<br />

with colorectal cancer with a rigid endoscopic device<br />

in combination with fluorescence-guided detection<br />

using a fiber endoscope.<br />

Acknowledgement: This work was supported by grant<br />

03IP614 from German ministry of research (BMBF)


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

Assessment of effectiveness and toxicity of the therapy with somatostatin analogue labelled<br />

90Y-DOTATATE in patients with non-functional pancreatic neuroendocrine tumours (PNT)<br />

Sowa-Staszczak A. .<br />

Nuclear Medicin Unit, Krakow, Poland<br />

sowiana@gmail.com<br />

Introduction: Therapy with labelled somatostatin<br />

analogues is the modern approach to the patients<br />

with disseminated or unresectable NETs expressing<br />

somatostatin receptors (SSTR). Octreotate is<br />

the somatostatin analogue with very high affinity<br />

to the SSTR type 2, most commonly present in<br />

neuroendocrine tumours. In non-functional PNT,<br />

grading and systemic metastases have a significant<br />

impact on survival. Overall, the 5-year survival<br />

rate is about 33%. The chemotherapy is the most<br />

common treatment approach. The aim of the study<br />

was to assess the efficacy and toxicity of peptide<br />

receptor radionuclide therapy (PRRT) with the use<br />

of the high affinity somatostatin receptor subtype<br />

2 analogue, 90Y labelled Tyr3-octreotate, (90Y-<br />

DOTATATE) in non-functional pancreatic neuroendocrine<br />

tumours.<br />

Methods: 19 patients with metastatic NET were diagnosed<br />

in the Department of Endocrinology UJCM.<br />

5 patients with high proliferation index (Ki >20%, 4-<br />

negative SRS, tumour size 3-7cm) were directed to<br />

chemotherapy. 14 patients with positive SRS scan due<br />

to disseminated and/or unoperable PNTs (2 patients)<br />

were qualified to PRRT (6 men, 8 women, mean age 54,7<br />

years old, Karnofsky’s index > 70-100%). The size of the<br />

tumour was 2,3-12cm, Ki-67 was


142<br />

WarSaW, poland May 26 – 29, 2010<br />

P-029 Intra operative near-infrared fluorescent imaging of colorectal liver metastases using<br />

clinically available Indocyanine Green in a syngene rat model<br />

Van Der Vorst J. , Hutteman M. , Mieog J. , De Rooij K. , Kuppen P. , Kaijzel E. , Löwik C.W.G.M. , Van De Velde C. ,<br />

Vahrmeijer A. .<br />

Leiden University Medical Centre, The Netherlands<br />

j.r.van_der_vorst@lumc.nl<br />

Introduction: The survival of patients with colorectal<br />

carcinoma is mostly determined by the occurrence<br />

of distance metastases. When liver metastases occur,<br />

surgical resection can offer a 5-year survival of 35-<br />

40%. However, during resection, an adequate assessment<br />

of the extent of disease is limited, resulting in<br />

a high percentage of recurrence (40-50%). Ishizawa<br />

et al. reported that liver cancer could be identified<br />

using near-infrared (NIR) fluorescence and the clinically<br />

available indocyanine green (ICG) (1). However,<br />

the optimal dose of ICG and the interval between the<br />

administration of ICG and surgery is unclear.<br />

Methods: In the current study, the NIR probe ICG<br />

and the Mini-FLARE (Dr. J.V. Frangioni, Boston,<br />

USA) camera system were used. In 6 rats, 125,000<br />

CC531 cells were inoculated subcapsularly in<br />

three different lobes of the liver. After four weeks,<br />

tumors of approximately 3-5 mm diameter were<br />

present. In each rat, fluorescence was measured at<br />

24 and 48 hours post-injection of 0.04 mg (n = 3)<br />

or 0.08 mg (n = 3) ICG.<br />

Color<br />

Fig1: Intraoperative NIRF identification of colorectal liver metastases<br />

imaging life<br />

Results: In 6 rats, all colorectal liver metastases<br />

(n = 10) were intraoperatively identified using<br />

ICG and the Mini-FLARE camera system (Figure<br />

1). The fluorescent signal of the colorectal liver<br />

metastases was significantly higher than the signal<br />

of surrounding normal liver tissue (P < 0.001).<br />

No significant difference in mean signal to background<br />

ratio was found between imaging at 24 and<br />

48 hours post-injection. Furthermore, no significant<br />

difference in mean signal to background ratio<br />

was found between injection of 0.04 and 0.08 mg<br />

of ICG.<br />

Conclusions: This study demonstrated that colorectal<br />

liver metastases can be clearly identified during<br />

surgery using the clinically available NIRF probe<br />

ICG and the Mini-FLARE camera system. When<br />

the current intraoperative identification can be improved<br />

using this technique, resections can be performed<br />

more accurately and preoperatively missed<br />

metastases can be involved in surgical decision<br />

making during surgery.<br />

NIR Merge<br />

References:<br />

1. Ishizawa et al.; Cancer. 1;115(11):2491-504. (2009)


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

Molecular imaging of resistance to EGFR tyrosine kinase inhibitors by 18F-FLT PET/CT and its<br />

reversal in non small cell lung cancer<br />

Zannetti A. (1) , Iommelli F. (1) , Lettieri A. (1) , Pirozzi G. (2) , Salvatore M. (3) , Del Vecchio S. (3) .<br />

(1) Institute of Biostructures and Bioimages, National Research Council, Naples, Italy<br />

(2) Department of Experimental Oncology, National Cancer Institute, Naples, Italy<br />

(3) Institute of Biostructures and Bioimages, National Research Council; Department of Biomorphological and Functional Sciences, University of Naples “Federico II”, Naples, Italy<br />

antonella.zannetti@ibb.cnr.it<br />

Introduction: Multiple molecular mechanisms<br />

may underlie the resistance to EGFR tyrosine kinase<br />

inhibitors (TKIs), including the occurrence<br />

of secondary mutations such as T790M in the kinase<br />

domain of EGFR, redundant lateral signalling<br />

or alterations of apoptotic program mainly due to<br />

dysregulation of Bcl-2 family members. The aim<br />

of our study was to test whether 18F-Fluorothymidine<br />

(18F-FLT) could detect EGFR TKI refractory<br />

tumors and identify the mechanisms underlying<br />

such resistance so that specific therapeutic strategies<br />

can be adopted in patients with non-small cell<br />

lung cancer (NSCLC).<br />

Methods: EGFR TKI sensitive and resistant<br />

NSCLC cells were evaluated for drug-induced<br />

apoptosis and growth arrest. Cells were also<br />

tested for inhibition of downstream signalling<br />

and expression or drug-induced upregulation<br />

of Bcl-2 family members. Nude mice bearing<br />

sensitive and resistant NSCLC were i.v. injected<br />

with 7.4 MBq of 18F-FLT and then subjected to<br />

microPET/CT (eXplore Vista Pre-Clinical PET<br />

Scanner GE Healthcare) before and after treatment<br />

with reversible or irreversible EGFR TKIs.<br />

Results: We found that NSCLC bearing T790M<br />

mutations showed a persistent high uptake<br />

of 18 F-FLT after treatment with reversible inhibitors<br />

and lack of growth arrest. Treatment<br />

of the same animals with irreversible inhibitors<br />

caused a reduction of 18F-FLT uptake in<br />

tumors indicating the reversal of T790M mutation-dependent<br />

resistance. Conversely, NSCLC<br />

cells that were resistant due to dysregulation of<br />

Bcl-2 family members became sensitive to EGFR<br />

TKIs when treatment included Bcl-2 inhibitors.<br />

Conclusions: Resistance to EGFR TKI may<br />

be caused by multiple mechanisms. Molecular<br />

imaging with 18F-FLT may contribute to the<br />

selection of patients that may benefit from<br />

treatment with irreversible EGFR TKI inhibitors.<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

P-030<br />

poStEr<br />

CANCER from BENCH to BEDSIDE


P-031<br />

144<br />

WarSaW, poland May 26 – 29, 2010<br />

Synchronised Cardiac and Lung CT in Rodents Using the Mobile CT Scanner LaTheta<br />

Glowalla A. .<br />

Zinsser Analytic GmbH, Frankfurt, Germany<br />

a.glowalla@zinsser-analytic.com<br />

Introduction: Historically, in vivo CT images of<br />

the chest region of small animals were blurred and<br />

streaked due to movement artefacts caused by the<br />

heartbeat and breathing during scanning. However,<br />

synchronised Cardiac and Lung CT in rodents has the<br />

enormous potential to contribute greatly to research in<br />

heart and lung diseases. Using the mobile CT Scanner<br />

LaTheta, scanning artefacts are greatly reduced. Here<br />

we present scanning results of the chest region of mice<br />

and discuss their relevance in pre-clinical research.<br />

Methods: Manufacturers standard protocols for synchronised<br />

CT scanning were used to scan the chest<br />

region of anaesthetised mice with respiratory rates<br />

below 75bpm, as below:<br />

Synchronised CT scanning results for A) heart in systole phase<br />

and B) lung in mouse<br />

A) Cardiac Sync. Scan (systole + diastole)<br />

- scan range axial: heart, 15mm length<br />

- scan time: 13.1min<br />

B) Respiratory Sync. Scan<br />

- scan range axial: heart, 10mm length<br />

- scan time: 4.5min<br />

X-ray tube voltage: 50kV at 0.5mA; Pixel resolution:<br />

48um; Slice thickness: 192um; Contrast media: Iopamiron<br />

300, 1.5ml/h in 30g mouse given 10min.<br />

before scanning.<br />

imaging life<br />

A<br />

CT images were constructed using methods for reduction<br />

of motion artefacts and with focus on soft<br />

tissues.<br />

Results: CT images of heart (diastole and systole<br />

phases) and lung were acquired and reconstructed<br />

without common blurring and streaking effects.<br />

Conclusions: Synchronised scanning modes with<br />

LaTheta LCT -200 produce high-resolution CT<br />

cross-sections of heart and lung in living mice<br />

within a short time period. Similar methods are<br />

applicable also for rats. Since no additional sensor<br />

hardware is necessary, handling of the animal is as<br />

simple as for standard CT.<br />

The potential for the detection of lung diseases such<br />

as pulmonary fibrosis, pulmonary emphysema and<br />

also lung tumours has already been validated by<br />

manufacturer. With this technology LaTheta LCT<br />

-200 can also contribute to the studies of cardiovascular<br />

diseases in rodents.<br />

Acknowledgement:<br />

1. Manufacturer of LaTheta(TM): Aloka Co., Ltd., 6-22-1<br />

Mure, Mitaka-shi, Tokyo, 181-8622, Japan<br />

2. Distributor: Zinsser Analytic GmbH, Eschborner<br />

Landstr. 135, 60489 Frankfurt am Main, Germany<br />

B


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

Molecular imaging of neurovascular inflammation in a mouse model of focal cerebral<br />

ischemia using Ultra small Superparamagnetic Particles of Iron Oxide (USPIOs) targeted to<br />

vascular cell adhesion molecule-1 (VCAM-1)<br />

Montagne A. (1) , Gauberti M. (1) , Orset C. (1) , Macrez R. (1) , Rubio M. (1) , Raynaud J. S. (2) , Vivien D. (1) , Maubert E. (1) .<br />

(1) Inserm U919 UMR 6232 CNRS Ci-Naps, Caen, France<br />

(2) Experimental Imaging, MRI unit. Research Division. GUERBET, Roissy CdG, France<br />

amontagne@cyceron.fr<br />

Introduction: Among several mechanisms, brain<br />

inflammation is thought to have critical functions<br />

in lesion progression during acute and<br />

subacute stages of ischemic stroke. Accordingly,<br />

anti-inflammatory strategies are neuroprotective<br />

in preclinical studies [1]. However, to date, all<br />

clinical trials with anti-inflammatory agents have<br />

failed to improve clinical outcome in acute ischemic<br />

stroke patients. New tools for non-invasive<br />

monitoring of inflammation following stroke are<br />

needed to select patients who are more susceptible<br />

to benefit from anti-inflammatory treatment.<br />

Vascular cell adhesion molecule 1 (VCAM-1), an<br />

endothelial adhesion molecule, is overexpressed<br />

in the injured brain and is therefore thought to be<br />

a good target for the molecular imaging of inflammatory<br />

processes [2]. The aim of this study was<br />

to investigate the use of targeted USPIOs against<br />

VCAM-1 to follow neurovascular inflammation in<br />

a mouse model of in situ thromboembolic stroke<br />

with recombinant tissue-type plasminogen activator<br />

(rt-PA) induced reperfusion [3].<br />

Results: Our present data confirm an increase of<br />

vascular cell adhesion molecule-1 (VCAM-1) immunoreactivity<br />

(-ir) in the ischemic brain with<br />

a peak at 24 hours post-ischemia. Interestingly,<br />

V-CAM-1-ir was significantly increased in late<br />

rt-PA-thrombolyzed animals (4 hours after clot<br />

formation) compared to early (20 minutes) or<br />

unthrombolyzed mice. These data suggest that<br />

inflammation could be involved in the deleterious<br />

clinical outcome reported with late rt-PA<br />

mediated thrombolysis. Targeted USPIOs against<br />

VCAM-1 showed numerous signal voids in the ipsilateral<br />

side by 7T magnetic resonance imaging<br />

(MRI) at 24 hours post-ischemia and the use of<br />

an antibody directed against polyethylene-glycol<br />

(USPIOs coating) confirmed histologically our<br />

MRI analysis.<br />

Conclusions: Our data indicate that non invasive<br />

imaging of VCAM-1 with targeted USPIOs allows<br />

reliable imaging of brain inflammation after<br />

stroke. Such molecular MRI approaches could be<br />

used to evaluate inflammatory processes in stroke<br />

patients and thus to adapt therapy on an individual<br />

basis. Furthermore, molecular imaging of<br />

VCAM-1 could also provide valuable clinical information<br />

in other diseases involving inflammatory<br />

processes (atherosclerosis, multiple sclerosis …).<br />

References:<br />

1. Machado L et al; Stroke. 40;3028-3033 (2009).<br />

2. Hoyte L et al; J Cereb Blood Flow Metab.1-10 (2010).<br />

3. Orset C et al; Stroke. 38;2771-2778 (2007).<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

P-032<br />

poStEr<br />

MI in CARDIOVASCULAR DISEASE


WarSaW, poland May 26 – 29, 2010<br />

P-033 Scintigraphy with the use of 123 I-IL-2: a new promising tool for cardiovascular risk assessment<br />

in patients with high cardiovascular risk<br />

146<br />

Opalinska M. (1) , Hubalewska-Dydejczyk A. (1) , Stompor T. (2) , Krzanowski M. (3) , Mikołajczak R. (4) , Sowa-Staszczak A. (1) ,<br />

Karczmarczyk U. (5) , Glowa B. (1) , Kuśnierz-Cabala B. (6) , Pach D. (1) , Sułowicz W. (3) .<br />

(1) Nuclear Medicine Unit, Department of Endocrinology, Jagiellonian University Medical School, Krakow, Poland (2) Chair and Department of Nephrology,<br />

Hypertesiology and Internal Medicine, University of Warmia and Mazury , Olsztyn, Poland (3) Chair and Department of Nephrology, Jagiellonian University<br />

Medical School, Krakow, Poland (4) Radioisotope Center POLATOM, Otwock-Swierk, Poland (5) Departament of Radiopharmaceuticals, National Medicines<br />

Institute, Warsaw, Poland (6) Chair of Clinical Biochemistry, Jagiellonian University Medical School, Krakow, Poland<br />

mkal@vp.pl<br />

Introduction: Cardiovascular diseases are the<br />

main cause of deaths in general population<br />

(0,28%/year) whereas in selected population like<br />

in end-stage renal disease patients, mortality<br />

reaches even 20%. More than half of the cardiovascular<br />

events occur among the patients without<br />

any previous symptoms of cardiovascular disease,<br />

when atherosclerotic plaques obliterate less than<br />

50% of arterial lumen.<br />

The majority of widely available methods of cardiovascular<br />

risk estimation are based on arterial<br />

lumen assessment, estimation of atherosclerotic<br />

plaque size or on intima-media complex measurements.<br />

It means that those methods may by imprecise<br />

in the identification of plaques of the highest<br />

risk of rupture. For that reason, the methods<br />

which enable the visualization and estimation of<br />

the intensity of inflammatory process within atherosclerotic<br />

lesions, seem to play the most promising<br />

diagnostic role.<br />

The histopathological studies of the atherosclerotic<br />

plaque have revealed that activated lymphocytes<br />

T, which contain IL-2 receptors on their surfaces,<br />

comprise at least 20 % of inflammatory cells in unstable<br />

plaques. Those receptors can be identified<br />

with the use of scintigraphy with labeled IL-2.<br />

Methods: 10 patients (5 women, 5 men, aged 62,4<br />

± 10,4), with the highest cardiovascular risk, were<br />

chosen from 67 peritoneal dialysis cohort patients.<br />

All of them underwent 123 I-IL2 scintigraphy, coronary<br />

calcium scoring by CT and common carotid<br />

artery intima-media thickness assessment by USG.<br />

The levels of some atherogenic, inflammatory and<br />

calcium-phosphate indicators were measured. Target/non-target<br />

ratio of 123 I-IL-2 uptake in atherosclerotic<br />

plaque confirmed by carotid artery USG<br />

with IMT, calcium score result and concentration<br />

measured agents were compared.<br />

Results: In the performed scintigraphies increased<br />

focal 123 I-IL-2 uptake in 16/16 (100%) atherosclerotic<br />

plaques previously visualized by neck ultrasound<br />

was detected. Mean T/nT ratio of focal 123 I-<br />

imaging life<br />

IL-2 uptake within atherosclerotic plaques was<br />

3,15 ± 0,54 (median 3,22, range 2,0 – 3,6). The<br />

levels of the inflammatory and atherogenic factors<br />

and IMT (mean 0,975 ± 0,337 mm) were increased<br />

in the majority of patients. High positive correlation<br />

between 123I-IL-2 uptake within atherosclerotic<br />

plaques and IMT in corresponding artery<br />

was observed (R = 0,92, p = 0,01). Significantly<br />

higher 123 I-IL-2 uptake on scintigraphy in patients<br />

who developed cardiovascular event during observation<br />

period (average 36 months), compared<br />

to patients without cardiovascular event, was revealed<br />

(3,45 ± 0,22 vs 2,91 ± 0,54, p = 0,046). No<br />

statistically significant association was found between<br />

123 I-IL-2 uptake and the levels of measured<br />

agents, calcium score or classical cardiovascular<br />

risk factors.<br />

Conclusions: Scintigraphy with the use of labeled<br />

123 I-IL-2 enables the visualization of inflamed<br />

atherosclerotic (vulnerable) plaque within common<br />

carotid arteries in end-stage renal disease<br />

patients.<br />

Quantitative results of the carotid arteries scintigraphy<br />

with 123 I-IL-2 correlate with the results of<br />

IMT and the risk of cardiovascular event during 3<br />

years of follow-up.<br />

Acknowledgement: This work is supported by the<br />

Polish Committee for Scientific Research (KBN)<br />

within Research Project 2 P05B 003 28<br />

References:<br />

1. Annovazzi A et al; 99mTc-interleukin-2 scintgraphy for<br />

the in vivo imaging of vulnerable atheroslerotic plaque.<br />

Eur J Nucl Med Mol Imaging 2006; 33: 117 – 126.<br />

2. Buyukhatipoglu H et al; Inflammation as a risk factor<br />

for carotid intimal-medial thickening, a measure of<br />

subclinical atherosclerosis in haemodialysis patients:<br />

the role of chlamydia and cytomegalovirus infection.<br />

Nephrology 2007; 12: 25 – 32.


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

MEMRI-DTI study of focal transient ischemia in immature rat brain<br />

Dupont D. , Bogaert-Buchmann A. , Sebrie C. , Gillet B. .<br />

IR4M , UMR 8081 CNRS-Univ. Paris-Sud, ORSAY, France<br />

damien.dupont1@u-psud.fr<br />

Introduction: The aim of the study was to investigate<br />

anatomo-functional changes in immature rat brain<br />

after focal transient cerebral ischemia occurred<br />

when they were 7-day-old. The MRI study was carried<br />

out 14 days after ischemia by serial MEMRI<br />

combined with DTI.<br />

Methods: Focal ischemia was induced in P7 rats as<br />

previously described [1], and the size of the lesion<br />

was pointed out by DWI 3h after ischemia. MRI<br />

studies were carried out at 7T on P21 rats (n=5), 14<br />

days after focal transient ischemia and compared to<br />

normal P21 rats (n=5). DTI sequence used was DTI-<br />

EPI (2 b-values=500/1000s/mm2; 30 directions;<br />

resolution=0.156*0.156*1 mm3). For the MEMRI<br />

experiments, 160nl of a 50mM isotonic manganese<br />

chloride aqueous solution (pH=7.3) was injected<br />

in S1 at 1.5mm below the dura, at a rate of 20nl/<br />

min. T1-weighted experiments were achieved at 3,<br />

5, 7 and 24h post-injection, using 3D MP-RAGE<br />

sequence (resolution=0.156*0.156*0.5mm3; TR/<br />

TE=15/4.5ms, α=20°; Ti=1s). The manganese containing<br />

voxels were defined as hyper intense voxels<br />

and selected as those with a significant higher signal<br />

(p


148<br />

WarSaW, poland May 26 – 29, 2010<br />

P-035 A clinically relevant model of in situ embolic stroke in the anesthetized monkey (macaca<br />

mulatta): long-term electrophysiological and mri analyses<br />

Gauberti M. (1) , Guedin P. (1) , Etard O. (2) , Diependaele A.S. (2) , Chazalviel L. (3) , Lamberton F. (3) , Vivien D. (1) , Young A. (1) ,<br />

Agin V. (1) , Orset C. (1) .<br />

(1) INSERM, Caen, France<br />

(2) CHU Caen, France<br />

(3) CNRS, France<br />

gauberti@cyceron.fr<br />

Introduction: The lack of relevant stroke models in<br />

large animals is a limitation for the development of<br />

innovative therapeutic/diagnostic approaches. The<br />

aim of the present study was to develop an original<br />

and clinically relevant pre-clinical model of embolic<br />

stroke in the monkey.<br />

Methods: During full physiological and biochemical<br />

monitoring, six monkeys underwent enucleation,<br />

the right MCA was exposed and alpha-thrombin<br />

was injected into the MCA. The monkeys were subjected<br />

to somatosensory evoked potentials (SEPs)<br />

and MRI studies (T2, FLAIR, DWI, PWI and MRA)<br />

prior to, and following the acute (2 h) and chronic<br />

stages (24 h to 3 months) of stroke.<br />

imaging life<br />

Results: This feasibility study showed that, it is possible<br />

to induce ischemic lesions in the MCA territory<br />

of the monkey following the direct injection of<br />

thrombin into the lumen of the M1 segment of the<br />

MCA. This procedure leads to cortical or subcortical<br />

ischemic lesions and to a persistent impairment<br />

of somatosensory responses as evidenced by MRI<br />

and SEPs data, respectively.<br />

Conclusions: In situ induction of an endogenous fibrin<br />

clot together with the lack of mortality make<br />

this original model of stroke in large non-human<br />

primates, highly relevant to determine the effectiveness<br />

of drug administration including thrombolytic<br />

therapy and to validate new imaging procedures.


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

Automated radiosynthesis of [ 18 F]MPPF derivatives for imaging 5-HT 1A receptors<br />

Goblet D. (1) , Thonon D. (1) , Plenevaux A. (1) , Defraiteur C. (2) , Wouters L. (2) , Franci X. (2) , Luxen A. (1) .<br />

(1) University of Liege, Liege, Belgium<br />

Z2) GE Healthcare Diagnostic Imaging, Belgium<br />

david.goblet@ulg.ac.be<br />

Introduction: Dysfunction of the central serotoninergic<br />

system is implicated in numerous neurodegenerative<br />

disorders such as Alzheimer’s disease,<br />

dementia, depression, anxiety, schizophrenia, and<br />

Parkinson’s disease. 5-HT 1A receptors are involved<br />

in several physiological functions including sleep,<br />

mood, neurogenesis and learning [1]. Consequently,<br />

there have been huge efforts to find ligands for this<br />

receptor. [ 11 C]WAY-100635 is a high affinity radioligand<br />

used for quantifying 5-HT 1A receptors with<br />

positron emission tomography. An 18 F-labeled radioligand<br />

would be advantageous because of higher<br />

specific activity and physical/nuclear properties<br />

(t 1/2 = 109 min, 97% of positron decay and positron<br />

energy of 635 keV maximum). [ 18 F]MPPF, a selective<br />

5-HT 1A antagonist derived from WAY-100635,<br />

is currently one of the most successful PET ligands<br />

used for 5-HT 1A receptor imaging [2]. However the<br />

affinity is lower than WAY-100635 and the amount<br />

of [ 18 F]MPPF reaching the brain is relatively low,<br />

as MPPF is a substrate for P-glycoprotein [3].<br />

Methods: In order to improve the brain uptake of<br />

the radiotracer, a desmethylated analog has been<br />

developed in our lab and preliminary in vitro<br />

studies show positive results [4]. Nevertheless,<br />

the radiosynthesis takes place in two steps, as the<br />

removal of a protecting group is needed. A one<br />

step procedure with a MPPF derivative could be<br />

of very great interest. We have synthesized many<br />

MPPF derivatives in our lab (modification on the<br />

phenylpiperazine moiety) and developed an automated<br />

radiosynthesis procedure for the production<br />

of these radiotracers. [ 18 F]MPPF was chosen<br />

as the model compound. We used a GE Healthcare<br />

FASTlab TM module and made modifications to the<br />

[ 18 F]FDG synthesis sequence and cassette. [ 18 F]<br />

MPPF was synthesized by coupling of [ 18 F]FBA<br />

with the corresponding amine. After coupling, the<br />

crude solution was diluted with water and passed<br />

through a tC 18 cartridge for prepurification. After<br />

elution, the [ 18 F]MPPF was purified by semi-preparative<br />

HPLC.<br />

Results: Total synthesis time, including purification<br />

was approximately 100 min. [ 18 F]FBA and [ 18 F]MPPF<br />

were obtained at a corrected yield of 60% (n=20)<br />

and 45% (n=5) respectively. The radiochemical purity,<br />

checked by radio-TLC and UPLC, was >95%.<br />

Conclusions: We have developed an automated<br />

method for [ 18 F]MPPF (and derivatives) production<br />

using a commercial synthesizer (FASTlab TM<br />

from GE Healthcare) and a conventional HPLC<br />

system resulting in good yields and high (radio)<br />

chemical purity. By simply switching the vial containing<br />

the modified amine, an 18 F-labeled MPPF<br />

derivative could be obtained. Radiosynthesis is<br />

still under optimization and the radiotracers<br />

synthesized need to be tested as suitable 5-HT 1A<br />

radioligands.<br />

Acknowledgement: This work was supported by<br />

the Fondation Rahier of the University of Liege.<br />

This work was supported by a GE Healthcare Diagnostic<br />

Imaging grant.<br />

References:<br />

1. Filip M., Bader M. et Al, Pharmacol Rep. 2009 Sep-Oct;<br />

61(5):761-77<br />

2. Aznavour N, Zimmer L. Et Al, Neuropharmacology.<br />

2007 Mar; 52(3):695-707<br />

3. Laćan G., Plenevaux A. et Al, Eur J Nucl Med Mol<br />

Imaging. 2008 Dec;35(12):2256-66<br />

4. Defraiteur C., Plenevaux A. et Al., Br J Pharmacol. 2007<br />

Nov; 152(6):952-8<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

P-036<br />

poStEr<br />

NEUROIMAGING from BENCH to BEDSIDE


P-037<br />

150<br />

WarSaW, poland May 26 – 29, 2010<br />

The vitamine E analogue CR6 protects against the long-term microstructure damage induced<br />

by MCA occlusion: a longitudinal Diffusion Tensor Imaging study<br />

Justicia C. (1) , Soria G. (1) , Tudela R. (2) , Van Der Linden A. (3) , Messeguer A. (4) , Planas A. (5) .<br />

(1) Investigaciones Científicas (CSIC), Institut d‘Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain<br />

(2) CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Group of Biomedical Imaging of the University of Barcelona, Spain<br />

(3) University of Antwerp, Antwerp, Belgium<br />

(4) Institut de Química Avançada de Catalunya (IQAC), CSIC, ,Spain<br />

(5) Institut d‘Investigacions Biomèdiques de Barcelona (IIBB)-Consejo Superior de Investigaciones Científicas (CSIC), Institut d‘Investigacions Biomèdiques<br />

August Pi i Sunyer (IDIBAPS), Spain<br />

cjmfat@iibb.csic.es<br />

Introduction: Oxidative and nitrosative stress are<br />

targets for intervention after ischemia/reperfusion.<br />

CR-6 is a synthetic analogue of vitamin-E, with the<br />

additional capacity to scavenge nitrogen-reactive<br />

species. Recently it has been demonstrated that<br />

CR-6 exerts a protective action against cerebral ischemia/reperfusion<br />

injury. The aim of this study was<br />

to investigate whether CR-6 can protect the brain<br />

microstructure after brain ischemia as assessed by<br />

longitudinal Diffusion Tensor Imaging (DTI).<br />

Methods: Sprague–Dawley rats had the middle<br />

cerebral artery occluded for 90 mins to induce brain<br />

ischemia. CR-6 (100 mg/kg) or vehicle (oliva oil)<br />

was orally administered at 2 and 8 h after ischemia<br />

onset. Longitudinal MRI scans were performed under<br />

isofluorane anaesthesia in a BioSpec 70/30 horizontal<br />

animal scanner (Bruker BioSpin, Ettlingen,<br />

Germany), equipped with a 12 cm inner diameter<br />

actively shielded gradient system (400 mT/m). Receiver<br />

coil was a phased-array surface coil for rat<br />

brain. Animals were scanned before surgery and<br />

1, 3 and 5 weeks later. The lesion was monitored<br />

by T2 mapping of coronal slices acquired with<br />

a MSME sequence by applying 16 different echo<br />

times (TE), repetition time (TR) = 4764 ms, Field of<br />

view (FOV) = 40 x 40 x 21 mm3, matrix size = 256<br />

x 256 x 21 pixels, resulting in a spatial resolution of<br />

0.156 x 0.156 mm in 1.00 mm slice thickness. DTI<br />

images were acquired by using a EPI DTI sequence<br />

applying TR = 3800 ms, TE = 30.85 ms, 4 segments,<br />

b-value = 1000, 30 different diffusion directions, 5<br />

A0 images, slice thickness = 1 mm, number of slices<br />

= 14, FOV = 2.0 x 2.0 x 1.4 mm3, matrix size = 96<br />

x 96 x 14 pixels, resulting in a spatial resolution of<br />

0.21 × 0.21 mm in 1 mm slice thickness. T2 maps<br />

were analysed using ImageJ. DTI maps of the tensor<br />

diffusivities, fractional anisotropy (FA), apparent<br />

diffusion coefficient (ADC), axial diffusivity (λ =<br />

λ1) and radial diffusivity (λ = [λ2+ λ3/2]), were<br />

calculated using Paravision 5.0 software (Bruker<br />

Biospin, Etlingen, Germany) and custom programs<br />

written in Matlab (The MathWorks, Inc., Natick,<br />

MA, USA). ROIs were individually drawn for each<br />

imaging life<br />

subject and hemisphere, by an experienced neurobiologist<br />

blinded to the treatments, overlaying the<br />

MRI images with the digital Paxinos and Watson<br />

rat brain atlas.<br />

Results: DTI diffusion indices, particularly axial and<br />

radial diffusivities, demonstrated that microstructure<br />

was better preserved after brain ischemia in<br />

CR6-treated animals than in animals receiving the<br />

vehicle. The latter showed a significant increase of<br />

MD, axial and radial diffusivities in the ipsilateral<br />

anteroventral thalamus, caudate putamen, globus<br />

pallidus and internal capsula 3 and 5 weeks after<br />

ischemia versus pre-scan. However, CR6 treated<br />

animals did not show such increase of DTI indexes<br />

in any of the ROIs analysed revealing preserved microstructure<br />

of the brain tissue.<br />

Conclusions: This work demonstrates that transient<br />

cerebral ischemia induces long-term microstructure<br />

alterations of brain tissue that are detected by DTI<br />

magnetic resonance imaging technique, and that the<br />

vitamin-E analogue CR6 attenuates these alterations.<br />

Acknowledgement: S and RT are supported by CSIC<br />

(JaeDoc) and CIBER-BBN, respectively. We acknowledge<br />

the European Erasmus program supporting<br />

Sofie de Prins and Stephan Cools that helped in this<br />

study. Work supported by national grants (SAF2008-<br />

04515-C02-01, FIS PI081880), European Network<br />

of Excellence DiMI (LSHB-CT-2005-512146), and<br />

FP7/2007-2013 project (grant agreement n°201024).


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

Design and synthesis of fluorescent probes for serotonin 5-HT1A receptors<br />

Leopoldo M. (1) , Lacivita E. (1) , Berardi F. (1) , Perrone R. (1) , Jafurulla M. (2) , Saxena R. (2) , Rangaraj N. (2) , Chattopadhyay A. (2) .<br />

(1) Università degli studi di Bari “A. Moro”, Bari, Italy<br />

(2) Council of Scientific and Industrial Research, Hyderabad, India<br />

leopoldo@farmchim.uniba.it<br />

Introduction: The serotonin1A<br />

receptor subtype (5-HT1A)<br />

OCH3 belongs to the large family of<br />

5-HT receptors that comprises<br />

at least 14 receptor subtypes.<br />

5-HT1A receptor is likely the<br />

most extensively studied serotonin<br />

receptor. The 5-HT1A<br />

receptor has been initially<br />

N<br />

N<br />

implicated in anxiety and de-<br />

O<br />

pression. Recent studies have<br />

O<br />

evidenced its implication in<br />

neuroprotection, cognitive<br />

N<br />

impairment, and pain [1].<br />

Many information are avail-<br />

1<br />

able about the molecular pharmacology of 5-HT1A<br />

receptor. However, the study of 5-HT1A pharmacology<br />

at the single cell and single molecule level<br />

by fluorescence-based techniques has not been possible<br />

due to the lack of an effective 5-HT1A fluorescent<br />

ligands. We here describe design, synthesis<br />

and preliminar pharmacological evaluation of two<br />

fluorescent probes for 5-HT1A receptor.<br />

Methods: The new fluorescent probes have been<br />

designed following a classic approach [2]: a pharmacophore<br />

moiety, selected from the literature, has<br />

been conjugated through a linker to a fluorescent<br />

dye. One red-emitting and one near infrared dye<br />

were selected because such excitation wavelengths<br />

cause reduced light scattering and do not cause cell<br />

damage. The synthesis and purification of the target<br />

compounds was accomplished by standard methods.<br />

The compounds 1 and 2 underwent binding assays to<br />

test their ability to displace [3H]-8-OH-DPAT from<br />

5-HT1A receptors overexpressed in HEK-298 cell<br />

membranes. The fluorescent ligand 1 was evaluated<br />

in visualization experiments of 5-HT1A receptor on<br />

CHO cells by confocal laser scanning microscopy.<br />

Results: Compounds were obtained in good yields<br />

and in good quantities to allow further biological<br />

studies. Both compounds 1 and 2 possessed good<br />

5-HT1A receptor affinities (Ki = 35 nM and 68.9<br />

O<br />

N +<br />

Br- O<br />

OCH 3<br />

N N +<br />

Cl- NaO 3S SO 3Na<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

N<br />

N<br />

nM, respectively). The target compounds displayed<br />

the same fluorescent properties as the corresponding<br />

fluorophores. In visualization experiments by confocal<br />

laser scanning microscopy compound 1 was able<br />

to label 5-HT1A receptors on CHO cells overexpressing<br />

the receptor. This interaction revealed to be specific<br />

since the compound was unable to bind wildtype<br />

CHO cells and also because 5-HT significantly<br />

displaced competitively 1 from the binding site.<br />

Conclusions: This study indicates that fluorescent<br />

probes for 5-HT1A receptor can be successfully<br />

designed on the basis of knowledge of structure-<br />

activity relationships of 5-HT1A receptor agents.<br />

The 5-HT1A fluorescent ligand that have been identified<br />

possess suitable characteristic for visualization<br />

of 5-HT1A receptors.<br />

References:<br />

1. Lacivita E et al; Curr Top Med Chem. 8:1024-1034<br />

(2008)<br />

2. Leopoldo M et al: Drug Disc Today 14:706-712 (2009)<br />

H<br />

N<br />

2<br />

S<br />

O<br />

P-038<br />

poStEr<br />

NEUROIMAGING from BENCH to BEDSIDE


152<br />

WarSaW, poland May 26 – 29, 2010<br />

P-039 Comparative evaluation of cerebral blood flow in a rat model of cerebral ischemia using 15 O-<br />

H 2 0 positron emission tomography and 99m Tc-HMPAO single-photon emission tomography<br />

Martin A. (1) , Boisgard R. (1) , Gervais P. (2) , Thézé B. (1) , Vuillemard C. (2) , Tavitian B. (1) .<br />

(1) CEA, DSV, I²BM, SHFJ, Laboratoire Imagerie Moléculaire Expérimentale; INSERM U803, Orsay, France<br />

(2) CEA, DSV, I²BM, SHFJ, France<br />

abraham.martin-munoz@cea.fr<br />

Introduction: Ischemic stroke occurs when cerebral<br />

blood flow is interrupted in a specific region of the<br />

brain. After ischemic stroke, cerebral parenchyma<br />

suffers perfusion changes over time which have been<br />

correlated with biological processes as compensatory<br />

growth of blood vessels to supply metabolic demand<br />

[1], underlying inflammation [2] and angiogenesis<br />

[3].Thereafter, a precise understanding of the cerebral<br />

blood flow evolution after ischemic stroke is essential<br />

for the comprehension of cerebral ischemia physiopathology.<br />

Here, a comparative imaging evaluation<br />

of cerebral blood flow using PET and SPECT during<br />

and after cerebral ischemia was performed in rats.<br />

Methods: [ 15 O] H20 and [ 99m Tc] HMPAO were used<br />

in a rat model of 2 hours transient middle cerebral<br />

artery occlusion (tMCAO) during occlusion, during<br />

early reperfusion and later on at 1, 2, 4 and 7 days.<br />

The tissue was analized ex vivo using histological<br />

(HE) and immunohistochemistry (CD31).<br />

Results: In vivo PET imaging showed a significant<br />

decrease of the ipsilateral or lesioned area versus<br />

contralateral ratios during the occlusion in<br />

relation to control, days 4 and 7 after reperfusion<br />

(P


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

Comparison of dopamine transporter density in Parkinson’s disease patients with and<br />

without autonomic dysfunction using F-18 FP-CIT PET/CT<br />

Park E. , Park K.W. , Hwang Y.M. , Oh S.Y. , Choe J.G. .<br />

Korea University Anam Hospital, Korea University College of Medicine, Seoul<br />

angela_ekpark@yahoo.co.kr<br />

Introduction: Autonomic dysfunction is a common<br />

non-motor feature of Parkinson’s disease (PD)<br />

which can severely impair the life quality of PD patients.<br />

However, evaluation of striatal presynaptic<br />

dopaminergic function with SPECT or PET imaging<br />

using various radiotracers has been limited in<br />

the differential diagnosis of parkinsonism and in<br />

the severity assessment of Parkinson’s disease. We<br />

compared the dopamine transporter (DAT) density<br />

of PD patients with and without autonomic dysfunction<br />

using F-18 FP-CIT PET/CT, which has been<br />

approved recently by Korean FDA for clinical uses.<br />

Methods: Twenty clinically diagnosed PD patients<br />

(mean age 68.6±8.1 y, M:F=5:15) and 8 age-matched<br />

healthy normal controls (65.1±4.7 y, M:F=2:6) were<br />

studied with F-18-FP-CIT PET/CT. Among 20 PD<br />

patients, 10 had no significant autonomic dysfunction<br />

(PD-AD) while the other 10 had autonomic dysfunction<br />

(PD+AD). F-18-FP-CIT PET/CT images were<br />

obtained 120 minutes after injection of 185 MBq<br />

F-18-FP-CIT. Semi-quantitative analysis was performed<br />

using manual ROI method. A DAT parameter<br />

V3”, a measure directly related to the density of DAT,<br />

was calculated in striatal regions as (striatal ROI–<br />

cerebellar ROI mean radioactivity)/cerebellar ROI<br />

mean radioactivity on F-18-FP-CIT PET/CT images.<br />

Results: PD patients demonstrated significantly<br />

lower DAT V3” in the striatum (2.29±0.69), caudate<br />

nucleus (2.56±0.67), and putamen (1.99±0.77)<br />

than those of healthy normal controls (4.02±0.55,<br />

3.88±0.54 and 4.16±0.61, respectively) (p


154<br />

WarSaW, poland May 26 – 29, 2010<br />

P-041 Synthesis of tosylate and mesylate precursors for one-step radiosynthesis<br />

of [ 18 F]FECNT<br />

Pijarowska J. , Jaron A. , Mikolajczak R. .<br />

Insitute of Atomic Energy POLATOM, Otwock, Poland<br />

j.pijarowska@polatom.pl<br />

Introduction: Dopamine transporter (DAT) is critical<br />

to the regulation of dopamine neurotransmission and<br />

is decreased by Parkinson’s disease. Several tropane<br />

analogues of cocaine have been developed and used in<br />

PET studies to evaluate the physiology and pharmacology<br />

of the dopamine transporter (DAT). However,<br />

low selectivity and unfavourable kinetics of most of the<br />

compounds limit their use in quantitive PET studies.<br />

The fluorine-18 labelled ligand 2-beta-carbomethoxy-3-<br />

-beta-(4-chlorophenyl)-8-(2-fluoroethyl)-nortropane<br />

(FECNT) has promising properties and appears to be<br />

an excellent imaiging PET agent. The development of<br />

automated [18F]FECNT synthesis system is a crucial<br />

because high amounts of radioactivity and availability<br />

of radiotracer for multiple PET study is necessary. A<br />

semi-automated synthesis of [18F]FECNT based on<br />

the two-steps has been developed at 16% decay corrected<br />

yield [1]. We hypothesize that N-[18F]fluoroalkylnortropane<br />

analogs could be synthesized at hight<br />

yield by direct [18F]fluorination from appropriate precursors:<br />

N-tosylate and N-mesylate derivatives. This<br />

compounds and non-radioactive FECNT as a standard<br />

in order to characterize [ 18 F]FECNT on HPLC are<br />

synthesized in accordance with requirements for Investigational<br />

Medicinal Product (IMP).<br />

Methods: The synthetic approach which we adopted<br />

based upon the published procedures [2,3]<br />

with some modifications. The essential feature<br />

of this route was the reaction of Grignard reagent<br />

with the critical intermediate anhydroecognine<br />

methyl ester, which was obtained from<br />

cocaine hydrochloride by hydrolysis in HCl and<br />

esterification with methanol. 3-β-substituted tropane<br />

derivative obtained in Grignard reaction<br />

was subjected to demethylation, as described [4].<br />

Non-radioactive FECNT was prepared by direct<br />

N-(2- fluoroethyl) alkylation of analytically pure<br />

3-β-substituted nortropane precursor. The alkylating<br />

agent 2-fluoroethyl brosylate was prepared<br />

from 2-fluoroethanol and 4-bromobenzenesulfonoyl<br />

chloride. The crude product was purified<br />

by recrystallization. The tosylate (TsOECNT)<br />

and mesylate (MsOECNT) precursors were<br />

imaging life<br />

synthetised from 3-β-substituted nortropane precursor<br />

in two steps by N-hydroxyethylation with<br />

2-bromoethanol and subsequent tosylation of the<br />

obtained alcohol with appropriate anhydride. The<br />

crude products were purified by preparative HPLC.<br />

Results: In the present study we investigated and<br />

optimized synthetic route of non-radioactive<br />

FECNT, tosylate and mesylate analogs. Overall<br />

production yield were 74% for FECNT, 58%<br />

for TsOECNT and 81% for MsOECNT synthesis<br />

and a purity of this products were over<br />

99% measured by the analytical HPLC(UV, 220<br />

nm). The 1 H NMR and MS analysis confirmed<br />

a structure of this compounds.<br />

Conclusions: The synthesis method of tosylate<br />

and mesylate precursors were established and an<br />

automated radiosynthesis of [18F]FECNT will be<br />

evaluated. We expect that the new one–step method<br />

will provide a facile and reliable procedure<br />

for [18F]FECNT preparation in routine clinical<br />

applications.<br />

Acknowledgement: This work is supported by DiMI,<br />

LSHB-CT-2005-512146<br />

References:<br />

1. Voll R.J. et al. Appl. Rad. Isot. 2005 (63) 353<br />

2. Zirkle C. L et al. J. Org. Chem. 1962 (34) 1269<br />

3. Clarke R. L. et al. J. Med. Chem. 1973 (16) 1260<br />

4. Meegalla S. K. et al. J. Med. Chem. 1997 (40) 9


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

Sex differences inDopamine D 2 Receptor Occupancy in the Amygdala using AMPT with PET<br />

and [ 18 F]fallypride<br />

Riccardi P. (1) , Park S. (2) , Carroll X. (1) , Anderson S. (3) , Benoit D. (2) , Li R. (2) , Bauernfeind A. (4) , Schmidt D. (2) .<br />

(1) Mercer University, Macon, United States<br />

(2) Vanderbilt university, United States<br />

(3) Georgia State University ,United States<br />

(4) George Washington university, United States<br />

riccardip@aol.com<br />

Introduction: Dopaminergic neurotransmission<br />

plays an important role in many psychiatric disorders<br />

which show sex differences in incidence, clinical<br />

course, and treatment outcomes. We examined<br />

whether PET studies using [ 18 F]fallypride performed<br />

prior to and following alphamethylparatyrosine<br />

(AMPT) administration could be used to estimate<br />

sex differences in baseline Dopamine D 2 receptors<br />

(DAD2r) occupancy.<br />

Methods: Four females and four males normal subjects<br />

were recruited with no history of psychiatric,<br />

neurological or medical illness. PET studies were<br />

performed using a GE Discovery LS PET scanner<br />

with 3-D emission acquisition and transmission<br />

attenuation correction. [ 18 F]fallypride PET scans<br />

(5.0 mCi, specific activity > 2,000 Ci/mmol) were<br />

performed prior to and following AMPT administration<br />

over 26 hours. Serial scans were obtained<br />

for 3.5 hours. Blood samples for HVA plasma levels<br />

were collected.<br />

Results: Analysis of variance of the ROI data with<br />

treatment status, region, sex and laterality as factors<br />

revealed significant effects of sex in the right<br />

(F = 9.403, P = 0.002) and left amygdala (F = 3.486,<br />

P = 0.05). No difference in b.p was found in the<br />

left amygdala at baseline, but significant differences<br />

were determined at baseline and post treatment in<br />

the right amygdala (p=0.005 and p=0.002) and post<br />

treatment in the left amygdala (p=0.019) with females<br />

demonstrating lower levels of D 2 /D 3 receptors<br />

compare to males. Effect of treatment was observed<br />

in females in the left amygdala with a percentage of<br />

change of 6.7%.<br />

Conclusions: The amygdala has been shown to be<br />

functionally asymmetrical in animals and humans,<br />

is involved in stress and emotion processing, and is<br />

importantly modulated by dopamine. We report here<br />

for the first time a sex differences in the occupancy<br />

of DA D 2 r by dopamine in vivo in the amygdala. Previous<br />

AMPT administration and [18F]fallypride PET<br />

studies to assess the baseline occupancy of DAD 2r by<br />

endogenous dopamine in-vivo reported no significant<br />

effects of treatment in the amygdala when males and<br />

females were grouped together. The results of this<br />

study underscore the importance of considering sex<br />

differences in the context of DAD 2r availability.<br />

Acknowledgement: Funding for this reaserch<br />

was provided by a NIH grant entltled “PET imaging<br />

of Extrastriatal Dopamine levels” NIMH 5RO1<br />

MH6898-03<br />

References:<br />

1. Verhoeff NP, Kapur S, Hussey D et al (2001). A simple<br />

method to measure baseline occupancy of neostriatal<br />

dopamine D2 receptors by dopamine in vivo in healthy<br />

subjects. Neuropsychopharmacology 25; 213-223<br />

2. Riccardi P., Baldwin R, Salomon R., et al. (2008).<br />

Estimation of baseline Dopamine D2 receptor<br />

occupancy in Striatum and extrastriatal regions in<br />

humans with Positron emission tomography with<br />

[18F]fallypride.<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

P-042<br />

poStEr<br />

NEUROIMAGING from BENCH to BEDSIDE


156<br />

WarSaW, poland May 26 – 29, 2010<br />

P-043 Manual versus automatic delineation of VOIs for analysis of nuclear medicine images<br />

Svarer C. (1) , Hammers A. (2) , Heckemann R. A. (2) , Knudsen G.M. (1) .<br />

(1) Neurobiology Research Unit, Rigshospitalet,, Copenhagen, Denmark<br />

(2) The Neurodis Foundation, Lyon, France<br />

csvarer@nru.dk<br />

Introduction: Quantification and analysis of nuclear<br />

medicine images is performed by visual inspection in<br />

most cases. More objective and less observer-dependent<br />

analysis often requires automatic identification<br />

of volumes of interest (VOIs) in the images. In this<br />

study we investigated the levels of precision achievable<br />

with manual delineation on high resolution MR<br />

images and compared them to the precision of a fully<br />

automatic method.<br />

Methods: The method described in Svarer et al(1) has<br />

been refined to handle neighborhood voxels in labeled<br />

template volumes using a maximum-probability algorithm.<br />

We applied it to 19 MR data sets, each with 67<br />

manually delineated VOIs(2). To test the precision of<br />

the automatic algorithm, we performed a leave-oneout<br />

cross-validation-comparison: using each data set<br />

in turn as the target, we automatically generated two<br />

independent VOI sets by randomly bisecting the template<br />

pool, leading to two independent segmentations<br />

of each target by nine atlases each, and calculated the<br />

average VOI voxel overlap (intersection divided by<br />

average labeled volume). To test the reproducibility/<br />

precision of the manually delineated VOI sets, a third<br />

VOI set was generated for each target using the entire<br />

template pool but itself, and its overlap with the<br />

manual VOI set were calculated.<br />

Results: The voxel overlap for the two automatically<br />

generated VOI sets was 88.3±5.2%, whereas the voxel<br />

overlap between the automatically generated and the<br />

manually delineated VOI set was 76.3±9.8%.<br />

Conclusions: Of the 24% non-overlap between manual<br />

and automatic methods approx. 12 percentage points<br />

may be attributable to variation in the automatic method<br />

using different VOI template sets whereas precision<br />

in manual delineation of the VOIs may largely underlie<br />

the other half of the difference between manual and<br />

automatically generated VOI sets. This variation will<br />

be removed using an automatic approach based on<br />

multiple template sets.<br />

Acknowledgement: Supported by The Lundbeck Foundation,<br />

Rigshospitalet, and the Danish Medical Research<br />

Council. These studies were funded in part by<br />

the EC - FP6-project DiMI, LSHB-CT-2005-512146.<br />

imaging life<br />

References:<br />

1. Svarer C, Madsen K, Hasselbalch SG, Pinborg LH,<br />

Haugbøl S, Frøkjaer VG, Holm S, Paulson OB, and<br />

Knudsen GM (2005) MR-based automatic delineation<br />

of volumes of interest in human brain PET images<br />

using probability maps. Neuroimage 24: 969-979.<br />

2. Hammers A, Allom R, Koepp MJ, Free SL, Myers R,<br />

Lemieux L, Mitchell TN, Brooks DJ, and Duncan JS<br />

(2003) Three-dimensional maximum probability atlas<br />

of the human brain, with particular reference to the<br />

temporal lobe. Hum Brain Mapp 19: 224-247.


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

PET Amyloid and Tau Ligand [18F]FDDNP uptake in early Alzheimer disease<br />

Tauber C. (1) , Beaufils E. (1) , Kepe V. (2) , Vercouillie J. (1) , Venel Y. (1) , Baulieu J.L. (1) , Barrio J. (2) , Hommet C. (1) , Camus V. (1) ,<br />

Guilloteau D. (1) .<br />

(1) UMRS INSERM U930 - CNRS ERL3106 - University of Tours, France<br />

(2) Dpt of Molecular and Medical Pharmacology, D. Geffen School of Medecine, UCLA, United States<br />

clovis.tauber@univ-tours.fr<br />

Introduction: The PET tracer [18F]FDDNP specifically<br />

binds amyloid-beta plaques and tau neurofibrillary<br />

tangles, which are typical in the development<br />

of the Alzheimer disease (AD). We evaluated<br />

the feasibility of a non-invasive quantification of<br />

[18F]FDDNP to detect these proteins and discriminate<br />

early AD patients from healthy controls (HC).<br />

This abstract presents the preliminary results on the<br />

currently included cohort of 4 AD and 2 controls.<br />

Methods: All the potential subjects underwent Neuropsychological<br />

Assessment (MMSE, Trail Making<br />

Test, Semantic Fluency Task, Free and Cued<br />

Recall Test, 80 item Boston Naming Test). Patients<br />

meeting research criteria for AD and cognitively<br />

normal controls underwent PET imaging with<br />

2-(1-{6-[(2-[F-18]fluoroethyl) (methyl)amino]-2naphthyl}ethylidene)malononitrile<br />

(FDDNP) and<br />

[18F]FDG. All subjects underwent MR 3D axial<br />

T1 weighted imaging. FDDNP distribution volume<br />

ratios (DVR) parametric images were generated using<br />

Logan graphical analysis with the cerebellum<br />

grey matter as a reference region. Regions of interest<br />

(ROIs) were defined in the Posterior Cingulate,<br />

Parietal, Frontal and Temporal regions. The DVR<br />

scores were measured as the mean DVR values of<br />

each of these regions. A global score was calculated<br />

as the mean DVR scores of all these regions.<br />

Results: All the AD patients had positive FDDNP<br />

scans by visual inspection, while all the HC had negative<br />

scans. Global values of the FDDNP-PET binding<br />

were significantly higher (p


158<br />

WarSaW, poland May 26 – 29, 2010<br />

P-045 Cryogenic brain injury as a model of brain trauma: Use of GFAP-luc mice to assess GFAP<br />

expression as an indication of neural injury<br />

Van Beek E., Blankevoort V., Snoeks T., Kaijzel E., Löwik C.W.G.M. .<br />

Leiden University Medical Center, The Netherlands<br />

e.r.van_beek@lumc.nl<br />

Introduction: Applying a cryogenic lesion of the<br />

cerebral cortex provides a simple and highly reproducible<br />

model of brain trauma without entering the<br />

intracranial cavity. In the present study we assessed<br />

and mapped the sequence of events that occur following<br />

induction of a cryogenic brain lesion using<br />

transgenic glial fibrillary acidic protein (GFAP)-luc<br />

reporter expressing mice. In these mice, the GFAP<br />

reporter is inducible following injury to the CNS<br />

and this model provides an easy model feasible for<br />

the study of transcriptional regulation of the GFAP<br />

gene and indications of possible neural injury.<br />

Methods: A brain lesion was induced by placing a<br />

liquid nitrogen cooled cone shaped copper device<br />

with a tip diameter of 1 mm for 1 min. onto the skinfree<br />

surface of the cranium of the cerebral cortex of<br />

a mouse. At different time points after lesion induction<br />

GFAP-luc activity was measured, mice were injected<br />

with Evans blue and brains were stained with<br />

2,3,5-Triphenyltetrazolium chloride (TTC).<br />

Results: 24h after induction of the cryogenic lesion, BLI<br />

measurement showed that, at the lesion site, GFAP-luc<br />

expression was increased 2-3 times over control level<br />

and gradually returned to basal after 72h. Evaluation<br />

imaging life<br />

Gfap-luc expression (Relative Light Units)<br />

Control GF AP–luc<br />

Control<br />

GFAP-luc expression expression<br />

24h<br />

24h after after freeze freeze injury injury<br />

6.0E+06<br />

5.0E+06<br />

4.0E+06<br />

3.0E+06<br />

2.0E+06<br />

1.0E+06<br />

0.0E+00<br />

of tissue viability using TTC staining showed that 24h<br />

after lesion, the primary lesioned cortex appeared as<br />

an unstained white area (dead tissue), whereas, the<br />

rest of the brain appeared red (living tissue). Moreover,<br />

at the lesion site, i.v. injected Evans blue leaked<br />

into the brain tissue, indicative for disruption of the<br />

blood brain barrier. This was also visualised in intact<br />

isolated brains by Fluorescence Imaging (FLI).<br />

Conclusions: Cryogenic lesion in the GFAP-luc mouse<br />

model showed a transient induction of GFAP expression,<br />

indicative for astrocyte activation in reaction<br />

Base 1 Base 2 24h 48h 72h<br />

to neural injury. Brain injury was confirmed by TTC<br />

staining and leakage of Evans blue over the blood<br />

brain barrier. GFAP-luc mice provide an easy and convenient<br />

model to study cryogenic brain trauma.<br />

Acknowledgement: This project was supported by<br />

EU-FP7 ENCITE (HEALTH-F5-2008-201842) and<br />

Volkswagen Stiftung.


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

Plaque burden in the APPPS1 mouse picked up with diffusion kurtosis magnetic resonance<br />

imaging<br />

Vanhoutte G. , Geys R. , Pereson S. , Veraart J. , Van Broeckhoven C. , Kumar-Singh S. , Sijbers J. , Van Der Linden A. .<br />

University of Antwerp, Belgium<br />

greetje.vanhoutte@ua.ac.be<br />

Introduction:Transgenic mouse models are essential<br />

in understanding the pathogenic role of the<br />

β-amyloidogenic pathway in Alzheimer(AD).In vivo<br />

detection of the amyloid deposits in the brain would<br />

be beneficial in terms of diagnosis and therapy follow-up.Therefore<br />

we investigated the sensitivity of<br />

a new method, diffusion kurtosis imaging (DKI), to<br />

detect amyloid burden, a correlate for brain damage<br />

in AD patients and transgenic APP (swe) -PS1 (L166p)<br />

mice. The use of this model was carefully chosen<br />

for its effective amyloidosis in the brain without the<br />

occurrence of neurodegeneration, tau-pathology or<br />

behavioural changes.All mice of 17months manifested<br />

amyloid burden in all brain regions(1).Since<br />

previous studies could show different ex vivo patterns<br />

for mean kurtosis in an APPPS1 mouse model,<br />

we hypothesize that the microstructural changes in<br />

the brain, due to extracellular amyloid deposits,can<br />

be measured in vivo by DKI(2). We investigated the<br />

neocortical and hippocampal regions linked to the<br />

cognitive impairment in AD. Further studies will be<br />

conducted to determine the longitudinal effects.<br />

Methods:Experiments were conducted on a 9,4T<br />

MR system(Bruker Biospec, Ettlingen Germany).<br />

DKI scans were performed on APP (swe) -PS1 (L166p)<br />

mice(n=5) and control WT mice(n=5). The mice<br />

were anaesthetized with isoflurane and monitored to<br />

maintain physiological parameters. The DKI protocol<br />

included 7 b 0 -images and 30 gradient directions<br />

with 7 b-values (400 to 2800s/mm²) acquired with<br />

multi-slice 2-shot SE-EPI(TR/TE=7500/24ms,δ=5ms,<br />

Δ=12ms,acquisition=96*64, FOV=19,2*12.8mm²,<br />

slice thickness=0,40mm, NEX=4). Realignment,<br />

was carried out by the vision lab (Univ. Of Antwerp)<br />

and diffusion kurtosis tensor and diffusion tensor<br />

derived parametric maps were computed (Matlab).<br />

These include mean kurtosis (MK), radial kurtosis<br />

(RK), axial kurtosis (AK), fractional kurtosis anisotropy<br />

(KA), fractional anisotropy (FA), mean diffusion<br />

(MD), axial and radial diffusion (AD, RD)<br />

maps. Regions of interest (cortex and hippocampus)<br />

were chosen based on the AD pathology and delineated<br />

on grey values of FA, MD and magnitude maps<br />

in AMIRA (Mercury Computer systems, San Diego,<br />

USA). Differences of diffusion parameters between<br />

WT and APPPS1 mice were computed by means of<br />

the Mann-Withney non-parametric statistical test in<br />

SPSS 16.0 (SPSS Inc. Chicago, USA).<br />

Results: This is the first in vivo study on AD mouse<br />

models using DKI.Already with a small set of<br />

number of animals, we could detect differences according<br />

to genotype. MK(p


160<br />

WarSaW, poland May 26 – 29, 2010<br />

P-047 In vivo Imaging of Rat Glioma using the TSPO-ligand [ 18 F]DPA-714<br />

Winkeler A. (1) , Boisgard R. (1) , Dubois A. (1) , Awde A. (1) , Zheng J. (1) , Ciobanu L. (2) , Siquier-Pernet K. (1) , Jego B. (1) , Dollé F. (1) ,<br />

Tavitian B. (1) .<br />

(1) CEA\DSV\I2BM\SHFJ, Orsay, France<br />

(2) roSpinCEA\DSV\I2BM\NeuroSpin,France<br />

alexandra.winkeler@cea.fr<br />

Introduction: In the last years there has been an<br />

enormous increase in the development of radioligands<br />

targeted against the translocator protein<br />

TSPO (18 kDa). TSPO expression is nearly absent<br />

in the intact CNS parenchyma but increases rapidly<br />

upon inflammation in activated microglia and<br />

serves as a biomarker for imaging cerebral inflammation<br />

(1). In addition, TSPO has also been reported<br />

to be over-expressed in a number of cancer cell<br />

lines (2, 3) and human tumours including glioma<br />

(4). Here, we investigated the use of the PET-radioligand<br />

[ 18 F]DPA-714 (5) as new marker to image<br />

glioma in vivo.<br />

Methods: 9L rat glioma cells have been stereotactically<br />

implanted in the striatum of Fisher, Wistar and<br />

Sprague Dawley rats. Dynamic [ 18 F]DPA-714 PET<br />

imaging was performed 11-14 days after implantation.<br />

The injected dosed was 1.24 + 0.30 mCi (mean<br />

+ std). T2w-MRI and/or [ 11 C]Methionine PET were<br />

acquired prior to the [ 18 F]DPA-714 PET imaging<br />

session in order to monitor tumor growth. The [ 18 F]<br />

DPA-714 PET images were then co-registered to<br />

the corresponding MRI. For quantitative analysis<br />

a volume-of-interest (VOI) analysis was performed<br />

on both the kinetic and summed image data sets. In<br />

addition, the expression of TSPO 9L rat glioma cells<br />

was investigated using Western Blot.<br />

Results: 9L glioma tumors grown in Fisher (n=5),<br />

Wistar (n=4) and Sprague Dawley (n=6) rats were imaged<br />

by [ 18 F]DPA-714 PET. Tumors grown in Fisher<br />

and Wistar rats were also monitored by MRI. All rats<br />

showed significant [ 18 F]DPA-714 PET accumulation<br />

at the site of tumor implantation compared to the<br />

contralateral site. Tthe %ID/cc in Fisher, Wistar and<br />

Sprague Dawley rats is listed in the following table:<br />

imaging life<br />

control (mean+std) tumor (mean+std)<br />

Fisher 0.15 + 0.02% 0.49 + 0.05%<br />

Wistar 0.13 + 0.06% 0.35 + 0.09%<br />

Sprague Dawley 0.11 + 0.05% 0.26 + 0.06%<br />

TSPO expression was confirmed by Western Blot in<br />

9L cells in vitro and by immunohistochemistry ex vivo.<br />

Conclusions: This study demonstrated the feasibility<br />

of using the TSPO-radioligand [ 18 F]DPA-714 to<br />

characterize 9L glioma in vivo in different rat models<br />

with PET imaging. [ 18 F]DPA-714 therefore has<br />

the potential to become a promising radiotracer to<br />

image human glioma.<br />

Acknowledgement: This work has been supported<br />

by the 6th FW EU grant EMIL (LSHC-CT-2004-<br />

503569) and DiMI (LSHB-CT-2005-512146).<br />

References:<br />

1. Winkeler A, Boisgard R, Martin A, & Tavitian B (2009) J<br />

Nucl Med 51, 1-4.<br />

2. Hardwick M, Fertikh D, Culty M, Li H, Vidic B, &<br />

Papadopoulos V (1999) Cancer Res 59, 831-842.<br />

3. Starosta-Rubinstein S, Ciliax BJ, Penney JB, McKeever<br />

P, & Young AB (1987) Proc Natl Acad Sci U S A 84, 891-<br />

895.<br />

4. Black KL, Ikezaki K, Santori E, Becker DP, & Vinters HV<br />

(1990) Cancer 65, 93-97.<br />

5. Damont A, Hinnen F, Kuhnast B, Schollhorn-<br />

Peyronneau MA, James M, Luus C, Tavitian B, Kassiou<br />

M, & Dolle F (2008) Journal of Labelled Compounds &<br />

Radiopharmaceuticals 51, 286-292.


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

A novel 19F MRI-based migration assay: application to primary human dendritic cells<br />

Bonetto F. (1) , Srinivas M. (1) , Weigelin B. (1) , Cruz Ricondo L.J. (1) , Heerschap A. (2) , Figdor C. (1) , De Vries I.J. (1) .<br />

(1) Nijmegen Center for Molecular Life Science, Radboud University Nijmegen Medical Centre, The Netherlands<br />

(2) Radboud University Nijmegen Medical Centre, Nijmegen, , Netherlands<br />

bonetto.fernando@gmail.com<br />

Introduction: The decisive role of dendritic cells<br />

(DCs) in inducing immunity formed the rationale<br />

for DC immunotherapy: DCs loaded with tumor<br />

antigens are injected into cancer patients to stimulate<br />

T cells to eradicate tumors. However, success<br />

in clinical trials has been limited mainly due to<br />

an inefficient migration rate post-vaccination. As<br />

DC migration can be affected by several factors, a<br />

suitable in vitro assay is required to reproduce in<br />

vivo conditions. Here we present a novel 19 F MRIbased,<br />

quantitative assay to measure cell migration<br />

in varying chemokine environments in a 3D<br />

scaffold specially designed to mimic biological tissue.<br />

We obtained migration rates comparable with<br />

clinical results (using scintigraphy for quantification)<br />

[1] showing the potential application of this<br />

assay to simulate different in vivo migration conditions.<br />

Moreover, we observed that the percentage<br />

of migrating cells strongly depends on the initial<br />

cell density.<br />

Methods: Primary human DCs were cultured, as<br />

per standard protocols for DC vaccination trials<br />

[2]. For 19 F-labeling, a biocompatible polymer<br />

currently in clinical use, poly(D,L-lactide-co-glycolide)<br />

(PLGA) was used to entrap a perfluorcarbon<br />

tracer. After maturation, cells were harvested,<br />

and viability and maturation marker expression<br />

determined. The presence of label within the cells<br />

was checked by optical microscopy. For migration<br />

assays, a variable number of cells were embedded<br />

in a fixed volume of a scaffold matrix (cell layer)<br />

and a chemokine gradient was created above it. A<br />

chemokine-free gel set below the cell layer served<br />

as a control. The sample was placed vertically in<br />

the MRI scanner and only upward migration was<br />

considered to exclude the effects of gravity. Temperature<br />

was maintained at 37 o . All experiments<br />

were performed on a 7T horizontal bore MR system<br />

with a 1 H/ 19 F volume coil. 1 H 2D spin echo images<br />

and chemical shift spectroscopy (CSI) were used<br />

to track and quantify the migration of 1-15x10 6<br />

cells. Nine 19 F-CSI experiments (1.1 hours each)<br />

were sequentially performed in order to keep track<br />

of the migrating cells.<br />

Results: Cell migration was assessed by measuring<br />

cell movement relative to their initial position above<br />

and below the cell-gel layer. Minimal migration was<br />

observed in samples with 10-15×10 6 cells. However,<br />

with smaller cell numbers, 5×10 4 from 5×10 6 cells<br />

(1%) and 2.5×10 4 from 1×10 6 cells (2%) were found<br />

to migrate up. Migration was found to stop 10 hrs<br />

after the start of the experiment. No cell migration<br />

was found below the initial cell layer during the<br />

whole study for all cell layer numbers, indicating<br />

that the upward cell movement was truly caused by<br />

the chemokine gradient.<br />

Conclusions: In the present study we showed that<br />

19 F-CSI and 19 F-MRI can be used to track and quantify<br />

cell migration in 3D collagen scaffold assays.<br />

The number of cells in the initial cell layer was in the<br />

order of the typical cell bolus injection in patients<br />

[1] and the migration rates were measured to be in<br />

the order of 2%, similar to that obtained in clinical<br />

results. The study showed that the initial cell density<br />

plays a decisive role in DCs migration, which<br />

is important information to optimize cell migration<br />

in a clinical setting. Thus, this assay is suitably to<br />

simulate in vivo clinical conditions, giving the potential<br />

chance for detailed analysis of DC migration<br />

by MRI.<br />

Acknowledgement: This work was partially supported<br />

by NWO (VISTA grant) and ZONMW (911-06-<br />

021) for investments.<br />

References:<br />

1. P. Verdijk, T. W. Scheenen, et al.. Int J Cancer 120(5):<br />

978-84 (2007).<br />

2. I. J. de Vries, W. J. Lesterhuis et. al.. Nat. Biotechnol.<br />

23(11), 1407-1412 (2005).<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

P-048<br />

poStEr<br />

GENE and CELL based THERAPIES


P-049<br />

162<br />

WarSaW, poland May 26 – 29, 2010<br />

In vivo magnetic resonance imaging reveals altered migration of endogenous neural<br />

progenitor cells following cuprizone-induced central nervous system demyelination<br />

Guglielmetti C. , Praet J. , Vreys R. , Ponsaerts P. , Van Der Linden A. .<br />

University of Antwerp, Belgium<br />

carolineguglielmetti@hotmail.com<br />

Introduction: The Cuprizone mouse model represents<br />

a highly reproducible tool to study in vivo demyelination<br />

and remyelination processes. The time<br />

course of demyelination-remyelination during Cuprizone<br />

administration has been well characterized<br />

and it has been suggested histologically that endogenous<br />

subventricular zone neural stem/progenitors<br />

cells (NSPC) are recruited during the remyelination<br />

process [1] . We tested whether the recently validated<br />

in situ NSPC labelling technique with micron-sized<br />

iron oxide particles in combination with the transfection<br />

agent Poly-L-lysine [2] (MPIOs-PLL) is suitable<br />

to visualize NSPC migration towards demyelinated<br />

areas.<br />

Methods: 20 C57BL/6 mice (8 weeks old) were used.<br />

All mice were injected in the right lateral ventricle<br />

with 2 µl MPIOs-PLL (9.1 10 5 particles, 0.67 mg Fe/<br />

ml). 10 mice were fed a diet containing Cuprizone<br />

(0.2%) during a 4-week period post-injection (p.i).<br />

In vivo T 2 *-weighted 3D-MGE (78 µm isotropic resolution)<br />

was performed at 4 weeks (peak of inflammation),<br />

6 weeks (peak of proliferation of NSPC)<br />

and 12 weeks (complete remyelination) pi. All images<br />

were acquired on a 9.4T Bruker console. 5 animals<br />

of each group received an injection of 5-bromo-2’-deoxyuridine<br />

(BrdU; 50 mg/kg i.p.) twice a<br />

day over a period of 5 days before either the first or<br />

the second imaging time point.<br />

Results: MRI revealed hypointense voxels along the<br />

rostral migratory stream (RMS) and in the olfactory<br />

bulb (OB) for controls. In contrast, for the Cuprizone<br />

treated mice, hypointense areas towards the<br />

OB were detected only in 2 mice at week 4. From<br />

week 6 onward they were detected in both groups.<br />

Hypointense voxels are also found in the region of<br />

the external capsule (EC) at the ipsilateral side of<br />

injection from 4 weeks onward in the Cuprizone<br />

treated group. Each MR image is a compilation of<br />

5 consecutive MR slices. Histological analyses are<br />

being performed in order to confirm whether the<br />

hypointense voxels are MPIO particles in oligodendrocyte-differentiated<br />

NSPC.<br />

imaging life<br />

Conclusions: In situ labelling of endogenous NSPC<br />

by direct injection of MPIOs-PLL in the lateral ventricle<br />

revealed NSPC migration impairment towards<br />

the OB, as well as the presence of MPIOs in the EC<br />

in the Cuprizone mouse model. Histological analysis<br />

is ongoing in order to further characterise migrated<br />

MPIO-labelled cells.<br />

Acknowledgement: This work is supported by IWT<br />

(SBO/030238), EC-FP6-project DiMI (LSHB-CT-<br />

2005-512146) and IUAP-NIMI-P6/38<br />

References:<br />

1. Matsushima G K et al; Brain Pathology, 11: 107-116<br />

(2001)<br />

2. Vreys R et al; NeuroImage 49, 2094-2103 (2010)


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

Evolution pathways of compartmentalization and distribution of labeling iron-oxide<br />

particles in tumor tissue<br />

Kotek G. , Van Tiel S. , Wielopolski P. , Krestin G. , Bernsen M. .<br />

Erasmus University Medical Center Department of Radiology, Rotterdam, Netherlands<br />

g.kotek@erasmus.nl<br />

Introduction: In our study we addressed the effects<br />

and the evolution of SPIO distribution on R2 and<br />

R2* values according to specific pathways, such as<br />

cell death, cell mixing and cell division. Also we<br />

investigated the effects of technical variations in<br />

labeling efficacy, cell density, imaging resolution<br />

and aggregation of labeling particles. We identify<br />

areas for improvement in cell labeling technique<br />

and imaging for quantitative cell tracking.<br />

Methods: Brown Norway 175 (BN175) sarcoma<br />

cells were labeled with iron-oxide (SPIO) particles.<br />

Various intravoxel SPIO distributions were<br />

prepared by methods mimicking biologically relevant<br />

processes (compartmentalization, mixing,<br />

division). R2* and R2 relaxometry was performed<br />

at high resolution at 3.0T, iron concentration<br />

was measured by optical emission spectrometry.<br />

Effects of spatial distribution and compartmentalization<br />

of SPIO on relaxivity (dR/dcFe) was<br />

analyzed.<br />

Results: We showed that relaxivity is sensitive to<br />

variations of cell labeling, cell density and imaging<br />

resolution. Our data suggest that intracellular<br />

variance of cell division rate potentially lead to<br />

breakdown of unique relaxation rate vs. iron concentration<br />

relationship. Our results indicate that<br />

cell death can be identified by parallel monitoring<br />

of R2 and R2* values, and we confirm that R2’ differentiates<br />

between intracellular and extra-cellular<br />

SPIO. We found a unique relaxation rate vs. iron<br />

concentration relationship in case of dominance<br />

of cell division. Our results suggest that tighter<br />

control on labeling variance put in place for stem<br />

cell labeling.<br />

Conclusions: We challenge labeled cell quantification<br />

by identifying sensitivity of the relaxivity<br />

to imperfections of labeling technique and imaging<br />

method; besides the numerous complications<br />

in quantification, with thorough consideration<br />

of limitations and monitoring of biologically relevant<br />

factors, quantification is feasible.<br />

In case of mixing labeled and non-labeled cells<br />

(likely process in stem cell tracking) a strong<br />

control on labeling efficiency is required, since<br />

R2*(cFe) and R2(cFe) curves are sensitive to initial<br />

intracellular iron content and cell density.<br />

References:<br />

1. Bowen CV, Zhang X, Saab G, et al.<br />

2. Application of the static dephasing regime theory to<br />

superparamagnetic iron-oxide loaded cells. Magn<br />

Reson Med 2002;48:52–61.<br />

3. Rad AM, Arbab AS, Iskander AS, et al. Quantification<br />

of superparamagnetic iron oxide (SPIO)-labeled cells<br />

using MRI. J Magn Reson Imaging 2007;26:366–74.<br />

4. Kuhlpeter R, Dahnke H, Matuszewski L, et al.R2 and R2*<br />

mapping for sensing cell-bound superparamagnetic<br />

nanoparticles: in vitro and murine in vivo testing.<br />

Radiology. 2007 Nov;245(2):449-57. Epub 2007 Sep 11.<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

P-050<br />

poStEr<br />

GENE and CELL based THERAPIES


WarSaW, poland May 26 – 29, 2010<br />

P-051 Acupuncture Works on Endorphins via Activating Stretch-Activated Cation Channels<br />

164<br />

Liang J. (1) , Zhong P. (2) , Yang X. (3) , Li G. (3) , Yang E. (3) , Cheung P. (1) .<br />

(1) The University of Hong Kong, Hong Kong, Hong Kong<br />

(2) First Affiliated Hospital of Sun Yat-Sen University,<br />

(3) Hong Kong Applied Science and Technology Research Institute Company Limited, Hong Kong<br />

cmleung@eee.hku.hk<br />

Introduction: Acupuncture has been in use for three<br />

thousand years in China but was not practiced in the<br />

West until the last three decades. Among acupuncture<br />

therapies, the acupuncture-induced analgesic effect<br />

has been used widely to alleviate diverse pains, particularly<br />

chronic pain. To shed light on the 2500-yearold<br />

acupuncture analgesia, pain control by acupuncture<br />

through Ca2+-regulated release of opioid peptides<br />

is proposed.<br />

Methods: 4-week-old C57BL/6N mice were kept in<br />

a specially designed holder, with their hind legs and<br />

tails exposed during acupuncture. An acupuncture<br />

needle (Hwato) 0.4 mm in diameter, 50 mm in length<br />

was inserted into the right hind leg, between the<br />

tibia and fibula, approximately 5 mm lateral to the<br />

anterior tubercle of the tibia. The acupuncture needle<br />

was driven by a piezoelectric bending element at 1<br />

Hz. After receiving acupuncture treatment for 40<br />

minutes, animals were sacrificed. Blood samples were<br />

harvested from the left ventricle of the heart. Plasma<br />

was separated by centrifuging samples at 1000 × g<br />

for 15 minutes at 4 ˚C and diluted 1:10 in a sample diluent<br />

before assay. The levels of plasmaβ-endorphin<br />

were measured using a Mouse β-Endorphin ELISA kit<br />

from USCN Life Science. Method for in vivo monitoring<br />

of Ca2+ excitation in mouse skeletal muscle during<br />

acupuncture was described in [1].<br />

Results: Acupuncture was initiated by the stimulation<br />

of needle driver describe above in the hind-limb muscle<br />

of the mouse. Plasma β-endorphin was measured<br />

40 min after the acupuncture treatment by immunoassay.<br />

Elevation of β-endorphin levels is observed<br />

(Fig.1b) along with cytosolic Ca2+ activation in muscle<br />

fibers in vivo (Fig.1a) 40 min after the stimulation<br />

imaging life<br />

is applied. The acupuncture induced β-endorphin<br />

secretion is blocked by the intraperitoneal injection<br />

of Gd3+, thestretch-activated Ca2+ channels blocker.<br />

Conclusions: These findings suggest that acupuncture<br />

analgesia depends on the physiological afferent signal<br />

elicited in the mechanosensation pathway. The acupuncture<br />

needle manipulation induces a Ca2+-dependent<br />

secretion of β-endorphin via interactions with<br />

stretch-activated Ca2+ channels. The depolarizationevoked<br />

opioid peptide β-endorphin then penetrates<br />

the fenestrated capil ary vessels in the pituitary by diffusion,<br />

and so enters the general circulation. Our results<br />

are in agreement with the studies on endogenous<br />

neuropeptide release during electroacupuncture [2]<br />

and may lead to new directions for acupuncture analgesia<br />

research by unveiling its cellular signal transduction<br />

pathways.<br />

References:<br />

Fig1: Acupuncture stimulates<br />

ß -endorphin release via<br />

activating stretch-activated<br />

cation channels.<br />

1. Liang J.M., Li G., Yang E.S., Cheung P.Y.S. In Vivo Monitoring<br />

of Ca2+ Excitation in Mouse Skeletal Muscle during<br />

Acupuncture. WMIC 2009, p.527. [2] Han J.S. Acupuncture:<br />

neuropeptide release produced by electrical stimulation<br />

of different frequencies. Trends Neurosci. 26, 17-22 (2003).


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

Labeling protocols for MRI and optical imaging of human muscle cells precursors<br />

Libani I.V. (1) , Lui R. (1) , Martelli C. (1) , Clerici M. (1) , Fiorini C. (2) , Lucignani G. (1) , Ottobrini L. (1) .<br />

(1) University of Milan, Rescaldina, Italy<br />

(2) Politecnico of Milan, Italy<br />

ilaria.libani@unimi.it<br />

Introduction: The interest about cell-mediated<br />

therapy finalized to tissues regeneration research<br />

is increased in the last few years. Although numerous<br />

protocols which include extraction of stem cells<br />

from healthy animals and implantation in diseased<br />

models were set up, important parameters such as<br />

the distribution and localization of the injected cells,<br />

cell survival, proliferation and differentiation cannot<br />

be evaluated in vivo. Here we refined and tested specific<br />

labelling protocols for in vivo visualization by<br />

MRI, SPECT and Optical Imaging of a human muscle<br />

cells precursor cell line as a proof of principle for<br />

the application of these procedure to stem cells in the<br />

evauation of muscle stem cell mediated treatments.<br />

Methods: Human Skeletal Muscle Cells (HSkMC)<br />

were labelled for 24 or 48h with different amounts<br />

of Endorem® (0-100–200 μg Fe/mL) in presence or<br />

not of Poly-L-Lysine (PLL), Protamine Sulfate (PrS),<br />

Polybrene (PB) or infected, with a lentiviral vector<br />

[1] carrying Luciferase gene under control of the<br />

muscle specific Myogenin promoter (pGZ.Myo.L<br />

vector). Labelled HSkMC were analized for viability,<br />

iron content (Perl’s Staining and spectrophotometer<br />

analysis [2]), morphology, differentiation capability<br />

or intrarterially (i.a.) injected into NUDE mice for<br />

in-vivo imaging by means of MRI, FLI or BLI. Initial<br />

cell distribution was also followed with scintigraphy<br />

after cell labelling with 111Indium-oxime (60<br />

μCi/106 cells) and gamma counting of explanted organ<br />

was performed to validate imaging data ex-vivo.<br />

Results: HSkMC incubated for 24 or 48h with 0-100-<br />

200 μg Fe/mL did not show significant differences,<br />

in terms of viability, between labelled/non-labelled<br />

cells in the presence or absence of PLL, PB or PrS<br />

(n=3) remaining higher than 84+8% after 24h and<br />

higher than 80,6+4,6% after 48h. The percentage of<br />

Iron+ cells increased in proportion to the iron content<br />

in the medium even if there are not significat<br />

differences between CTRL and PLL. In particular we<br />

obtained more than 90% Iron+ cells in the samples<br />

incubated with 200 μg Fe/mL for 24 or 48h. Intracellular<br />

iron content reached the higest levels in the<br />

samples loaded with 200ug/ml+PLL: 61,5+2,8 pg/<br />

cells after 24h and 114,9+9,6 pg/cells after 48h. For<br />

this reason 200 ug/mL endorem+PLL was deemed as<br />

the ideal condition for cell labeling for in vivo visualisation<br />

by MRI. Loaded HSkMC were injected i.a. into<br />

NUDE murine model of muscle inflammation. MRI<br />

permitted to follow over time HSkMC distribution<br />

into the injured muscle. SPECT and BLI of HSkMC<br />

not only confirmed celldistribution to muscle but also<br />

revealed an early localisation into the lung. HSkMC infected<br />

with pGZ.Myo.L, i.a. injected in the same mouse<br />

model, were detectable in muscle up to 5 weeks after<br />

injection. Interestingly, at this timepoint, the signal<br />

is still present only near the lesion area while disappeared<br />

in the rest of the body suggesting that this construct<br />

could be used to evaluate not only localisation<br />

but also the differentiation in vivo by means of BLI.<br />

Conclusions: We set up protocols to efficiently<br />

visualize human muscle cells precursors localization<br />

and differentiation by MRI, SPECT or in vivo<br />

optical imaging. These protocols will be useful to<br />

study the fate of cells once injected into recipient<br />

NUDE mice with different techniques and will<br />

make it possible to study their behaviour in vivo<br />

over time. Furthermore, it will be possible to use<br />

these instruments for the in vivo study of muscular<br />

stem cells in restoring skeletal muscles after<br />

damage.<br />

Acknowledgement: This work was supported by a<br />

Cariplo Foundation grant, the FP6 Hi-CAM project<br />

(LSHC-CT-2006-037737) and from the Doctorate<br />

School of Molecular Medicine, University of Milan.<br />

References:<br />

1. Naldini L. et al; Science 272(5259):263-7 (1996)<br />

2. Boutry N. et al; Contrast Media Mol Imaging 3(6):223-<br />

32 (2008)<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

P-052<br />

poStEr<br />

GENE and CELL based THERAPIES


WarSaW, poland May 26 – 29, 2010<br />

P-053 Studying molecular processes in-vivo: A framework for quantifying variability in molecular<br />

MRI<br />

166<br />

Plenge E. (1) , Kotek G. (1) , Guenoun J. (1) , Doeswijk G. (1) , Krestin G. (1) , Niessen W. (2) , Meijering E. (1) , Bernsen M. (1) .<br />

(1) Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands<br />

(2) Erasmus University Medical Center, Rotterdam and Faculty of Applied Sciences, Delft University of Technology, The Netherlands<br />

e.plenge@erasmusmc.nl<br />

Introduction: Quantitative characterization of cellular<br />

and molecular processes in-vivo is dependent<br />

on consistent image data. To justify conclusions regarding<br />

observed biological changes over time, the<br />

changes should exceed those attributed to imaging<br />

artifacts and inconsistencies. The aim of this work<br />

is to establish a framework for quantifying the variability<br />

inherent to molecular magnetic resonance<br />

imaging (mMRI) protocols.<br />

Methods: Our framework consists of variance maps,<br />

variance histograms, visualization and quantification<br />

of the precision of expert contouring. To demonstrate<br />

the efficacy of our framework we have compared the<br />

variability of two different acquisition protocols in<br />

the challenging environment of a rodent heart. SPIO<br />

labeled stem cells were injected into the myocardium<br />

of a rat and cardiac MRI performed. Without moving<br />

the anesthetized rat at any point, seven 3D nontriggered<br />

images and seven 2D cine sequences were<br />

acquired intermittently to capture inevitable changes<br />

due to e.g. breathing, coil-heating etc. in both datasets.<br />

The assumption was made that no biological changes<br />

occur within the acquisition time. After rigid registration<br />

of the images in each dataset, the per-voxel variance<br />

over each dataset was calculated. Variance maps<br />

were generated for visualization, variance distributions<br />

for quantification. Three experts manually segmented<br />

the cell cluster in each slice/frame of each 3D/cine sequence.<br />

The inter- and intra-observer variability was<br />

visualized and quantified by a similarity index (SI) [1].<br />

imaging life<br />

Results: The created framework allows objective<br />

quantification of the variability inherent to mMRI<br />

protocols. Fig. 1 and table 1 visualize and quantify<br />

the variance distribution of the datasets due to imaging<br />

inconsistencies, and the inter- and intra-observer<br />

variability of the expert segmentation. Through<br />

such a quantitative assessment a lower limit on the<br />

longitudinal changes in the biological process to be<br />

studied can be defined.<br />

Conclusions: We have proposed a framework for<br />

evaluating mMRI protocols in terms of variability.<br />

The proposed framework allows anyone to objectively<br />

assess how well-suited an mMRI protocol is<br />

for a longitudinal study of a given biological process.<br />

Acknowledgement: Supported by ENCITE, funded<br />

by the European Community under FP7.<br />

Inter-obs.<br />

variability, SI<br />

Intra-obs.<br />

variability, SI<br />

3D 2D cine<br />

0.70 0.79<br />

0.56 0.64<br />

Fig1: Top and bottom row shows visualizations related to the 3D<br />

and 2D cine datasets, respectively. Left: Slice/frame of each dataset.<br />

Middle: Histograms of per-voxel variance of each dataset. Right: Variability<br />

among three expert’s segmentations of cell cluster (colors represent<br />

the number of experts including the voxel in the cell cluster).<br />

References:<br />

1. Pohl KM et al; Med.Img.Analysis, 11:5:465-477 (2007)


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

Clinically applicable cell tracking by MRI in cartilage repair using Superparamagnetic Iron<br />

Oxide (SPIO)<br />

Van Buul G. , Kotek G. , Wielopolski P. , Farrell E. , Uijtterdijk A. , Bos P. , Weinans H. , Verhaar J. , Krestin G. , Van Osch G. ,<br />

Bernsen M. .<br />

Erasmus MC, Rotterdam, The Netherlands<br />

g.vanbuul@erasmusmc.nl<br />

Introduction: Cell tracking is a useful tool for<br />

optimizing cell-based cartilage repair. Cell labeling<br />

using superparamagnetic iron oxides (SPIOs)<br />

enables non-invasive in vivo cell tracking by MRI,<br />

and has already been used experimentally in a<br />

clinical setting[1]. We investigated the clinical,<br />

intra-articular applicability of this cell tracking<br />

technique regarding safety, MRI traceability and<br />

label re-uptake.<br />

Methods: Part 1: Human bone marrow stromal<br />

cells (hBMSCs) were labeled with SPIO (ferumoxides-protamine<br />

sulphate complexes) in a range<br />

of 0 - 250 μg/ml. Cell viability was asses-sed and<br />

metabolic cell activity was quantified up to seven<br />

days. Part 2: SPIO-labeled hBMSCs (100,000 to<br />

5,000,000 cells) were injected ex vivo in pig knees,<br />

to mimic a clinically relevant sized model. Furthermore,<br />

SPIO-labeled cells (10,000 - 1,000,000<br />

per 75 μl) were seeded in cartilage defects in vitro.<br />

Scanning was performed on a clinical 3.0 T MRI<br />

scanner. Part 3: To show possible SPIO re-uptake<br />

by synovial cells, viable and dead GFP-SPIO double-labeled<br />

chondrocytes were seeded on human<br />

synovium explants for five days. Samples were<br />

analyzed using fluorescence- and light microscopy.<br />

Results: Part 1 Cell labeling<br />

and -behaviour:<br />

SPIO labeling resulted<br />

in labeling efficiencies<br />

of approximately 90%<br />

and did not negatively<br />

affect cell viability or<br />

cell proliferation for<br />

dosages up to 250 μg/ml.<br />

Part 3 SPIO re-uptake: GFP+-SPIO+ cells, indicating<br />

originally seeded cells, were seen in synovium<br />

samples containing living cells. GFP – -SPIO+ cells,<br />

indicating SPIO re-uptake by synovial cells, were<br />

found in samples containing dead cells.<br />

Conclusions: Although possible SPIO re-uptake<br />

by host cells might limit duration of accurate cell<br />

tracking, we showed promising results for the use<br />

of SPIO labeling for cell tracking in clinical cartilage<br />

repair procedures. This approach provides the<br />

extra advantage to simultaneously track cells and<br />

evaluate cartilage repair in one MRI session.<br />

Acknowledgement: This work is supp. by the Smart-<br />

Mix Prog. of the Neth. Min. of Econ. Aff. & the<br />

Neth. Min. of Educ., Cult. & Science; and the EC-<br />

FP7 project ENCITE (HEALTH-F5-2008-201842).<br />

Part 2 MRI traceability: Fig1: Intra-articular injected SPIO-labeled cells (1A) and cells seeded in circular cartilage defects in a volume of 75<br />

All SPIO-labeled cell dosages,<br />

both intra-articular<br />

µl (1B) were accurately visualized by MRI. The amount of intensity of signal voids was related to used cell number.<br />

injected or seeded in cartilage defects, were visualized<br />

by MRI (Fig. 1). The amount of signal voids<br />

was related to the used cell number. SPIO-labeled<br />

cells seeded in cartilage defects in vitro could be visualized<br />

and quantified using a T2* mapping MRI<br />

References:<br />

technique.<br />

1. Bulte JW; AJR Am J Roentgenol. 193(2):314-25 (2009)<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

P-054<br />

poStEr<br />

GENE and CELL based THERAPIES


168<br />

WarSaW, poland May 26 – 29, 2010<br />

P-055 Labeling of HUVEC with different iron oxide particles: An in vitro study about incorporation,<br />

distribution, retention and toxicity<br />

Van Tiel S. , Wielopolski P. , Houston G. , Krestin G. , Bernsen M. .<br />

ErasmusMC, Rotterdam, The Netherlands<br />

s.vantiel@erasmusmc.nl<br />

Introduction: For in vivo cell tracking it is essential<br />

that the cells have incorporated a label so that<br />

they can be distinguished from their surroundings<br />

in Magnetic Resonance Imaging (MRI)[1-3]. A vast<br />

amount of studies have been published dealing with<br />

labeling of various cell types with iron oxide nanoparticles.<br />

In these studies a large variety of labeling<br />

protocols have been described. While for every cell<br />

type tested efficient labeling and subsequent detection<br />

by MRI[4] has been reported, it is not clear<br />

how different labeling protocols may influence labeling<br />

efficiency. The purpose of this study was to<br />

systematically investigate the effect of variations in<br />

dose and duration of labeling on label incorporation,<br />

distribution, retention and toxicity using two commonly<br />

used types of iron oxide nanoparticles; the<br />

so-called SPIO and MPIO particles.<br />

Methods: Primary culture Human Umbilical Vein<br />

Endothelial Cells (HUVECs) were grown to 80-<br />

90% confluence and labeled with SPIO or MPIO<br />

at concentrations ranging from 0 - 100 µg Fe and<br />

incubation times of 4-48hrs. Cellular iron load was<br />

measured by Inductible Coupled Plasma- Optical<br />

Emission Spectrometry (ICP-OES). Cellular localization<br />

and retention over time of iron oxide complexes<br />

was assessed on cytospin slides. Cell functionality of<br />

labeled cells was tested by a tube forming assay. MRI<br />

traceability of labelled cells was tested using a 3D-<br />

SPGR sequence with TR/TE 41.1/10.5 ms, and a flip<br />

angle (α) of 50º with a resolution of 38 μm x 38 μm x<br />

100 μm and a FOV of 2 cm x 2 cm.<br />

Results: Under the conditions tested a maximal<br />

iron load of 18.2 pg per cell was obtained for SPIO.<br />

A much higher iron load of 661 pg per cell was<br />

achieved with MPIO. Inter and intra cellular distribution<br />

of label are both strongly dependent on<br />

the labeling protocol used. Labeling with high doses<br />

and short incubation times may result in large<br />

intra-cellular vesicles with multiple iron-oxide<br />

complexes. Labeling with low doses and long incubation<br />

times may result in small intra-cellular<br />

vesicles with just one iron-oxide complex. A better<br />

retention of label was observed after short incubation<br />

times and high labeling doses. For both MPIO<br />

imaging life<br />

and SPIO higher doses were tolerated at shorter incubation<br />

times. At equal incubation times, MPIO<br />

was better tolerated than SPIO. In FACS studies,<br />

clear changes in forward scatter and side scatter<br />

plots, corresponding to changes in cell size and cell<br />

granularity respectively were observed following<br />

labeling. Both effects were more pronounced after<br />

labeling with MPIO than with SPIO. At the highest<br />

doses of MPIO tested cell sizes increased 4-7<br />

times in cell volume compared to unlabeled control<br />

cells. The effects of SPIO and MPIO labeling<br />

on tube forming capacity of HUVECS was tested at<br />

all doses that did not significantly affect cell survival.<br />

For all these conditions tested HUVECs still<br />

displayed tube forming capacity. Sensitive imaging<br />

by MRI at the single cell level in vitro was possible<br />

for all conditions tested.<br />

Conclusions: HUVECs can be labeled efficiently with<br />

SPIO and MPIO, but dose and duration of exposure<br />

of cells to the particles strongly influence label incorporation,<br />

distribution, retention and toxicity.<br />

Acknowledgement: This research has been done in<br />

part through support from ENCITE - funded by the<br />

European Community under the 7th Framework<br />

program.<br />

References:<br />

1. Bulte JW. Methods Mol Med.124:419-439 (2006)<br />

2. Modo M et al. Mol Imaging. 4:143-164 (2005)<br />

3. Hoehn M et al. J Physiol. 584:25-30. (2007)<br />

4. van Buul GM et al. Contrast Media Mol Imaging. 4:230-<br />

236 (2009)


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

Visualization of aberrant migration in the YAC 128 mouse model for Huntington’s disease by<br />

in situ labelling of neural progenitor cells with iron oxide particles<br />

Vreys R. (1) , Blockx I. (1) , Kandasamy M. (2) , Nguyen H.H.P. (3) , Verhoye M. (1) , Aigner L. (2) , Van Der Linden A. (1) .<br />

(1) University of Antwerp, Belgium<br />

(2) Paracelsus Medical University Salzburg, Austria<br />

(3) University of Tübingen, , Germany<br />

ruth.vreys@ua.ac.be<br />

Introduction: The outcome of a recent validation<br />

study of various in situ cell labelling strategies is<br />

that intraventricular injection of a small number of<br />

micron-sized iron oxide particles (MPIOs) in combination<br />

with the transfection agent Poly-L-lysine<br />

(PLL) is a successful method for the MRI visualization<br />

of endogenous neural progenitor cell (NPC)<br />

migration in the adult mouse brain [1] . As it was<br />

recently reported that NPC migration is impaired<br />

in a mouse model of Huntington’s disease (HD) [2] ,<br />

we tested if the in situ labelling technique using<br />

MPIOs-PLL is suitable to visualize aberrant NPC<br />

migration in YAC128 mice, a mouse model of HD.<br />

Methods: YAC128 transgenic mice (n=5; 66-69<br />

weeks old) maintained on a FVB/N background<br />

and age-matched non-transgenic littermates (n=5;<br />

wild-types) were used. Mice were injected intraventricular<br />

with 1.5 µl MPIOs-PLL (9.1 x 10 5 particles,<br />

0.67 mg Fe/ml). In vivo T 2 *-weighted 3D-GE<br />

(78 µm isotropic resolution; 9.4T) was performed<br />

at 1 week up to 13 weeks post injection. Ex vivo<br />

T 2 *-weighted 3D-GE (66 µm isotropic resolution;<br />

9.4T) was performed on perfused and fixed brains<br />

in their skulls. Four days before perfusion, mice<br />

received 5 times (every 3 h) an injection of 5-bromo-2’-deoxyuridine<br />

(BrdU; 50 mg/kg i.p). Immunohistochemistry<br />

was performed for BrdU on 25<br />

µm sagittal sections.<br />

Results: MRI revealed hypointense pixels along the<br />

rostral migratory stream (RMS) and in the olfactory<br />

bulb (OB) for all wild-types (WT). In contrast, for the<br />

YAC 128 mice (YAC HD), hypointense areas towards<br />

the OB were detected in only two mice. As neurogenesis<br />

is reduced upon ageing, the fraction of MPIOlabelled<br />

NPCs was very limited. The first column in<br />

the figure shows for both groups minimum intensity<br />

projection (mIP) compositions of ex vivo MR images<br />

comprising the RMS of five animals. The mIP of YAC<br />

HD shows less hypointense areas in the RMS and OB<br />

compared to the WT. Histological staining for BrdU<br />

shows that there are less BrdU + cells present in the<br />

RMS and OB of the YAC HD compared to the WT<br />

(figure, second and third column).<br />

Conclusions: In situ labelling of endogenous NPCs<br />

by intraventricular injection of MPIOs-PLL revealed<br />

aberrant NPC migration towards the OB in<br />

the YAC128 mouse model for HD.<br />

Acknowledgement: Supported in part by: IWT (Ph.D.<br />

grant; SBO/030238), EC-FP6-project DiMI (LSHB-<br />

CT-2005-512146) and IUAP-NIMI-P6/38<br />

References:<br />

1. Vreys R et al; NeuroImage 49, 2094-2103 (2010)<br />

2. Moraes L et al; Neuropathology 29, 140-147 (2009)<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

P-056<br />

poStEr<br />

GENE and CELL based THERAPIES


WarSaW, poland May 26 – 29, 2010<br />

P-057 Optimization of in vitro radiolabeling of mesenchymal stem cells with 18 Ffluorodeoxyglucose<br />

170<br />

Wolfs E. (1) , Bormans G. (2) , Van Santvoort A. (3) , Vermaelen P. (3) , Mortelmans L. (3) , Verfaillie C. (4) , Van Laere K. (3) , Deroose C. (3) .<br />

(1) K.U.Leuven, Leuven, Belgium<br />

(2) Laboratory for Radiopharmacy, K.U.Leuven, Belgium<br />

(3) Division of Nuclear Medicine, K.U.Leuven, Belgium<br />

(4) Stem Cell Institute, K.U.Leuven, Belgium<br />

Esther.Wolfs@med.kuleuven.be<br />

Introduction: Because of their great differentiation<br />

potential and immunomodulatory properties, mesenchymal<br />

stem cells (MSCs) are considered to be a<br />

potential source for tissue regeneration and immunomodulatory<br />

therapy [1-4]. For the optimization<br />

of this type of stem cell therapy, novel methods are<br />

required to follow the in vivo fate of stem cells after<br />

injection [5]. Stem cells can be labeled with radioisotopes<br />

in vitro, such as 18 F-fluorodeoxyglucose<br />

( 18 F-FDG), a positron emitting glucose-analogue<br />

which is taken up by cells in a similar manner as<br />

glucose. After phosphorylation, 18 F-FDG is trapped<br />

intracellularly, since it misses the 2’-hydroxyl group<br />

blocking further steps of the glycolysis pathway. The<br />

molecule stays in the cell until its decay [6]. The aim<br />

of this study is to optimize the radioactive labeling<br />

of MSCs in vitro with 18 F-FDG.<br />

Methods: Mouse MSCs were obtained from the<br />

Stem Cell Institute Leuven, and were seeded in 24<br />

well dishes for uptake experiments. 18 F-FDG (5µCi)<br />

was added to each well, in the presence of different<br />

insulin concentrations, and for uptake kinetics for<br />

up to 6 hours. In addition, 18 F-FDG washout after<br />

labeling was assessed. Additionally, 3 mice were<br />

injected with 500 000 18 F-FDG-labeled MSCs, and<br />

stem cell biodistribution was investigated with a<br />

µPET scanner 10 minutes after injection. Furthermore,<br />

a µCT image was acquired for coregistration<br />

with the µPET images.<br />

Results: 18 F-FDG uptake by MSCs was slightly higher<br />

with the addition of insulin (0,01 IU) but with only<br />

limited additional effect of higher concentrations.<br />

Furthermore, experiments also showed significantly<br />

higher 18 F-FDG uptake with longer incubation periods,<br />

slowly reaching a plateau after 6h of incubation.<br />

However, a significant tracer washout could be<br />

observed after 1 hour. In addition, in vivo µPET experiments<br />

with radiolabeled MSCs in mice showed<br />

almost exclusive pulmonary activity, confirming the<br />

intracellular location of the tracer.<br />

Conclusions: This study demonstrates that mouse<br />

MSCs can be successfully labeled with 18 F-FDG<br />

in vitro. Labeling efficiency increases even when<br />

very low insulin concentrations are added. Uptake<br />

imaging life<br />

kinetics assessment showed a significantly higher<br />

uptake with increasing incubation time for up to 6<br />

hours of incubation. Despite the tracer washout after<br />

one hour in vitro, in vivo µPET studies confirmed<br />

the intracellular location of the tracer through an<br />

almost exclusive pulmonary signal. Furthermore,<br />

in vitro labeling with the given dose (5µCi) permits<br />

solid imaging of stem cells injected in mice.<br />

References:<br />

1. Caplan A. Adult mesenchymal stem cells for tissue<br />

engineering versus regenerative medicine. J Cell<br />

Physiol 2007;213(2):341-7.<br />

2. Jackson L, Jones D, Scotting P, Scottile V. Adult<br />

mesenchymal stem cells: differentiation potential<br />

and therapeutic applications. J Postgrad Med<br />

2007;54(2):121-8.<br />

3. Caplan A. Why are MSCs therapeutic? New data: new<br />

insight. J Pathol 2009;217(2):318-24.<br />

4. Jiang Y, et al. Pluripotency of mesenchymal<br />

stem cells derived from adult marrow. Nature<br />

2002;418(6893):41-9.<br />

5. Lauwers E, et al. Non-invasive imaging of<br />

neuropathology in a rat model of a-synuclein<br />

overexpression. Neurobiol Aging 2007;28(2):248-57.<br />

6. Caracó C, et al. Cellular release of [18F]2-fluoro-<br />

2-deoxyglucose as a function of the Glucose-6phosphatase<br />

Enzyme system. J Biol Chem 2000; 275<br />

(24): 18489-2000


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

Responsive MRI contrast agent for specific cell imaging of inhibitory, GABAergic neurons<br />

Aswendt M. (1) , Gianolio E. (2) , Kruttwig K. (1) , Brüggemann C. (1) , Aime S. (2) , Himmelreich U. (3) , Hoehn M. (1) .<br />

(1) Max Planck Institute for Neurological Research, Cologne, Germany<br />

(2) University of Turin, Italy<br />

(3) Katholieke Universiteit Leuven, Belgium<br />

Markus.Aswendt@nf.mpg.de<br />

Introduction: γ-Aminobutyric acid (GABA) is the<br />

major known inhibitory neurotransmitter. The ratelimiting<br />

step in the synthesis of GABA is the decarboxylation<br />

of glutamate by the enzyme glutamic acid<br />

decarboxylase (GAD), which exists in two isoforms:<br />

GAD65 and GAD67 [1].<br />

We exploited the feasibitity of cell specific contrast<br />

generation with the new smart contrast agent Gd-<br />

DOTAgad responsive to GAD activity. It consists of<br />

the chelate Gd-DOTA coupled to a long hydrocarbon<br />

backbone with glutamic acid residues. Upon cleavage<br />

of the glutamate moieties the hydration of the<br />

paramagnetic metal ion increases,<br />

leading to a higher relaxivity.<br />

Methods: Postnatal day 14,<br />

Wistar rat tissues were mechanically<br />

homogenized, and<br />

lysed with cell lysis reagent<br />

M-PER (Pierce, USA). After<br />

protein determination, same<br />

amounts were incubated at<br />

37°C for 24 hours with 1 mM<br />

Gd-DOTAgad together with<br />

the GAD activator pyridoxal-<br />

5’-phosphate. MR measurements<br />

were made at 4.7T (Bruker,<br />

Germany) using a custom<br />

made coil system. The MR phantoms consist of<br />

plastic tubes embedded in 1.5% agarose (Fluka,<br />

Switzerland). RAREVTR scans (TE=12.0ms,<br />

TR=30–250 ms, matrix=128x128; FOV=30.0 mm,<br />

resolution=0.234x0.234 mm; slice thickness=2.0<br />

mm). CGR8 murine embryonic stem cells were differentiated<br />

in a 3 step protocol using chemically<br />

defined media.<br />

Results: Tissue lysates of cerebellum, cortex and<br />

liver (as a negative control) were incubated with<br />

Gd-DOTAgad. Corresponding controls were Gd-<br />

DOTAgad in PBS and tissue lysates without contrast<br />

agent. T1 maps calculated from RAREVTR with an<br />

in-house software were used to evaluate the change<br />

in relaxation rate ΔR1 (Fig. 1A) identifying the highest<br />

changes with brain lysates. CGR8 cells were successfully<br />

differentiated in a 10 day-lasting protocol<br />

resulting in a high proportion of GAD65/67 positive<br />

cells, proven by immunocytochemistry (Fig. 1B).<br />

Conclusions: Gd-DOTAgad incubated with tissue<br />

lysates leads to enhanced ΔR1 rates in highly expressing<br />

GAD65/67 brain regions. A differentiation<br />

protocol to obtain GABAergic neurons from ES cells<br />

was successfully established. Efficient labelling with<br />

Gd-DOTAgad of murine ES and neuronal precursor<br />

cells is in progress for evaluation of differentiation<br />

induced contrast. With this approach it will be possible<br />

to label cells prior to transplantation and to<br />

follow their differentiation fate in vivo with MRI.<br />

Acknowledgement: This work was supported in part<br />

by grants from the European Union under FP6 program<br />

(StemStroke, LSHB-CT-2006-037526) (DiMI,<br />

LSHB-CT-2005-512146), and under FP7 program<br />

(ENCITE, HEALTH-F5-2008-201842).<br />

References:<br />

1. Erlander, M.G., et al., Two genes encode distinct<br />

glutamate decarboxylases. Neuron, 1991. 7(1): p. 91-100.<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

P-058<br />

poStEr<br />

PROBE DESIGN


172<br />

WarSaW, poland May 26 – 29, 2010<br />

P-059 Novel ultrasound contrast agents for drug delivery<br />

Berti R.P. (1) , Freret L. (1) , Haiat G. (2) , Pisani E. (3) , Díaz-Lópeza R. (3) , Tsapis N. (3) , Pucci B. (4) , Fattal E. (4) , Taulier N. (1) , Urbach W. (1) .<br />

(1) Laboratoire d’Imagerie Paramétrique, PARIS, France<br />

(2) Laboratoire de Biomécanique et Biomatériaux Ostéo-Articulaires, France<br />

(3) Laboratoire de Pharmacie Galénique, France<br />

(4) Laboratoire de Chimie Bioorganique et des Systèmes Moléculaires Vectoriels, France<br />

romain.berti@upmc.fr<br />

Introduction: We are working on novel Ultrasound<br />

Contrast Agents (UCA) to improve the signal<br />

for echographic diagnosis and at the same time<br />

to extend their applications as possible drug carriers.<br />

Methods: We have investigated two types of agents:The<br />

first one is made of polymer PLGA (Poly Lactic Glycolic<br />

Acid) that encapsulates a liquid PFOB (PerFluoro-<br />

Octyl Bromide) core. The agent radius can be varied<br />

from 150 nm to 10 µm and it remains stable for weeks [1] .<br />

The second type is made of a telomer FTAC (fluorinated<br />

polymer) encapsulating a fluorinated liquid or gas core.<br />

It forms agent with a radius of about 100 nm and are<br />

stable over days. The size of both types of agent is weakly<br />

polydisperse. In addition, for a better understanding<br />

of the relationship between mechanical and ultrasound<br />

properties of the above contrast agents, we performed<br />

2D and 3D time domain simulations to compute the<br />

acoustic wave propagation in an our UCA solution.<br />

Results: We have measured in vitro the signal-to-noise<br />

ratio, backscattered signal, and destruction by ultrasound<br />

waves of the two nano-size ultrasound contrast<br />

agents, where the applied ultrasound signal has a frequency<br />

of either 5 or 50 MHz. In addition, the comparison<br />

between 2D numerical and experimental results<br />

shows a good agreement [3] , the 3D simulations are still<br />

under ways and should permit an improvement of our<br />

predictions. Our first tests performed in vivo showed<br />

that our agents induce a significant enhancement in the<br />

backscattered signal. For targeting purpose, the surface<br />

chemistry of the first UCA particles type was modified<br />

by incorporating phospholipids (fluorescent, pegylated,<br />

and biotinylated phospholipids) in the organic phase<br />

before emulsification [2] . Microscopy shows that phospholipids<br />

are present within the shell and that the core/<br />

shell structure is preserved. The functionalization did<br />

not modify the echographic signal arising from capsules.<br />

Conclusions: These results demonstrates that these<br />

nano and polymeric capsules are suitable ultrasound<br />

contrast agent and can be easily modified<br />

for targeted therapy or molecular imaging purpose.<br />

Acknowledgement: This work is supported by a<br />

grant from ANR (n° NT05-3-42548) and EC-<br />

FP6-project DiMI (LSHB-CT-2005-512146)<br />

imaging life<br />

References:<br />

1. Pisani et al. Adv. Funct. Mater. 18 (2008) 1-9<br />

2. Diaz-Lopez et al. Biomaterials 30 (2009) 1462-1472<br />

3. Haiat & Al, J. Acoust. Soc. Am. 127 1, (2010)


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

Methods to study the interaction between aptamer probes and cell surface biomarkers<br />

Cibiel A. , Pestourie C. , Gombert K. , Janssens I. , Thézé B. , Tavitian B. , Ducongé F. .<br />

CEA,DSV,I²BM,SHFJ,INSERMU1023, Orsay, France<br />

agnes.cibiel@cea.fr<br />

Introduction: Aptamers are specific nucleic acidsbased<br />

ligands selected by an in vitro molecular evolution<br />

process (named SELEX). Recently, SELEX<br />

has been performed against living cells and allows<br />

for successful selection of aptamers against a specific<br />

transmembrane protein [1] or against cell surface<br />

biomarkers without prior knowledge of the target [2] .<br />

Here, we compared different techniques to study<br />

the interaction between aptamers and their target.<br />

Methods: For the binding experiments, 10 5 cells were<br />

first incubated with 5’- 32 P-labeled aptamers or control<br />

sequences for 15 minutes at 37°C and then washed.<br />

Bound sequences were recovered in 400µL of lysis<br />

buffer and the amount of radioactivity was counted.<br />

To optimise this protocol, we evaluated the number<br />

of washings and different combinations of unspecific<br />

competitors (tRNA, ssDNA and Polyinosine) at different<br />

concentrations. This allowed us to determine the<br />

protocol that yielded the highest signal/background<br />

ratio and to measure the affinity and the Cmax of different<br />

aptamers. In addition, an automation of the<br />

protocol has been performed. For microscopy analysis,<br />

aptamers were first labelled using Ulisys Alexa fluor<br />

546. Then, they were imaged using epifluorescence<br />

microscopy (Axio Observer Z1 Zeiss) at different time<br />

points during incubation (up to 40 minutes).<br />

Results:We observed that 5 washing cycles during the<br />

binding experiments were enough to decrease the<br />

background. In addition, we observed that the best<br />

combination of competitors seemed to be dependent<br />

on the cell type. In contrast, the optimal concentration<br />

of competitors was always around 100µg/ml. The use<br />

of higher concentrations did not have any effect suggesting<br />

that unspecific targets were saturated at this<br />

concentration. Finally, the use of competitor during<br />

pre-incubation and/or incubation of aptamers had the<br />

same impact on the signal/background ratio. Labelling<br />

aptamers with different amounts of Ulysis Alexa<br />

546 showed that when the ratio of Alexa/aptamer was<br />

increased, fluorescence intensity decreased, due to<br />

self-quenching phenomenon. This labelling method<br />

has been optimised to study the cellular localisation<br />

of aptamers by microscopy. In addition to providing<br />

information on the membrane or intracellular location<br />

of aptamers (after internalisation), microscopy<br />

allowed for evaluation of the kinetics of association<br />

between the aptamer and the target.<br />

Conclusions: The use of the above described two<br />

methods allowed us to obtain complementary results.<br />

Binding experiments led to the quantification<br />

of the kD or the number of targets on the cell surface<br />

whereas microscopy provided the localisation<br />

of the aptamer on the cell surface and its evolution<br />

over time. Aptamers selected against cell surface<br />

biomarkers could have many applications in the biomedical<br />

field (e.g. cell sorting, biomarker discovery,<br />

imaging, drugs design, etc.). Our methods can be<br />

used to rapidly assess their potential.<br />

Acknowledgements: This work was supported by<br />

grants from the European Molecular Imaging Laboratory<br />

(EMIL) network (EU contract LSH-2004-<br />

503569), the FMT-XCT program (EU contract<br />

201792) and l’Agence Nationale pour la Recherche<br />

(project ANR-Emergence ARTIC and ANR-TecSan<br />

DOT-IMAGER). AC was supported by a fellowship<br />

(Irtélis) from the CEA.<br />

References:<br />

1. Cerchia L et al. (2005) PLoS Biol 3(4)<br />

2. Pestourie C, Tavitian B, Duconge F (2005) Biochimie<br />

87(9-10): 921-930<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

P-060<br />

poStEr<br />

PROBE DESIGN


WarSaW, poland May 26 – 29, 2010<br />

P-061 MRI intelligent contrast agents as enzyme responsive nanosystems<br />

174<br />

Figueiredo S. (1) , Cittadino E. (1) , Terreno E. (1) , Moreira J.N. (2) , Geraldes C. F. (3) , Aime S. (1) .<br />

(1) Department of Chemistry IFM and Molecular Imaging Center, Torino, Italy<br />

(2) cLaboratory of Pharmaceutical Technology, Faculty of Pharmacy and Center for Neurosciences and Cell Biology, University of Coimbra, , Portugal<br />

(3) Department of Life Sciences, Faculty of Sciences and Technology, and Center for Neurosciences and Cell Biology, University of Coimbra, Portugal<br />

Saharic@gmail.com<br />

Introduction: When Magnetic Resonance is the imaging<br />

modality of choice it is necessary to design<br />

highly sensitive systems in order to overcome MRI<br />

relatively low sensitivity. We have envisaged an approach<br />

to enzyme-responsive agents based on the<br />

use of liposomes loaded in the aqueous cavity with a<br />

high number of paramagnetic complexes. Liposomes<br />

are self-assembled vesicles formed by saturated and<br />

unsaturated phospholipids commonly used in drug<br />

delivery. The overall relaxation enhancement of solvent<br />

water protons depends upon the permeability/<br />

disruption of the liposome membrane to water molecules.<br />

Our work has addressed the objective of i)<br />

modifying the permeability of liposome membrane<br />

thus pursuing an enhancement of the observed proton<br />

relaxation rate upon the enzymatic cleavage of<br />

peptides covalently bound to the phospholipid moieties<br />

or ii)promoting the disruption of low relaxivity<br />

aggregates formed by the binding capabilities of a<br />

macromolecular substrate that is selectively cleaved<br />

by the enzyme.<br />

Methods: Paramagnetic liposomes were prepared using<br />

the proper membrane by the thin film hydration<br />

method followed by extrusion. The lipidic film was<br />

hydrated with a 200mM solution of GdHPDO3A,<br />

followed by dialysis to remove non-entrapped material.<br />

The T1 measurements in vitro were carried<br />

out at 0.5T on a Stelar Spinmaster, whereas the in<br />

vivo measurements were acquired at 7T on a Bruker<br />

Avance 300 spectrometer. The temporal evolution of<br />

T1 contrast was determined in vivo after intratumor<br />

injection to mice bearing xenografted B16 melanoma.<br />

Results: i)The relaxometric properties of the liposomes<br />

loaded with the lipopeptide was tested in vitro<br />

measuring the r1 over time of three samples: a) the<br />

suspension of liposomes incorporating the lipopeptide<br />

in the presence (I) and absence (II) of MMP1<br />

and b) liposomes incorporating the stearic acid in<br />

the presence of MMP1. The results reported indicated<br />

that sample I showed a slight increase in the relaxivity<br />

likely due to partial instability of the liposome.<br />

In the presence of MMP, a more pronounced enhancement<br />

was observed. Importantly, the liposomal<br />

sample in which the incorporated lipopeptide was<br />

replaced by stearic acid did not show any significant<br />

imaging life<br />

enhancement, confirming that the enhancement observed<br />

for sample a-I) is related to the MMP1 activity.<br />

This different behaviour can be well appreciated<br />

by looking at the different contrast exhibited in the<br />

corresponding T1weighted MR image. In vivo kinetic<br />

experiment following the intratumor injection of<br />

the lipopeptide-based paramagnetic liposome indicated<br />

a rapid washout of the imaging probe, consistent<br />

with a relevant release of the Gd(III) complex in<br />

the extracellular fluid, where MMPs accumulates. ii)<br />

The interaction of anionic liposomes and protamine<br />

yields supramolecular adduct with low relaxivity. The<br />

action of trypsin causes the digestion of protamine<br />

and the consequent de-assembly of the adduct. The<br />

process is accompanied by an overall relaxation enhancement<br />

as consequence of the recovering of the<br />

original permeability of the liposome membrane to<br />

water molecules. An illustrative example of the utilization<br />

of this responsive probe consists in the entrapment<br />

of the supramolecular assembly in alginate<br />

vesicles. The detected change in r1 due to trypsin is<br />

correlated to the one in the absence of alginate. Thus,<br />

an in vivo exploitation relies on the entrapment of<br />

micron-sized particles into an alginate matrix that<br />

has often been considered as a bio-compatible device<br />

in cell-based therapies.<br />

Conclusions: We have now envisaged a molecular<br />

probe that work as an enzymatic substrate in a particular<br />

microenvironment, hence enhancing the MRI signal<br />

as a function of enzymatic activity. As further goal,<br />

an anti-tumoral drug will be co-encapsulated with the<br />

MRI probe, allowing supervise the tumoral therapy.


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

Standardization of molecular PBCA-microbubbles for routine use<br />

Fokong S. , Liu Z. , Gätjens J. , Kiessling F. .<br />

Helmholtz Institute, Aachen, Germany<br />

sfokong@ukaachen.de<br />

Introduction: To develop standardized highly monodisperse<br />

targeted polybutylcyanoacrylate (PBCA)microbubbles<br />

for contrast enhanced molecular<br />

ultrasound imaging.<br />

Methods: PBCA-microbubbles were produced by<br />

mechanical agitation and size isolated by centrifugation.<br />

Physical parameters of the size optimized<br />

microbubbles and regular (Sonovist) microbubbles<br />

were compared. Targeting of the microbubbles for<br />

molecular imaging was achieved by covalently binding<br />

streptavidin molecules onto the shell of surface<br />

activated PBCA-microbubbles and the subsequent<br />

attachment of biotinylated markers. The amount of<br />

targeting ligands on the surface of the microbubbles<br />

was quantified using a fluorescence activated cell<br />

sorter (FACS). The suitability of these microbubbles<br />

for destructive imaging methods like power Doppler<br />

ultrasound, used for the evaluation of the degree of<br />

angiogenesis and the effect of antiangiogenic therapy,<br />

was investigated in gelatin phantoms.<br />

Results: Following a new protocol for the synthesis<br />

of PBCA microbubbles, highly monodisperse populations<br />

could be isolated. Curves obtained from<br />

particle sizer measurements showed size isolated<br />

microbubbles which are normally distributed with<br />

a narrower standard deviation (> 97% ± 1.0-3.0 µm)<br />

in comparison to regular Sonovist microbubbles<br />

(> 97% ± 0.2–10 µm). The high monodispersity was<br />

also confirmed by SEM (scanning electron microscopy)<br />

pictures. The population distribution remained<br />

constant even with prolonged storage in solution<br />

(over 4 months), indicating high stability. Comparison<br />

of the resonance frequencies and the persistence<br />

in an ultrasound field showed a significantly lower<br />

resonance frequency and a lower persistence in the<br />

ultrasound field for the monodispersed microbubbles<br />

compared to the polydispersed microbubbles.<br />

The high monodispersity was maintained even with<br />

covalent coupling of streptavidin molecules onto the<br />

shell of the microbubbles. A quantification of the<br />

number of streptavidin molecules on the surface by<br />

the use of a FACS gave approximately 2 x 10 4 streptavidin<br />

molecules per microbubble. This number can<br />

be tuned according to the degree of surface activation<br />

of the microbubbles. Destructive imaging using<br />

SPAQ (Sensitive Particle Acoustic Quantification)<br />

[1] was also possible with these monodispersed microbubbles<br />

showing a highly reproducible destruction<br />

with high mechanical index ultrasound waves.<br />

Conclusions: Size optimized non modified PBCAmicrobubbles<br />

are more suitable for use as ultrasound<br />

contrast agents in comparison to regular Sonovist<br />

microbubbles. The simplified synthetic protocol allows<br />

for a speed up in the synthesis and purification<br />

process (6 days vs. 3 h). The low resonance frequency<br />

and persistence lead to more efficient detection using<br />

power Doppler techniques. Also, given the relative<br />

high number of targeting ligands which can be<br />

easily attached to their surface, and the possibility to<br />

efficiently quantify their amounts in a region of interest,<br />

they are very suitable for molecular imaging of<br />

intravascular targets. In summary, the tuned properties<br />

of the microbubbles presented in this study, will<br />

make them very interesting as building blocks for<br />

tailoring of specific contrast agents for ultrasound.<br />

Acknowledgement: This work is supported by the<br />

German Federal Ministry of Education and Research<br />

(BMBF-0315017).<br />

References:<br />

1. M. Reinhardt, P. Hauff, A. Briel, V. Uhlendorf, M. Schirner,<br />

Sensitive particle acoustic quantification (SPAQ),<br />

Investigative Radiology 40 (2005) 2-7.<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

P-062<br />

poStEr<br />

PROBE DESIGN


WarSaW, poland May 26 – 29, 2010<br />

P-063 Novel Gd(III)-based probes for MR molecular imaging of matrix metalloproteinases<br />

176<br />

Gringeri C. (1) , Catanzaro V. (2) , Menchise V. (3) , Digilio G. (1) , Aime S. (2) .<br />

(1) Department of Environmental and Life Sciences, University of Eastern Piedmont “A. Avogadro”, Alessandria, Italy<br />

(2) Department of Chemisty IFM & Center for Molecular Imaging, University of Turin, Italy<br />

(3) Institute for Biostructures and Bioimages (CNR) c/o Molecular Biotechnology Center (University of Turin), Italy<br />

cgringeri@gmail.com<br />

Introduction: The assessment of the activity of a<br />

selected panel of MMPs in a given tissue or anatomical<br />

district is very important for the typization<br />

and staging of cancer and autoimmune diseases,<br />

and for the evaluation of the efficacy of therapies.<br />

[1] “Molecular Imaging” is a relatively new and rapidly<br />

growing branch of diagnostic imaging devoted<br />

to visualize tissue specific patho-physiological processes<br />

on the basis of their cellular/biochemical molecular<br />

signatures. MRI is one of the most clinically<br />

relevant imaging techniques, because of its great<br />

spatial resolution (


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

Functionalization of nanoparticles for molecular imaging; a covalent approach<br />

Herranz F. (1) , Salinas B. (1) , Rosell Y. (1) , Desco M. (2) , Ruiz-Cabello J. (1) .<br />

(1) Universidad Complutense de Madrid- CIBERES, Madrid, Spain<br />

(2) Hospital Gregorio Marañón, Spain<br />

fherranz@pdi.ucm.es<br />

Introduction: The key point in the development<br />

of new nanoparticles (NPs) for imaging is the<br />

surface functionalization. By the attachment of<br />

new molecules one should, ideally, provide stability<br />

in physiological conditions, with a narrow<br />

size distribution and a functional group for the<br />

binding of biomolecules. These changes on the<br />

NPs surface can be made by weak non-specific<br />

interactions or by strong specific covalent bonds.<br />

Methods: Iron oxide nanoparticles were synthesized<br />

by the decomposition of organic precursors redendering<br />

hydrophobic Fe 3 O 4 NPs. These nanoparticles<br />

have oleic acid as surfactant, providing excellent size<br />

distribution, crystallinity and stability. To transfer<br />

the NPs to water the chemical structure of the oleic<br />

acid was modified by chemical methods, obtaining<br />

water stable NPs ready for further modification.<br />

Results: Fe 3 O 4 NPs were synthesized (7 ± 1 nm) by<br />

the decomposition of organic precursors. The surfactant<br />

of the NPs (oleic acid) was oxidized by means<br />

of potassium permanganate in a two phase system.<br />

This way we obtained water stable, carboxylic acid<br />

functionalized NPs, that were fully characterized<br />

(47 ± 4 nm, Zeta potential -46 mV, r 1 =4 s -1 mM -1<br />

and r 2 = 115 s -1 mM -1 , FTIR and 1 H-NMR). 1 These<br />

NPs are stable in buffer conditions and are being<br />

used for in vivo MRI. Besides, we took advantage of<br />

the carboxylic acid on the surface to covalently funcionalize<br />

the NPs with different molecules and dyes,<br />

like glucose, EDANS, 2 and streptavidin-alexa647.<br />

Conclusions: Here we report a new approach for the<br />

covalent functionalization of superparamagnetic<br />

nanoparticles. We demonstrate the usefulness of<br />

the method with several examples of covalent functionalization<br />

with different molecules and in vivo<br />

MRI use.<br />

Acknowledgement: This work was supported in part<br />

by MAT2008-01489 and SAF2008-0512-C02-01.<br />

References:<br />

1. Herranz, F.; Morales, M. P.; Roca, A. G.; Desco, M.; Ruiz-<br />

Cabello, J., Chemistry A European Journal 2008, 14,<br />

(30), 9126-30.<br />

2. Herranz, F.; Morales, M. P.; Roca, A. G.; Vilar, R.; Ruiz-<br />

Cabello, J., Contrast Media Mol. Imaging 2008, 3, (6),<br />

215-22.<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

P-064<br />

poStEr<br />

PROBE DESIGN


178<br />

WarSaW, poland May 26 – 29, 2010<br />

P-065 Comparison of different chelating systems for the synthesis of Ga-68 labelled peptides for<br />

molecular imaging using RGD-peptides as model compound<br />

Knetsch P. (1) , Petrik M. (1) , Rangger C. (1) , Griessinger C. (2) , Fani M. (3) , Wester H.J. (4) , Pichler B. (2) , Pietzsch H.J. (5) ,<br />

Decristoforo C. (1) , Haubner R. (1) .<br />

(1) Medical University Innsbruck, Innsbruck, Austria<br />

(2) University of Tuebingen, Germany<br />

(3) University of Freiburg, Germany<br />

(4) Technische Universitaet Muenchen, Germany<br />

(5) Research Center Dresden-Rossendorf, Germany<br />

peter.knetsch@i-med.ac.at<br />

Introduction: Due to its increasing availability<br />

Ga-68 attracts increasing interest in molecular imaging<br />

with PET. Moreover, due to the straightforward<br />

labelling protocols especially for labelling of<br />

peptides this is an interesting alternative to F-18<br />

labelling strategies. Here the imaging properties<br />

of c(RGDfK) conjugated to different chelating systems<br />

are compared.<br />

Methods: Peptides were synthesised using standard<br />

SPPS protocols. After cyclisation in solution<br />

and selective deprotection of the amino function<br />

of the lysine the chelating moieties were conjugated<br />

via in situ activation. The chelating systems<br />

include 1,4,7,10-tetraazacyclododecane-1,4,7,10acetic<br />

acid (DOTA), a 1,4,7–triaaza-10-oxocyclododecane-1,4,7-acetic<br />

acid derivative (B505),<br />

1,4,7-triaazacyclononane-4,7-acetic acid-1-2-glutaric<br />

acid (NODAGA), and a tris(2-mercaptoethyl)<br />

amine derivative (NS3). Labelling was carried out<br />

using the fractionated elution method in sodium<br />

acetate or phosphate buffer, respectively. In vitro<br />

evaluation included determination of the partition<br />

coefficient, protein binding properties, metabolic<br />

stability, binding affinity, and cell uptake characteristics.<br />

In vivo evaluation was carried out using nude<br />

mice bearing alpha(v)beta3-positive and alpha(v)<br />

beta3–negative tumours. For all tracer biodistribution<br />

data were collected. For the most promising<br />

also small animal PET imaging was carried out.<br />

Results: All peptides could be labelled with Ga-68<br />

in good radiochemical yields. Labelling of NOD-<br />

AGA-RGD could be achieved even at room temperature.<br />

Whereas labelling of NS3-RGD has to be<br />

followed by Seppak separation to obtain the product<br />

in high radiochemical purity. The compounds<br />

showed comparable partition coefficients, binding<br />

affinity for the alpha(v)beta3 integrin as well as receptor<br />

specific uptake. However, great differences<br />

were found in the protein binding properties. Out<br />

of the four peptides tested only NODAGA-RGD<br />

showed low protein binding. This is also reflected<br />

in the biodistribution data. Lowest activity concentration<br />

in blood and best tumour/background ratios<br />

imaging life<br />

were found for NODAGA-RGD. Subsequent small<br />

animal imaging showed best imaging properties<br />

for NODAGA-RGD, which seems to be comparable<br />

with F-18-Galacto-RGD.<br />

Conclusions: In this series NODAGA-RGD revealed<br />

most promising properties for molecular<br />

imaging applications. Easy radiolabelling at room<br />

temperature, low amount of protein bound activity<br />

and the resulting lower activity concentration<br />

found in blood compared to the other compounds<br />

makes it to an interesting alternative to F-18-Galacto-RGD<br />

for imaging alpha(v)beta3 expression.<br />

However, the general advantage of NOTA derivatives<br />

for imaging purposes have to be confirmed by<br />

additional studies using other peptide structures.


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

GdDOTA-PIB: a potential MRI marker for Alzheimer’s disease<br />

Martins A. (1) , Morfin J.F. (1) , Hamplova A. (1) , Kubicek V. (1) , Suzenet F. (2) , Salerno M. (3) , Lazar A. (4) , Duyckaerts C. (4) ,<br />

Geraldes C. (5) , Toth E. (1) .<br />

(1) Centre de Biophysique Moléculaire,CNRS, Orléans, France<br />

(2) Université d’Orléans, France<br />

(3) Université Paris 13, France<br />

(4) Hôpital de la Pitié-Salpêtrière, France<br />

(5) University of Coimbra, Portugal<br />

andre.martins@cnrs-orleans.fr<br />

Introduction: Alzheimer’s disease (AD) is the most<br />

frequent form of intellectual deterioration in elderly<br />

individuals, characterized by the brain deposition of<br />

amyloid plaques and neurofibrillary tangles. Early detection<br />

of the β-amyloid (Aβ) deposits in vivo is very<br />

difficult. Recently 11 C-radiolabeled small-molecules<br />

have been developed, capable of entering the brain<br />

and specifically targeting amyloid plaques for imaging<br />

with PET, such as several Thioflavin T derivatives<br />

[1-2]. In particular, the uncharged analogue 6-OH-<br />

BTA-1 (Pittsburgh compound B - PIB) is highly efficient<br />

both in crossing the BBB and in selective binding<br />

to AD amyloid aggregates. A major limitation of PET<br />

is the requirement for the markers to be labelled with<br />

short-lived isotopes. The use of Aβ marker linked to<br />

a MRI CA would constitute an attractive noninvasive<br />

in vivo imaging approach. Recently, Poduslo used CA<br />

aided MRI to image AD plaques with Gd(III)DTPA<br />

conjugated to a putrescine-modified human Aβ peptide<br />

able to cross the BBB and selectively target individual<br />

amyloid plaques in the brain of Alzheimer’s<br />

disease transgenic mice. Nevertheless, due to its large<br />

size, several days (weeks) of incubation with the CA<br />

are necessary to obtain the labeling of amyloid plaques<br />

in transgenic mouse brain in vivo. In an attempt to label<br />

Aβ plaques using small metal complexes for the<br />

diagnostics of Alzheimer disease, we synthesized a<br />

PIB-derivative of DOTA.<br />

Methods: DOTA-PIB was synthesized using a new,<br />

versatile strategy. The Gd 3+ complex has been characterized<br />

by relaxometric methods. The compound<br />

forms micelles in aqueous solution; the critical micellar<br />

concentration has been measured. Experiments<br />

on human brain slices have been carried out to assess<br />

binding of the compound to the β-amyloid deposits.<br />

HOOC<br />

N<br />

N<br />

O<br />

N<br />

H<br />

N N<br />

HOOC COOH<br />

DOTA-PIB<br />

S<br />

N<br />

O<br />

Results: The 1 H NMRD profiles evidence aggregation<br />

of the GdDOTA-PIB complex in aqueous solution,<br />

with a cmc of ~1.5 mM. The preliminary experiments<br />

on human brain slices show good binding affinity of<br />

the LnDOTA-PIB compound towards the amyloid<br />

plaques.<br />

Conclusions: We synthesized and investigated a<br />

novel Gd 3+ complex with good binding properties to<br />

β-amyloid deposits.<br />

Acknowledgement: We thank the support from the<br />

F.C.T. Portugal (project SFRH / BD / 46370 / 2008 and<br />

COST D38.<br />

References:<br />

1. Mathis CA, Bacskai BJ, Kajdasz ST, McLellan ME, Frosch<br />

MP, Hyman BT, Holt DP, Wang Y, Huang GF, Debnath ML,<br />

Klunk WE. (2002) Bioorg Med Chem Lett.12(3):295-8.<br />

2. Nesterov EE, Skoch J, Hyman BT, Klunk WE, Bacskai<br />

BJ, Swager TM. (2005) Angew Chem Int Ed Engl.<br />

26(34):5452-6.<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

P-066<br />

poStEr<br />

PROBE DESIGN


WarSaW, poland May 26 – 29, 2010<br />

P-067 A comparative study of the self-elimination of para-aminobenzyl alcohol and hemithioaminalbased<br />

linkers. Application to the design of Caspase-3 sensitive pro-fluorescent probes<br />

180<br />

Meyer Y. (1) , Richard J. A. (1) , Delest B. (2) , Noack P. (2) , Renard P. Y. (1) , Romieu A. (1) .<br />

(1) University of Rouen, Mont Saint Aignan, France<br />

(2) Quidd: smart molecular imaging, Mont Saint Aignan, France<br />

yvesmeyer@estvideo.fr<br />

Introduction: This study focuses on the disassembly-behavior<br />

of self-immolative pro-fluorescent<br />

linkers under physiological conditions occurring<br />

via an enzyme-initiated domino reaction. The targeted<br />

linkers are based on para-aminobenzylalcohol<br />

(PABA) or hemithioaminal derivatives of para-carboxybenzaldehyde<br />

or glyoxilic acid. We found that a<br />

fine tuning of the kinetic properties can be obtained<br />

through the modulation of the linker structure, giving<br />

either a fast signal response or customizable<br />

systems suitable for the design of protease-sensitive<br />

fluorogenic probes or prodrug systems.<br />

Methods: Five model compounds bearing specific<br />

self-immolative linkers were synthesised and all<br />

integrate both: (1) a phenylacetamide moiety to be<br />

cleaved by Penicillin G Acylase (PGA) functionning<br />

as the triggering agent, and (2) a masked umbelliferone<br />

(i.e., 7-hydroxycoumarin) unit to be released as<br />

the final product and to elicit turn-on fluorescence.<br />

All these fluorogenic probes were incubated at the<br />

same concentration (3 µM) at 37°C with PGA (0.12<br />

U) in phosphate buffered saline (PBS, pH 7.5) and<br />

the corresponding fluorescence time-courses were<br />

recorded at λ = 460 nm.<br />

Results: Comparison of the resulting fluorescence<br />

recovery curves clearly indicates that the decomposition<br />

of PABA-based self-immolative linkers was<br />

faster than with the hemithioaminal counterparts.<br />

No nonspecific cleavage of these pro-fluorescent<br />

probes was detected in control reactions when incubated<br />

only in PBS. These results demonstrate that<br />

the use of PABA or hemithioaminal based linkers<br />

affords enzyme-reactive pro-fluorophores with<br />

high chemical stability. Despite the slower release<br />

of umbelliferone from the hemithioaminal probe<br />

derived from glyoxylic acid, this linker presents a<br />

peptide-like structure able to be readily and widely<br />

functionalised.<br />

Conclusions: PABA derivatives will be used for getting<br />

fast-response probes whereas the hemithioaminal<br />

spacers will be preferred for the construction of<br />

finely tunable (bio)functionalised probes. A first<br />

generation of Caspase-3 (apoptosis process) sensitive<br />

pro-fluorescent probes using PABA linkers has<br />

imaging life<br />

been obtained (AcDEVD-PABA-Umbelliferone) and<br />

in vitro fluorescence assay was performed with the<br />

corresponding recombinant human protease: 10fold<br />

increase of the enzyme velocity was obtained as<br />

compared to the commercially available profluorophore<br />

AcDEVD-AMC.<br />

Acknowledgement: This work is supported by CNRS,<br />

Quidd: Smart Molecular Imaging and La Région<br />

Haute-Normandie.<br />

References:<br />

1. R. Erez and D. Shabat, Org. & Biomol. Chem., 2008, 15,<br />

2669-2672. H. Y. Lee, X. Jiang and D. Lee, Org. Lett.,<br />

2009, 11, 2065-2068.<br />

2. C. Fossey, A.-H. Vu, A. Vidu, I. Zarafu, D. Laduree, S.<br />

Schmidt, G. Laumond and A.-M. Aubertin, J. Enzyme<br />

Inhib. Med. Chem., 2007, 22, 591. C. Fossey, N.-T. Huynh,<br />

A.-H. Vu, A. Vidu, I. Zarafu, D. Laduree, S. Schmidt, G.<br />

Laumond and A.-M. Aubertin, J. Enzyme Inhib. Med.<br />

Chem., 2007, 22, 608.<br />

3. Y. Meyer, J. A. Richard, M. Massonneau, P. Y. Renard and<br />

A. Romieu, Org. Lett., 2008, 10, 1517-1520.<br />

4. Y. Meyer, J. A. Richard, B. Delest, P. Noack, P. Y. Renard<br />

and A. Romieu, Org. & Biomol. Chem., 2010, In Press<br />

DOI: 10.1039/b926316k.


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

Olefin Metathesis for the functionalization of superparamagnetic nanoparticles<br />

Salinas B. (1) , Ruiz-Cabello J. (1) , Desco M. (2) , Herranz F. (1) .<br />

(1) Universidad Complutense Madrid- CIBERES, Madrid, Spain<br />

(2) Hospital Gregorio Marañón, Spain<br />

besalina@pdi.ucm.es<br />

Introduction: One of the most important points in<br />

the synthesis of new nanoparticles (NPs) for molecular<br />

imaging is the functionalization of the surface<br />

with new molecules. There is a necessity for new approaches,<br />

allowing more versatile synthesis and the<br />

introduction of biomolecules for specific interactions.<br />

This modification of the surface should be done covalently<br />

in mild conditions. Olefin metathesis offers<br />

many of those features thanks to the new family of<br />

catalysts, especially Hoveyda-Grubbs 2 nd generation. 1<br />

Methods: Iron oxide nanoparticles were synthesized<br />

by the decomposition of organic precursors redendering<br />

hydrophobic Fe 3 O 4 NPs, with oleic acid as<br />

surfactant. We carried out the olefin metathesis between<br />

the double bond in oleic acid structure and<br />

methyl acrylate. After the hydrolysis of the ester,<br />

water stable dispersion of superparamagnetic nanoparticles<br />

were obtained.<br />

Results: Fe 3 O 4 NPs were synthesized (10 ± 2 nm) by<br />

the decomposition of organic precursors. 2 The NPs<br />

were reacted in dry conditions with methyl acrylate<br />

in the presence of catalytic amounts (4%mol) of<br />

Hoveyda-Grubbs 2 nd generation catalyst. After hydrolysis<br />

of the ester water stable NPs, with narrow<br />

size distribution (30 ± 5 nm) and a Zeta potential<br />

corresponding to the introduced carboxylic acid<br />

(-46 mV) were obtained. The presence of the new<br />

diacid on the surface was confirmed by 1 H-NMR<br />

and FTIR.<br />

Conclusions: Here we report, for the first time,<br />

the use of olefin metathesis for the synthesis of<br />

water stable Fe 3 O 4 NPs with excellent yield and<br />

selectivity. This synthesis opens up the possibility<br />

for the attachment of new molecules in one step<br />

from the hydrophobic nanoparticles in a mild<br />

and selective way.<br />

Acknowledgement: This work was supported in part<br />

by MAT2008-01489 and SAF2008-0512-C02-01.<br />

References:<br />

1. Rybak, A.; Meier, M. A. Green Chem. 2008, 1099-1104.<br />

2. Herranz, F.; Morales, M. P.; Roca, A. G.; Desco, M.; Ruiz-<br />

Cabello, J., Chemistry A European Journal 2008, 14,<br />

(30), 9126-30.<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

P-068<br />

poStEr<br />

PROBE DESIGN


182<br />

WarSaW, poland May 26 – 29, 2010<br />

P-069 Pyridine-based lanthanide complexes : towards bimodal agents operating as near infrared<br />

luminescent and MRI reporters<br />

Toth E. (1) , Bonnet C. (1) , Villette S. (1) , Suzenet F. (2) , Buron F. (2) , Shade C. (3) , Petoud S. (1) .<br />

(1) Centre de Biophysique Moléculaire, CNRS, Orleans, France<br />

(2) Université d’Orléans, France<br />

(3) University of Pittsburgh, USA<br />

eva.jakabtoth@cnrs-orleans.fr<br />

Introduction: Among the state of the art bioimaging<br />

modalities, some are characterized by high<br />

resolution but low sensitivity (magnetic resonance<br />

imaging, MRI) and others by high sensitivity but<br />

low macroscopic resolution (optical imaging). Luminescent/MRI<br />

bimodal imaging offers the advantage<br />

of combining the high resolution of MRI<br />

with the high sensitivity of luminescence and the<br />

development of contrast agents active for both<br />

techniques is of prime importance. Lanthanides<br />

offer unique opportunity to develop such bimodal<br />

contrast agents given their magnetic and optical<br />

properties. Nevertheless, it was long thought that<br />

the conditions required for both applications were<br />

non-compatible. Here, we report on a versatile pyridine-based<br />

scaffold for Ln 3+ complexation where<br />

MRI and near infrared (NIR) luminescence requirements<br />

are both satisfied using the same ligand.<br />

Methods: The synthesis of the ligands and the complexes<br />

will be briefly described. Potentiometric<br />

titrations have been performed to assess to thermodynamic<br />

stability of the complexes, together<br />

with kinetic measurements to quantify their inertness.<br />

To characterize the MRI properties, the exchange<br />

rate of the water molecules directly bound<br />

the Gd 3+ was determined by measuring 17 O longitudinal<br />

relaxation times. Finally the NIR spectra<br />

of the corresponding Nd 3+ and Yb 3+ were recorded,<br />

as well as the lifetimes of the excited states and the<br />

quantum yield of the complexes to quantify their<br />

luminescent properties.<br />

Results: The bishydrated complexes are found to be<br />

thermodynamically stable and the ligands show a significant<br />

selectivity for Ln 3+ over endogenous cations<br />

such as Zn 2+ , Ca 2+ and Cu 2+ . The kinetic inertness is<br />

also remarkable for such bishydrated complexes. The<br />

chelates do not form ternary complexes with endogenous<br />

donors which does not limit relaxivity in biological<br />

media. All the complexes give rise to NIR emission<br />

and the quantum yields are remarkable. 1 They are in<br />

the same range as those of non-hydrated complexes optimized<br />

for fully protecting the NIR emitting Ln 3+ for<br />

aqueous applications. The modification of the pyridine<br />

into quinoline was successful in shifting the excitation<br />

wavelength of the system towards higher values.<br />

imaging life<br />

Conclusions: The pyridine synthon is a prime candidate<br />

for the development of bimodal NIR/MRI imaging<br />

probes, as the bishydrated Ln 3+ complexes are thermodynamically<br />

and kinetically stable and display a high<br />

NIR quantum yield. The successful modification of the<br />

pyridine into a quinoline did not modify the thermodynamic<br />

properties of the complexes, but it resulted<br />

in a shift of the excitation energy towards lower values<br />

preventing damages to biological samples and allowing<br />

deeper tissue penetration of the excitation photons. The<br />

pyridine platform offers also easy routes for coupling<br />

the probes to biological vectors and optimizing the<br />

MRI properties.<br />

Acknowledgement: This work was financially supported<br />

by the Institut National du Cancer, La Ligue contre<br />

le Cancer, France, and was carried out in the frame of<br />

the COST action D38.<br />

References:<br />

1. L. Pellegatti, J. Zhang, B. Drahos, S. Villette, F. Suzenet,<br />

G. Guillaumet, S. Petoud, E. Toth, Chem. Commun.,<br />

2008, 6591-6593.<br />

2. S. Comby, D. Imbert, C. Vandevyver, J.-C. G. Bünzli,<br />

Chem. Eur. J., 2007, 13, 936.


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

Delivery of multiple F-18 tracers from a single automated platform (FASTlab TM synthesizer)<br />

Bhalla R. .<br />

GE Healthcare, Amersham, United Kingdom<br />

rajiv.bhalla@ge.com<br />

Introduction: The majority of F-18 radiotracers<br />

prepared in the clinic are synthesized on be-spoke<br />

automated rigs, which require modifications (and<br />

time) in order to switch to the production of different<br />

tracers. The increasing demand to facilitate<br />

the production of multiple F-18 tracers from a single<br />

clinical site (and preferably from a single Hot Cell)<br />

has necessitated the need to develop an easy to use<br />

automated platform which can readily accommodate<br />

the production of multiple F-18 tracers.<br />

O<br />

18<br />

TsO n F<br />

18<br />

F<br />

O<br />

R<br />

Fig1: Example of radiochemistry on FASTlabTM<br />

18<br />

R n F<br />

Methods: The FASTlab TM is an automated PET radiochemistry<br />

synthesis platform incorporating a<br />

disposable cassette. The disposable cassette contains<br />

a reaction vessel, pre-filled reagent vials and SPE<br />

cartridge(s). The FASTlab TM performs transfers (of<br />

F-18, reagents, solvents etc) in and out of the reaction<br />

vessel using a combinations of syringe drivers, pressure<br />

and vacuum – this combination of processes provides<br />

a high degree of control, allowing manipulations to be<br />

carried out with a high degree of precision and accuracy.<br />

The F-18 labelling is preformed in the reaction vessel<br />

and subsequent purification is performed either on the<br />

cassette using SPE cartridges or externally via a HPLC.<br />

Results: Figure 1 presents a snapshot of some the<br />

chemistry which has been successfully transitioned<br />

onto the FASTlab TM , demonstrating that this platform<br />

is capable of accommodating a diverse range<br />

of chemistry. Examples of F-18 tracers transferred<br />

to the FASTlab TM and currently in use in the clinic<br />

will be presented.<br />

N<br />

R<br />

18<br />

E n F<br />

R<br />

18<br />

F<br />

18<br />

F<br />

Synthon chemistry<br />

Fluoroalkylkation<br />

Direct labelling<br />

S N 2<br />

Direct labelling<br />

S N Ar<br />

Synthon chemistry<br />

Oxime synthesis<br />

Conclusions: FASTlab TM is an automated synthesizer<br />

which is capable of producing GMP quality F-18<br />

tracers. We have demonstrated the versatility of<br />

FASTlab TM by transitioning a wide variety of chemistry<br />

onto the platform to produce a large number of<br />

F-18 tracers, many of which are now being assessed<br />

in the clinic. We are now focussing on expanding<br />

the range of chemistry and the number of tracers<br />

available on FASTlab TM (both GE Healthcare proprietary<br />

and non proprietary).<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

P-070<br />

poStEr<br />

TECHNOLOGY


184<br />

WarSaW, poland May 26 – 29, 2010<br />

P-071 Monte-carlo modelling of a silicon detector insert combined with a PET scanner<br />

Brzezinski K. , Oliver J.F. , Llosá G. , Solevi P. , Linhart V. , Cabello J. , Lacasta C. , Rafecas M. .<br />

Instituto de Física corpuscular, CSIC/Universidad de Valencia, Spain<br />

brzezinski@ific.uv.es<br />

Introduction: Recent works have explored the capabilities<br />

of insert devices to improve the performance<br />

of clinical [1,2] as well as small animal [3,4] PET scanners.<br />

The development of a high resolution probe to<br />

be mounted inside a conventional PET scanner to<br />

increase its spatial resolution and sensitivity is one of<br />

the goals of the EU project MADEIRA. This paper describes<br />

the modelling of the MADEIRA set-up using<br />

Monte-Carlo (MC) techniques. These techniques are<br />

vital for optimizing the configuration of the proposed<br />

system by maximizing efficiency and spatial resolution.<br />

Furthermore, simulations allow characterization<br />

of the system and are useful for modelling its physical<br />

response, which can be included in image reconstruction.<br />

In the probe-and-ring system, a pair of annihilation<br />

photons has several ways of being detected. This<br />

work focuses on the classification and quantification<br />

of the basic detection modes.<br />

Methods: A preliminary study was conducted to validate<br />

the physics models of GEANT4 for the purpose of<br />

modelling the silicon (Si) detector probe. One layer of<br />

a Si detector was irradiated by a Ba-133 source and energy<br />

spectra measured. The full probe-and-ring system<br />

was modelled using the MC toolkit GATE (based on<br />

GEANT4). The model includes a probe consisting of<br />

ten layers of 26x40 pixel Si detectors, with 1x1x1 mm 3<br />

pixels, in coincidence with an existing partial-ring PET<br />

scanner with two opposing groups of 12 block detectors.<br />

The scanner diameter is 1 m and each partial ring<br />

spans 67.5 o out of 180 o so that it must be rotated to three<br />

different positions to cover the full FOV. Each detector<br />

block consists of an array of 32 BGO crystals of 6x2x30<br />

mm 3 . Initial simulations were run with points sources<br />

of 0.1 and 1.5 mCi in the centre of the FOV and the<br />

probe at a 50 mm. A temporal resolution of 5 ns was<br />

applied. The coincidences were classified as ring-ring<br />

and ring-probe; for each type, the contribution of accidental<br />

coincidences (randoms) was estimated.<br />

Results: The spectra measured with the Si detector<br />

were successfully reproduced by the GEANT4 code.<br />

For the full probe-and-ring system, coincidences<br />

where constructed using a 10 ns time window. The<br />

simulations permitted to quantify the kinds of coincidences<br />

as well as to evaluate the sensitivity and random<br />

fractions for each type (see table I).<br />

imaging life<br />

random fraction (%)<br />

Sensitivity (%)<br />

ring-ring ring-probe<br />

0.1 mCi 0.013 1.3<br />

1.5 mCi 0.2 15.5<br />

0.1 mCi 9.7 0.42<br />

1.5 mCi 9.0 0.39<br />

Table1: Random Fractions and Sensitivities<br />

Simulations are now being carried out with more<br />

complex activity distributions. An iterative reconstruction<br />

algorithm, based on ML-EM, developed<br />

for previous two-dimensional simulations of the<br />

MADEIRA system is now being modified to reconstruct<br />

the new three-dimensional data.<br />

Conclusions: The goal of a high resolution Si detector<br />

probe in coincidence with a conventional PET<br />

scanner is improving its spatial resolution and sensitivity.<br />

MC simulations of the proposed system<br />

have been performed and basic processes quantified.<br />

Other types of events such as those with various<br />

detections in the probe are now being studied and<br />

could potentially be exploited to increase the system<br />

sensitivity. The GATE toolkit will be shown to be<br />

useful in simulating such a non conventional scanner<br />

geometry. The coincidence data is now being applied<br />

to reconstruction.<br />

Acknowledgement: Supported by TEC2007-61047<br />

References:<br />

1. Janecek M et al; IEEE Trans Nucl Sci. 53 No.3: 1143-1149<br />

(2006)<br />

2. Zhou J et al; Phys Med Biol. 54:5193-5208 (2008)<br />

3. Wu H et al; J Nucl Med. 49:1668-1676 (2008)<br />

4. Park SJ et al; Phys Med Biol. 52 4653-4677 (2007)


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

Autofluorescence corrected multispectral red-shifted fluorescent protein tomography<br />

Deliolanis N. (1) , Wurdinger T. (2) , Tannous B. (2) , Ntziachristos V. (1) .<br />

(1) Helmholtz Zentrum Muenchen, Neuherberg, Germany<br />

(2) Massachusetts General Hospital, United States<br />

ndeliolanis@yahoo.com<br />

Introduction: The development of fluorescent proteins<br />

(FPs) that operate in the far-red and nearinfrared<br />

part of the spectrum, enable the macroscopic<br />

visualization of FP activity deep in tissues.<br />

We demonstrate herein a multispectral fluorescence<br />

tomography method that allowed the visualization<br />

of labeled glioma tumors in animal brains operating<br />

with two-orders of magnitude better sensitivity<br />

compared to imaging GFP. We discuss how the<br />

detection sensitivity can be improved by at least an<br />

order of magnitude using further shifted FP’s and<br />

the potential of the method for accelerating discovery<br />

associated with functional genomics, stem cell<br />

research and systems biology.<br />

Methods: The method makes use of a novel spectral<br />

inversion scheme that integrates three-dimensional<br />

image reconstruction and auto-fluorescence<br />

correction that works seamlessly in the steep absorption<br />

transition from visible to near-infrared.<br />

The method is based on non-contact full angular<br />

projection Fluorescence Molecular Tomography.<br />

Results: We have successfully imaged mCherry labeled<br />

glioma tumors located deep in tissue in animal<br />

brains. The results show almost perfect anatomical<br />

localization of the tumors that is verified<br />

by MRI and histology.<br />

Conclusions: The approach offers therefore the ability<br />

for tomographically visualizing the emerging<br />

new class of red-shifted fluorescent proteins though<br />

entire animals. We discuss how the detection sensitivity<br />

can be improved by at least an order of magnitude<br />

using further shifted FP’s and the potential<br />

of the method for accelerating discovery associated<br />

with functional genomics, stem cell research and<br />

systems biology.<br />

Acknowledgement: This research is supported by a<br />

Marie Curie Intra-European Fellowship within the<br />

7th European Community Framework Programme.<br />

References:<br />

1. Shaner, N. C. et al. “Improved monomeric red, orange<br />

and yellow fluorescent proteins derived from<br />

Discosoma sp red fluorescent protein”. Nat. Biotechnol.<br />

22, 1567-1572, 2004.<br />

2. N. Deliolanis et al. “Free-space fluorescence molecular<br />

tomography utilizing 360° geometry projections”, Opt.<br />

Lett. 32 382-384 (2007)<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

P-072<br />

poStEr<br />

TECHNOLOGY


P-073<br />

186<br />

WarSaW, poland May 26 – 29, 2010<br />

Acquiring the sample surface from co-registered FMT/MR measurements of a murine tumor<br />

model<br />

Dikaiou K. (1) , Stuker F. (1) , Vats D. (1) , Ratering D. (1) , Keist R. (1) , Klohs J. (1) , Ripoll J. (2) , Rudin M. (1) .<br />

(1) Institute for Biomedical Engineering, Zurich, Switzerland<br />

(2) Institute of Electronic Structure and Laser, Heraclion, Greece<br />

dikaiou@biomed.ee.ethz.ch<br />

Introduction: The combination of MRI and FMT<br />

is promising in preclinical imaging, combining the<br />

high spatial resolution and soft tissue contrast of<br />

MRI for deriving structural and physiological information<br />

with high sensitivity of fluorescence imaging<br />

for studying molecular targets/interactions. Moreover,<br />

MR information can be used as a prior in the<br />

FMT reconstruction. The necessary steps to this end<br />

are the co-registration of the two datasets, the acquisition<br />

and use of the sample surface for reconstructing<br />

fluorescence images, and the characterization of<br />

different tissue types within the sample. The first two<br />

steps are presented here for an in vivo measurement.<br />

Methods: We measured nude mice bearing<br />

subcutaneous tumors on the thigh flank. 106<br />

colon cancer-derived C51 cells had been injected<br />

9 days prior to measurement. The protease-activatable<br />

probe ProSense 680 (VisEn<br />

Medical, Bedford, USA) was administered<br />

via the tail vein in two doses of 13nmol each,<br />

48h and 24h prior to measurement.<br />

We used a custom-made FMT/MR compatible<br />

animal support equipped with an<br />

MR transceiver surface coil. Each animal<br />

was fixed under anesthesia on the stage and<br />

measured on FMT. The optical signal from a<br />

1.5x1.2cm2 ROI around the tumor was collected<br />

upon excitation with a 671nm cw laser<br />

at 680nm and 720 nm. Thereafter, the stage<br />

was inserted into a Biospec 94/30 MR scanner<br />

(Bruker BioSpin MR, Ettlingen, Germany) operating<br />

at 9.4 T. 14 transversal slices with a FOV of 3.8x1.5cm2<br />

and 1.0mm thickness covering the tumor were acquired<br />

with a FLASH sequence (TE/TR = 5/250ms)<br />

for high sample/background contrast. The elapsed<br />

time between FMT/MR measurements was 30min.<br />

The MR data was automatically segmented according<br />

to [1]. Iso-surfaces were determined in<br />

order to compute the top surface height map,<br />

which was interpolated to match the optical image<br />

pixel dimensions. As both the height map and<br />

the optical white light image are oriented along<br />

yz on the coordinate system, they were used for<br />

co-registration. Reference points on the tumor<br />

imaging life<br />

outline were interactively selected to compute<br />

an affine transformation between the two images.<br />

The co-registered surface was subsequently used<br />

in FMT reconstruction.<br />

Results: The co-registered FMT/MR dataset for one<br />

mouse is shown, zoomed on the ROI for clarity. The<br />

reconstructed fluorescence is plotted on the whitelight<br />

image. The top surface is shown in green.<br />

Conclusions: A framework for co-registering FMT/<br />

MR data and recording the sample surface was presented.<br />

It constitutes an essential step towards full<br />

FMT/MR data integration and future use of MR a<br />

priori information in the FMT reconstruction.<br />

Acknowledgement: This work was supported financially<br />

by the EU FP7 FMT/XCT project.<br />

References:<br />

1. N.Otsu, A Threshold Selection Method from Gray-Level<br />

Histograms, Automatica, 1975


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

fDOT/ PET/CT imaging of biological processes in tumors<br />

Garofalakis A. , Dubois A. , Kuhnast B. , Dupont D. , Jassens I. , Mackiewicz N. , Dollé F. , Tavitian B. , Ducongé F. .<br />

CEA, Institut d’Imagerie BioMédicale, Service Hospitalier Frédéric Joliot, Laboratoire d’Imagerie Moléculaire Expérimentale, Orsay, France, Metropolitan<br />

anikitos.garofalakis@cea.fr<br />

Introduction: Small animal Fluorescence Diffuse<br />

Optical Tomography (fDOT) is a relatively new<br />

technology that can provide quantitative imaging<br />

of molecular processes. It allows using smart activatable<br />

probes which can measure biological processes<br />

that are inaccessible using nuclear probes. For<br />

instance, fluorescent sensors have been designed<br />

to monitor specific proteolytic activities using<br />

quenched NIRF probes that become fluorescent<br />

when they are cleaved by a protease of interest. In<br />

this work, we performed experiments aiming in integrating<br />

fDOT imaging of fluorescent probes with<br />

PET-FDG and a X-ray micro-CT imaging.<br />

Methods: For performing combined fDOT/PET/CT<br />

measurements we have developed a simple approach<br />

that incorporates a multimodal mouse supporting<br />

system that can fit in all three modalities. We performed<br />

multimodal imaging of two processes in a<br />

tumour using the fluorine-18- FluoroDeoxyGlycose<br />

([ 18 F]-FDG) and a fluorescent activatable probe. For<br />

optical monitoring we used probes for monitoring<br />

cathepsin activity and the localization of integrin<br />

alpha-v-beta-3 in tumor models by using different<br />

commercially available probes (Prosense680, Integrisense680,<br />

Visen Medical, USA and AngioStamp,<br />

Fluoptics, France). We used as models nude mice<br />

bearing a subcutaneous xenograft of two different<br />

cell lines, the MDA-MB-231 and the NIH-MEN2A.<br />

For the multimodal measurements we followed a<br />

protocol that allows for the optimal correlation of<br />

the fDOT and the PET data. Each mouse has been<br />

firstly measured by PET and was sequentially placed<br />

on the fDOT system for optical imaging. Once the<br />

mouse was placed in the PET, an intravenous injection<br />

of FDG with an activity of 200 μCi was performed<br />

and the dynamic PET scan was initiated.<br />

Optical measurements were performed 24h after<br />

the probe injection.<br />

Results: We reconstructed the 3D cathepsin activity<br />

and the localization of Integrin with respect to the<br />

tumor volume as given by PET. In the all xenograft<br />

models, we observed that the glucose consumption<br />

and the fluorescent probes were just partially<br />

co-localized in the tumour. Indeed, the [ 18 F]-FDG<br />

labelled all the tumour xenograft whereas the<br />

fluorescence signal was predominantly located at<br />

the base and only partially overlapped with the tumor<br />

volume as imaged by μPET.<br />

Conclusions: In this study we explored the potential<br />

of integrating information collected by nuclear and<br />

optical imaging taking advantage of the unique possibilities<br />

that each modality can provide. We found<br />

that the activatable probes used are concentrated<br />

underneath the tumor predominantly connected to<br />

the stroma while the highest glucose consumption<br />

appears to be uniformly distributed in the tumour<br />

tissue. Cathepsin activity is regulated by complex<br />

interactions between extracellular matrix components<br />

and cells populating the tumor like stromal<br />

cells and therefore it is expected to be reconstructed<br />

underneath the FDG signal. We proved that cancer<br />

imaging can benefit from the development of<br />

dual PET/Optical methods can act synergistically<br />

and that can provide complementary information<br />

enhancing thus the information collected from a<br />

single subject.<br />

Acknowledgement: This work was supported by<br />

grants from the European Molecular Imaging Laboratory<br />

(EMIL) network (EU contract LSH-2004-<br />

503569 and the european program FMT-XCT “Hybrid<br />

Fluorescence Molecular Tomography (FMT)<br />

– X-ray Computed Tomography (XCT) method and<br />

system” contract No 201792.<br />

References:<br />

1. Ntziachristos, V., Ripoll, J., Wang, L.V. & Weissleder, R.<br />

Looking and listening to light: the evolution of wholebody<br />

photonic imaging. Nat. Biotechnol 23, 313-320 (2005).<br />

2. Weissleder, R., Tung, C.H., Mahmood, U. & Bogdanov, A.<br />

In vivo imaging of tumors with protease-activated nearinfrared<br />

fluorescent probes. Nat. Biotechnol 17, 375-378<br />

(1999).<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

P-074<br />

poStEr<br />

TECHNOLOGY


P-075<br />

188<br />

WarSaW, poland May 26 – 29, 2010<br />

Deep tissue molecular imaging with fluorescent biomarkers using multispectral optoacoustic<br />

tomography. A simulation study<br />

Glatz J. , Deliolanis N. , Schulz R. , Razansky D. , Ntziachristos V. .<br />

Helmholtz Zentrum Muenchen, Neuherberg, Germany<br />

ndeliolanis@yahoo.com<br />

Introduction: Fluorescent protein<br />

markers have established themselves<br />

as a valuable tool in biomedical<br />

research. Their applicability in<br />

multispectral optoacoustic tomography<br />

(MSOT) has recently been<br />

demonstrated with fluorochromes<br />

and fluorescent proteins [1,2].<br />

Methods: A simulation study was<br />

conducted for the proteins GFP, mRaspberry, IFP<br />

and AF750. Theoretical calculations, based on light<br />

and sound propagation models suggests that IFP and<br />

AF750 yield an acoustic signal that is acoustic signal<br />

that is three orders of magnitude higher than that of<br />

GFP. Two protein inclusions of a concentration of<br />

1 μM were simulated in a cervical mouse structure<br />

and reconstructed by the backprojection algorithm.<br />

Results: It was shown that the protein concentration<br />

can be unmixed using only three multispectral<br />

measurements. Using blind source separation<br />

techniques this could even be achieved without<br />

prior spectral information. The unmixed components<br />

of the different proteins are shown in Figure<br />

1. The distinctively weaker signal strength from<br />

GFP and mRaspberry is a consequence of the<br />

strong tissue absorption in their spectral region.<br />

In order to obtain a stronger signal from the center<br />

the reconstructions were normalized for the light<br />

fluence inside the sample. From the simulation study<br />

the optimal measurement wavelengths could be determined<br />

for each protein. Their usage, as well as the<br />

blind source separation, was verified in a practical<br />

experiment using a tissue mimicking phantom with<br />

fluorochrome inclusions.<br />

Conclusions: The new red-shifted proteins can boost<br />

the acoustic signal intensity by over three order of<br />

magnitude. Protein concentrations can be unmixed<br />

in deep tissue and correcting for the light attenuation<br />

is an important step towards quantitative unmixing.<br />

Acknowledgements: This research is supported by a<br />

Marie Curie Intra-European Fellowship within the<br />

7th European Community Framework Programme.<br />

imaging life<br />

Fig. 1: Unmixing of tissue and protein distribution for simulated data<br />

References:<br />

1. D. Razansky et al. Multispectral photoacoustic imaging<br />

of fluorochromes in small animals Opt. Letters, 32,<br />

2891-2893, 2007<br />

2. D. Razansky et al. Multispectral opto-acoustic<br />

tomography of deep-seated fluorescent proteins in<br />

vivo Nat. Photonics, 3, 412-417, 2009


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

Microfluidic [ 11 C]-carbonylation reactions for the rapid synthesis of radiolabelled compounds<br />

for PET<br />

Miller P. (1) , Audrain H. (2) , Bender D. (2) , Demello A. (1) , Gee A. (3) , Long N. (1) , Vilar R. (1) .<br />

(1) Imperial College London, London, United Kingdom<br />

(2) Arhus University Hospital PET Centre, Denmark<br />

(3) GSK, United Kingdom<br />

philip.miller@imperial.ac.uk<br />

Introduction: The synthesis of 11C compounds for<br />

PET requires fast and specialised chemical techniques<br />

owing to the short half-life of the 11C radioisotope<br />

(t1/2 = 20.4 min) and sub-micromolar reaction scales.<br />

[1] Microfluidic reactors are emerging as a valuable<br />

technology for the rapid and small scale synthesis of<br />

short-lived radiopharmaceuticals for PET imaging.<br />

[2] The palladium mediated 11C-carbonylation reaction<br />

is a highly versatile route for the preparation<br />

of a wide range of 11C-carbonyl compounds[3],<br />

however, the low molecular concentrations of<br />

11CO coupled with its poor solubility in organic<br />

solvents make this a particularly challenging<br />

transformation. Here we report the application of<br />

a microfluidic reactor for improving the synthesis<br />

of 11C labelled amide and ester molecules via the<br />

palladium mediated 11C-carbonylation reaction.<br />

Methods: The microfluidic reactor (figure 1) was<br />

fabricated from glass using chemical wet etching<br />

techniques and contains two inlets, one outlet and a<br />

5 metre long reaction channel. A simple mixing tee<br />

motif is used bring the gas and liquid reagents into<br />

contact with each other. The palladium mediated<br />

11C-carbonylation reaction of a range of aryl halides<br />

was investigated (scheme 1). In a typical 11CO<br />

labelling experiment the coupling reagents (aryl halide,<br />

palladium catalyst and amine) were premixed<br />

and loaded into a 50 uL loop on an injector port.<br />

11CO, produced via the high temperature reduction<br />

of 11CO2 over Mo, was preconcentrated and trapped<br />

onto molecular sieves. The coupling reagents were<br />

injected into the microfluidic device while at the<br />

same time 11CO was released from the molecular<br />

sieves and passed into the device for reaction.<br />

Results: The total reaction and processing times for<br />

a typical reaction, including trapping and release of<br />

X<br />

+<br />

H 2 N<br />

R<br />

X = I or Br;<br />

R = CN, CF3 or OCH3 Scheme 1<br />

11 CO, Pd catalyst<br />

microfluidic chip<br />

R<br />

O<br />

* N<br />

H<br />

11CO, was 15 min from end of bombardment. A series<br />

of amide and ester molecules were labelled with<br />

11C using our microfluidic reaction system (scheme<br />

1). Radiochemical yields (RCY) of labelled products<br />

were found to be dependent on the type of aryl halide<br />

and nucleophile used for the reaction. Generally,<br />

iodoaryl substrates with activating groups (CF3<br />

or CN) gave consistently higher RCYs (>80%) and<br />

radiochemical purities (>95%) than aryl halide substrates<br />

with deactivating groups (OCH3).<br />

Conclusions: A glass fabricated microfluidic device<br />

has been used to effectively perform high speed 11CO<br />

radiolabelling reactions. The larger surface area-tovolume<br />

ratio within the microfluidic reactor improves<br />

the gas liquid contact area and is thought to enhance<br />

the problematic CO insertion step of this reaction.<br />

Acknowledgement: PWM is grateful to the EPSRC for the<br />

award of a Life Sciences Interface fellowship (EP/E039278/1).<br />

References:<br />

Figure 1<br />

1. P. W. Miller et al., Angew. Chem. Int. Ed., 2008, 47,<br />

8998.<br />

2. P. W. Miller, J. Chem. Technol. Biotechnol. 2009, 84, 309.<br />

3. B. Langstrom et al., J. Labelled Compd. Radiopharm.,<br />

2007, 50, 794.<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

P-076<br />

poStEr<br />

TECHNOLOGY


WarSaW, poland May 26 – 29, 2010<br />

P-077 Boosting image quality in low-dose RC-gated 5D cone-beam micro-CT<br />

190<br />

Sawall S. (1) , Bergner F. (1) , Lapp R. (2) , Mronz M. (2) , Karolczak M. (1) , Kachelrieß M. (1) .<br />

(1) Institute of Medical Physics (IMP), Erlangen, Germany<br />

(2) CT Imaging GmbH, Erlangen, Germany<br />

stefan.sawall@imp.uni-erlangen.de<br />

Introduction: Micro-CT imaging of the animal<br />

heart typically requires respiratory and cardiac<br />

(RC) gating. This can either be done prospectively<br />

or retrospectively. For functional imaging, and<br />

for multi-modality imaging, it is often desired to<br />

obtain the full 5D information (volumetric + respiratory<br />

+ cardiac) and retrospective gating is<br />

the method of choice. The amount of information<br />

available to reconstruct one volume for a given respiratory<br />

and cardiac phase is significantly lower<br />

than the total amount of information acquired.<br />

For example the reconstruction of a volume from a<br />

10% wide respiratory and a 20% wide cardiac window<br />

uses only 2% of the data acquired. Achieving<br />

a similar image quality as a non-gated scan would<br />

typically require to increase the dose by a factor of<br />

up to 50. Our aim is to provide similarly high image<br />

quality at low dose levels (100 to 500 mGy).<br />

Methods: We implemented a two-step iterative image<br />

reconstruction algorithm based on the McKinnon-Bates<br />

approach. The first step, aiming at R<br />

gating only, uses a prior image consisting of the<br />

reconstruction of all data. The RC gating step uses<br />

the image of the first step as a prior. An edge-preserving<br />

anisotropic diffusion filter is applied to the<br />

volume data after each iteration to perform spatial<br />

and temporal resolution-preserving noise reduction<br />

in up to five dimensions. We demonstrate our<br />

new reconstruction approach using mouse data<br />

scanned with a dedicated in-vivo micro-CT scanner<br />

(TomoScope Synergy, CT Imaging GmbH, Erlangen,<br />

Germany). We derived an intrinsic gating<br />

signal (kymogram) from the rawdata to synchronize<br />

our reconstruction. The scan itself consisted<br />

of ten rotations with 7200 projections in total.<br />

imaging life<br />

The scan time was five minutes, the tube voltage 65<br />

kV. The gating window widths were set to 10% in the<br />

respiratory and to 20% in the cardiac cycle.<br />

Results: Although the mouse data available to us<br />

shows only an untypically low enhancement of about<br />

110 HU (blood vs. myocardium) our approach yields<br />

an image noise of as low as 35 HU while preserving the<br />

spatial resolution of about 100 µm. We also performed<br />

a standard RC-gated reconstruction. Its high noise<br />

levels of about 170 HU are prohibitive and significant<br />

artifacts are observed due to sparse view sampling.<br />

The dose of our protocol is about 500 mGy. Reconstructions<br />

using less than the available 10 rotations<br />

show that one can achieve good image quality in<br />

RC-gated micro-CT with about 100 mGy, provided<br />

that the contrast agent delivers sufficient contrast.<br />

Conclusions: Using advanced image reconstruction<br />

techniques enables us to perform high fidelity<br />

low-dose double-gated imaging of free breathing<br />

small animals.<br />

Acknowledgement: This work was supported by<br />

the Deutsche Forschungsgemeinschaft (DFG) under<br />

grant FOR 661.


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

Methodological approach using microscopy for quantitative evaluation of neo-angiogenesis<br />

and tumour progression in pre-clinical cancer models<br />

Thézé B. , Beynel A. , Chapotot L. , Boisgard R. , Tavitian B. .<br />

CEA /Inserm 1023, Orsay, France<br />

benoit.theze@cea.fr<br />

Introduction: In vivo molecular imaging methods can<br />

localize sites of angiogenesis and obtain functional<br />

data, whereas microscopic methods provide their<br />

highest resolution on preserved tissue specimens. To<br />

bridge the gap between molecular imaging methods<br />

and microscopy, we developed a method to evaluate<br />

neo-angiogenesis and tumour volume on formalinfixed<br />

tissues samples.<br />

Methods: In order to visualize the mammary tumour<br />

vessels density in the MMTV-PyMT transgenic model<br />

of mammary carcinoma, mice were injected i.v.<br />

with Horse Radish Peroxidase (HRP)– tomato lectin<br />

(at 6.66 g/kg), which binds surface glycoproteins of<br />

red blood cells and endothelial cells inside the vascular<br />

lumen. Tumours were immediately sampled, fixed<br />

in formalin and frozen. For this pilot study, four tumours<br />

from each of two mice were sectioned every<br />

300 µm . For every section, HRP-lectin was revealed<br />

with 3,3´-diaminodbenzidine (DAB) and the tissues<br />

were counterstained with haematoxylin. Panoramic<br />

views of each tissue section were acquired in brightfield<br />

with a motorized AxioObserver Z1 Zeiss microscope.<br />

Tissue staining and image acquisition parameters<br />

were standardized for quality control. Using<br />

imageJ image analysis software, a macro was developed:<br />

(i) to separate blue and brown colours from the<br />

RGB original image using the colour deconvolution<br />

plugin in batches of images, and (ii) to evaluate the<br />

area occupied by the tumour in the whole sample. A<br />

treatment pipeline was designed with the Cellprofiler<br />

software for threshold and segmentation of vessels<br />

(brown) versus tumour masses (blue). Object-based<br />

filtering was applied to vessels according to their size<br />

and to their localization inside or outside of the tumour<br />

masses in order to separate neo-vessels from<br />

pre-existing vessels and areas of necrosis.<br />

Results: Using this approach, we were able to measure<br />

areas occupied by vessels and tumours and, (i)<br />

to obtain an estimate of neo-angiogenesis defined as<br />

the ratio between tumour blood vessels’ volume and<br />

tumour volume (1.27% ± 0.61%), (ii) to determinate<br />

the real volume of tumour in the sample (75%<br />

± 7%), which also contain muscular, adipose and<br />

conjunctive tissue, (iii) to obtain the ratio between<br />

total blood volume and total sample volume (5.99%<br />

± 0.42%), and (iiii) to discriminate between small<br />

(55% ± 9.6%), medium (32% ± 2.8%), and large<br />

(12.9% ± 7.8%) blood vessels.<br />

Conclusions: The data from this feasibility study<br />

provides morphological information about the vascular<br />

network in the PyMT model. Further validation<br />

steps should now allow correlating the results<br />

with functional information obtained by PET imaging,<br />

and provide a method for the evaluation of antiangiogenic<br />

drugs in animal tumour models.<br />

Fig1 from left to right: original image, blue and brown colour deconvolved images and final segmented image<br />

Acknowledgement: This work was supported by the<br />

6th FW EU grants EMIL (LSHC-CT-2004-503569)<br />

and DiMI (LSHB-CT-2005-512146)<br />

References:<br />

1. Rasband, W.S., ImageJ, U. S. National Institutes of<br />

Health, 1997-2009<br />

2. Jones TR et al. (2008) CellProfiler Analyst: data<br />

exploration and analysis software for complex imagebased<br />

screens. BMC Bioinformatics<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

P-078<br />

poStEr<br />

TECHNOLOGY


192<br />

WarSaW, poland May 26 – 29, 2010<br />

P-079 Changes of heart stroke volume index established by 99m Tc MIBI GSPECT scintigraphy after<br />

radioiodine treatment of patients with subclinical hyperthyroidism<br />

Kaminski G. , Podgajny Z. , Szalus N. , Bilski M. , Dziuk M. .<br />

Military Institute of Health Services, Warsaw, Poland<br />

gkam@wim.mil.pl<br />

Introduction: Stroke volume index (SVI) is an indicator<br />

of heart load. Increased heart load is one of<br />

causes of cardiac death. Subclinical hyperthyroidism<br />

(SH yper ) increases mortality mostly due to cardiovascular<br />

events. SH yper affects about 1% of population including<br />

patients with cardiovascular diseases. The<br />

aim of the investigation was to estimate an influence<br />

of cure with radioiodine of SH yper on heart stroke volume<br />

index (SVI) measured by heart scintigraphy -<br />

99m Tc MIBI GSPECT.<br />

Methods: 44 patients (37 women, 7 men) aged<br />

45.9±11, with 12.8±9.8 month history of only autonomous<br />

SH yper (TSH=0.16±0.1 IU/l), were examined<br />

with 99m Tc MIBI GSPECT twice: before<br />

and 5.7±4.2 months after TSH normalization<br />

(TSH=1.32±0.1 IU/l) after radioiodine treatment<br />

(at dose 12.1±5.7 mCi) . The radioiodine ( 131 I)<br />

and 99m Tc MIBI were carried by POLATOM/Poland.<br />

The average time between examinations was<br />

12.5 ± 6 months. The Local Ethical Committee approval<br />

for this investigation has been obtained.<br />

Results: The cure of SH yper caused decrease of SVI<br />

from 26.36 to 24.21 ml/m 2 (p=0.042)<br />

Conclusions: Cure of autonomous subclinical hyperthyroidism<br />

with radioiodine decreases heart<br />

load. This finding indicates to treat subclinical hyperthyroidism,<br />

especially in patients with cardiovascular<br />

diseases.<br />

imaging life<br />

[ml/m 2 ]<br />

27<br />

26<br />

26<br />

25<br />

25<br />

24<br />

24<br />

23<br />

26,36<br />

przed po<br />

24,21<br />

Fig.1. Mean values of SVI before (left) and after (right) radioiodine<br />

treatment p = 0,042)<br />

References:<br />

1. Parle J.V., Maissoneuve P., Sheppard M.C., Boyle P.,<br />

Franklyn J.A.: Prediction of all- cause and cardiovascular<br />

mortality in elderly people from one low serum<br />

thyrotropin result: a 10 -year cohort study.<br />

2. Lancet, 2001, 358: 861 - 865.


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

PET/CT investigations with 68Ga-DOATATE in neuroendocrine tumors - first clinical<br />

experience<br />

Kunikowska J. (1) , Krolicki L. (2) , Pawlak D. (3) , Kobylecka M. (2) .<br />

(1) Medical University of Warsaw, Poland<br />

(2) Medical University of Warsaw, Poland<br />

(3) IEA POLATOM, Świerk, Poland<br />

jolanta.kunikowska@wum.edu.pl<br />

Introduction: Neuroendocrine tumors (NETs) have<br />

distinct biological and clinical characteristics, in<br />

particular a high density of somatostatin receptors<br />

at the cell membrane. It is this property that allows<br />

the use of radiolabeled somatostatin analogs for imaging<br />

of these tumors. Novel techniques PET/CT<br />

with 68Ga-DOTATATE open new possibilities in the<br />

diagnosis of patients with NET. Sensitivity of that<br />

techniques is depending on SSTR expression, but average<br />

range is 60-94%.<br />

The aim of this study was to evaluate the diagnostic<br />

usefulness of a new somatostatin analog, 68Ga-<br />

DOTATATE, for PET/CT in patients with diagnosis<br />

of neuroendocrine tumors.<br />

Methods: 55 patients with NET were examined (24<br />

men, 31 women; age range, 18-86 y; mean age +/- SD,<br />

51.4 +/- 12.5 y). For analysis, patients were divided into<br />

3 groups: detection of unknown primary tumor (n =<br />

13 patients), follow-up after surgery (n = 23 patients),<br />

staging of disease (n= 19). PET imaging was performed<br />

on PET/CT scanner Biograph 64, 60 minutes<br />

post injection of 120-185 MBq 68Ga-DOTATATE.<br />

Results: In the group patients with unknown primary<br />

tumors, 68Ga-DOTATATE revealed 9 primary<br />

foci. 7 were not visible in CT (4 in intestinum, 3 in<br />

pancreas) and 2 foci observed in CT and PET/CT<br />

- small nodule in lung. In 4 cases primary tumors<br />

were not found.<br />

In the cases of patients after surgery, examination<br />

shown new foci in 5 patients (liver-3, lymph nodes<br />

in abdominal cavity-2, peritoneum-1, pancreas-1).<br />

In the group of staging of disease in 12/19 cases<br />

PET and CT shown the same foci. In 7/19 (37 %)<br />

patients in 68Ga-DOTATATE examination was revealed<br />

new lesions (intestinum-1, liver-3, bone-1,<br />

pancreas-4, lymph nodes in abdominal cavity-2)<br />

Conclusion: Results of our study shown that 68Ga-<br />

DOTATATE PET/CT is very useful non-invasive<br />

techniques in diagnosis of patients with NET.<br />

68Ga-DOTATATE PET/CT examination should<br />

be performed for staging and restaging of disease<br />

and it gives more clinically useful information<br />

than CT.<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

P-080<br />

poStEr<br />

ENDOCRINE DISEASES


194<br />

WarSaW, poland May 26 – 29, 2010<br />

P-081 Influence of number of subsets and iterations of Ordered Subsets Expectation Maximization<br />

(OSEM 3D Flash) reconstruction on quantitative assessment of small and medium detected<br />

lesions in SPECT study with used in 99m Tc[EDDA/HYNIC]Octreotate for patients with GEP-NET<br />

Lenda-Tracz W. .<br />

Nuclear Medicin Unit, Krakow, Poland<br />

wtracz@su.krakow.pl<br />

Introduction: The algorithm OSEM 3D Flash generally<br />

is the most appropriate available method of<br />

reconstruction. The choice of appropriate sets of<br />

OSEM 3D Flash reconstruction is crucial in interpretation<br />

of scintigraphy images. The number<br />

of subsets and iterations significantly influences<br />

the lesion to noise ratio. Therefore the aim of the<br />

study was to find the optimum number of subsets<br />

and iterations which present the most effective<br />

lesion to noise ratio in quantitative assessment<br />

of small and medium lesions detected with<br />

the use of 99m Tc[EDDA/HYNIC]Octreotate.<br />

Methods: The results of 20 patients with confirmed<br />

neuroendocrine tumors (GEP-NET) were<br />

analyzed. SPECT studies, acquisition 3-4 h after<br />

injection of 740 MBq 99m Tc[EDDA/HYNIC]<br />

Octreotate, were performed. The E.CAM 180<br />

(Siemens), double-head gamma-camera, was<br />

equipped with parallel, low-energy, high-resolution<br />

collimators. Data were acquired with 180<br />

rotation with 128 non-circular projections (30s<br />

per view) using a 128x128 matrix with 1.23 zoom.<br />

OSEM 3D Flash reconstruction (subsets number<br />

8, 16, 32 for 2-30 iterations) was performed.<br />

Results: Lesion to noise ratio in voxels was analyzed<br />

for different setting combinations (n,m)<br />

of number of subsets (n) and iterations (m) of<br />

OSEM 3D Flash reconstruction. An increasing<br />

number of subsets and iterations influences the<br />

increasing values of lesion to noise ratio. No difference<br />

for settings (16,m) and (32,m) was observed<br />

for quantitative assessment of small and<br />

medium lesions detected. Differences between lesion<br />

to noise ratio were observed in a group with<br />

8 subsets (8,m). However, the quality of images is<br />

variable for all setting combinations (n,m).<br />

Conclusions: The high number of subsets improves<br />

the image quality and the images are<br />

smoother. The increasing number of iterations<br />

on the one hand gives a little better contrast but<br />

on the other hand the shape of the lesions and organs<br />

is sharper. In spite of the fact that the image<br />

quality is changed with the increasing number of<br />

subsets and iterations, for quantitative assessment<br />

imaging life<br />

only (8,m) setting is changed significantly. Therefore,<br />

for quantitative assessment the best choice<br />

is setting with 8 subsets and 2-30 iterations but at<br />

least 6 and no more than 22 iterations with step<br />

equal at least 4 iterations. The appropriate number<br />

of iterations depends on the image quality<br />

assessment.


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

The precise localization of metastatic lesion with used SPECT/CT with CoDe system after<br />

131I – MIBG therapy in patients with disseminated pheochromocytoma<br />

Szalus N. (1) , Podgajny Z. (2) , Kaminski G. (3) , Mazurek A. (1) , Giżewska A. (1) , Dziuk E. (1) .<br />

(1) Department of Nuclear Medicine,Military Institute of Medicine, Warsaw, Poland<br />

(2) Department of Endocrinology and Radioisotope Therapy2 - Military Institute of Medicine, Warsaw, Poland<br />

(3) Department of Endocrinology and Radioisotope Therapy - Military Institute of Medicine, Warsaw, Poland<br />

nszalus@wp.pl<br />

Introduction: Pheochromocytoma is a rare tumor<br />

that originates from chromaffin cells such as the adrenal<br />

medulla and sympathetic ganglia. Malignant<br />

pheochromocytoma is uncommon, and metastases<br />

typically affect the bones, liver, lungs, and lymph<br />

nodes. Meta-iodo-benzyl guanidine (MIBG) is a<br />

norepinephrine analog, and 131I- and 123I-MIBG<br />

have been widely used for the diagnosis of pheochromocytoma.<br />

This technique has high specificity and<br />

detectability not only for primary tumors but also<br />

metastatic lesions when compared with morphologic<br />

imaging such as computed tomography (CT) and<br />

magnetic resonance imaging (MRI). Co-registered<br />

data have been shown to be useful in the evaluation<br />

of patients with cancer at diagnosis and staging, in<br />

monitoring the response to treatment, and during<br />

follow up, for early detection of recurrence. Gamma<br />

camera with CoDe system is a new modality to the<br />

PET/CT and SPECT/CT imaging to the precise localization<br />

metastatic lesions.<br />

Aim of study: The aim of this study is to investigate<br />

the precise localization 131-MIBG with used SPECT/<br />

CT CoDe system for metastatic diseases in patients<br />

with malignant pheochromocytoma.<br />

Material and methods: Two patients with disseminated<br />

pheochromocytoma (the first patient with metastases<br />

to the liver, bones and lung; the second with<br />

metastases to the bone) were referred for study. Routine<br />

whole body scan with I-131 was performed with<br />

a dual head gamma camera (Infinia Hawkeye General<br />

Electric Milwaukee with CoDe system, USA) using<br />

a large field of view with high energy collimator<br />

a 20 % energy window centered at 364 keV and, 1024<br />

x 512 matrix. The data acquisition was performed 72<br />

hours after 131-MIBG therapy. Whole body anterior<br />

and posterior views were obtained. SPECT images of<br />

thorax and abdomen were obtained with 45 sec/frame,<br />

60 projections, 20 % window centered at 364 keV,<br />

matrix size of 128 x 128 and zoom factor of 1.0. This<br />

was followed by CT acquisition using single slice (1<br />

cm thickness). Using volumetrix software in Xeleris,<br />

fused images in coronal, sagittal and transaxial views<br />

were then obtained. Whole body planar images were<br />

first interpreted alone. Then, they were reassessed<br />

with the addition of SPECT/CT coregistered images.<br />

Results: 1. In these patients SPECT/CT revealed<br />

30 % more pathological lesions than planar studies<br />

alone. 2. SPECT/CT provided precise anatomical<br />

localization not clearly evident in planar images<br />

alone. 3. It also enabled exclusion of disease in sites<br />

of physiologic tracer deposition found suspicious in<br />

planar studies alone<br />

Conclusion: SPECT/CT allows more precise localization<br />

and interpretation of 131I-MIBG whole<br />

body scan thereby improving its diagnostic accuracy.<br />

It also has impact on correct restaging after therapy<br />

with 131I-MIBG preparation.<br />

References:<br />

1. Bas Havekes at al. Clinical Endocrinology (2010) 72,<br />

137–145<br />

2. Akie Takano at al. Ann Nucl Med (2008) 22:395–401<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

P-082<br />

poStEr<br />

ENDOCRINE DISEASES


196<br />

WarSaW, poland May 26 – 29, 2010<br />

P-083 Multimodal in vivo imaging of pancreatic beta-cells via antibody mediated targeting of<br />

beta-cell tumors<br />

Vats D. (1) , Dikaiou K. (1) , Stuker F. (1) , Honer M. (2) , Wang H. (3) , Schibli R. (2) , Rudin M. (1) .<br />

(1) Institute for Biomedical Engineering, ETH Zurich, Zurich, Switzerland<br />

(2) Institute of Pharmaceutical Sciences, ETH Zurich, Switzerland<br />

(3) F.Hoffman La-Roche, Basel, Switzerland<br />

vats@biomed.ee.ethz.ch<br />

Introduction: Pancreatic beta-cells regulate glucose<br />

metabolism by producing and secreting insulin in<br />

sufficient amounts. Progressive loss of beta-cell mass<br />

and function surmounts to acute diabetes. Diagnosis<br />

of diabetes and the evaluation of potential therapeutics<br />

suffer from lack of established methods for in<br />

vivo imaging of beta-cells. This study demonstrated<br />

the in vivo targeting of a novel antibody to a beta-cell<br />

surface protein in beta-cell tumor models. The pilot<br />

study evaluated the binding capacity of 89 Zr-lableled<br />

and alexa680 labeled monoclonal antibody against<br />

hTMEM27, a human beta-cell surface glycoprotein<br />

(1), for targeted imaging of beta-cell tumors in nude<br />

mice using positron emission tomography (PET) and<br />

fluorescence imaging.<br />

Methods: Insulinoma cell-line (beta-cells), Ins1E,<br />

was taken as the parental cell-line for generating<br />

stable cell lines (2) over-expressing hTMEM27 (Ins-<br />

TM) while blank Ins1E cells (Ins-only) were taken<br />

as controls. Ins-TM and Ins-only cells were used to<br />

generate subcutaneous tumors in nude mice. 1.5MBq<br />

of 89 Zr-anti-hTMEM27 monoclonal antibody ( 89 Zr-<br />

in vivo fluorescence imaging<br />

Ins-TM Ins-only Ins-TM-HA<br />

ROI ROI ROI<br />

alexa680-anti-hTMEM-mAb alexa680-anti-HA-mAb<br />

TM-mAb) and 2mg/kg alexa680-anti-hTMEM27<br />

monoclonal antibody (alexa680-TM-mAb) and<br />

alexa680-anti-HA control monoclonal antibody (alexa680-HA-mAb)<br />

were injected i.v. in tumor bearing<br />

nude mice. The animals were in vivo imaged three<br />

days after antibody administration: PET for 89 Z-TMmAb<br />

and fluorescence imaging (both near infrared<br />

fluorescence reflectance imaging and near infrared<br />

fluorescence molecular tomography (NIR-FMT)) for<br />

alexa680-TM-mAb and alexa680-HA-mAb. The antibody<br />

retention was quantified using gamma counting<br />

of excised tumors for the 89 Zr labeled antibody and<br />

the fluorescence intensity was derived from region<br />

of interest (ROI) quantification for reflectance mode<br />

and NIR-FMT data (using MATLAB tools). The<br />

data was compared between the hTMEM27 over expressing<br />

insulinomas (Ins-TM), control insulinomas<br />

imaging life<br />

(Ins-only) and control antibody in hTMEM27 over<br />

expressing tumors (TM-HA), for antibody specificity<br />

over a period of 6 days. Individual tumors from<br />

optical study were further subjected to histological<br />

analysis for accurate localization of the dye distribution<br />

within tumor tissue.<br />

Results: We found 6-7 times higher retention of the<br />

antibody in TMEM27 over expressing beta-cells,<br />

when compared to control beta-cells or with control<br />

antibody by both 89 Zr-PET and NIR-imaging (Figure<br />

1). The fluorescence microscopy results confirmed<br />

antibody retention to TMEM27 in target insulinomas<br />

(Ins-hTMEM-tumors).<br />

Conclusions: The pilot study could demonstrate<br />

the dual modal imaging of beta-cell mass, in a betacell<br />

tumor model, and identified a novel antibody<br />

for image-guided targeting of a beta-cell surface<br />

glycoprotein.<br />

Acknowledgement: This work is funded by<br />

F.Hoffmann La-Roche Ltd.<br />

Figure 1.<br />

Av. Fl. intensitiy<br />

References:<br />

1.40E+05<br />

1.20E+05<br />

1.00E+05<br />

8.00E+04<br />

6.00E+04<br />

4.00E+04<br />

2.00E+04<br />

0.00E+00<br />

Fluorescence quantification<br />

pre 24h 3d 6d<br />

Time after injection<br />

Ins-only<br />

Ins-TM<br />

Ins-TM-HA<br />

1. Akpinar P et al.; Cell Metab. 2(6):385-97 (2005)<br />

2. Wang H and Iyenedjian P B.; PNAS. 94: 4372-4377<br />

(1997)


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

6-[ 18 F]Fluoro-PBR28, a novel TSPO 18 kDa radioligand for imaging neuroinflammation with<br />

PET<br />

Boisgard R. (1) , Damont A. (1) , Blossier A. (1) , Jego B. (1) , Siquier K. (1) , Kassiou M. (2) , Dolle F. (1) , Tavitian B. (1) .<br />

(1) CEA, Orsay, France<br />

(2) Sydney University, Australia<br />

raphael.boisgard@cea.fr<br />

Introduction: The peripheral benzodiazepine receptor<br />

(PBR or TSPO 18 kDa) is expressed by microglial<br />

cells in many neuropathologies involving<br />

neuroinflammation. [ 11 C]PK11195 is today the<br />

most widely used radioligand for the in vivo imaging<br />

of PBR using PET, and this in spite of its low<br />

brain uptake and its high level of non-specific binding.<br />

Numerous PK11195 challengers are currently<br />

under investigation [1,2], and of particular interest<br />

are the N-benzyl-N-(2-phenoxyaryl)-acetamides, a<br />

series which includes [ 11 C]PBR28 [3]. A fluorinecontaining<br />

analogue, namely 6-fluoro-PBR28 (N-(2methoxybenzyl)-N-(6-fluoro-4-phenoxypyridinyl-<br />

3-yl)acetamide), has been labeled with the longer<br />

half-life positron-emitter fluorine-18 and pharmacologically<br />

evaluated in a rat model of neuroinflammation<br />

(unilaterally, AMPA-induced, striatum-lesioned<br />

rats) with PET.<br />

A<br />

18 F<br />

N<br />

N<br />

O<br />

O<br />

O<br />

B<br />

Fig1: Structure of 6-[18F]fluoro-PBR28 (A) and microPET images (B) obtained in AMPA lesioned rat (60 min p.i.)<br />

Methods: 6-Fluoro-PBR28, as well as the corresponding<br />

precursors for labeling, were synthesized<br />

from 4-chloro-3-nitropyridine. 6-Fluoro-PBR28 was<br />

labeled with fluorine-18 by nucleophilic heteroaromatic<br />

substitution using K[ 18 F]F-Kryptofix ® 222,<br />

purified by HPLC (Waters Symmetry ® C-18) and<br />

formulated for i.v. injection. The AMPA rat model<br />

was used to study in vitro and in vivo specific and<br />

non-specific binding using autoradiography and<br />

µPET imaging on a Concorde Focus P220 PET<br />

camera, including displacement with PK11195<br />

and non-labeled 6-fluoro-PBR28 (1 mg/kg).<br />

Results: Starting from a 37 GBq cyclotron-produced<br />

[ 18 F]fluoride batch, 3.3-3.7 GBq of 6-[ 18 F]<br />

fluoro-PBR28, > 99% radiochemically pure and<br />

ready-to-inject, were obtained within 90 minutes<br />

(Figure 1A). In PET experiments, 6-[ 18 F]fluoro-<br />

PBR28 showed a higher contrast between the lesioned<br />

area and the corresponding area in the intact<br />

contralateral hemisphere (ratio ipsi/contra at<br />

60 min post-injection: 6-[ 18 F]fluoro-PBR28 : 2.2)<br />

(Figure 1B). Furthermore, 6-[ 18 F]fluoro-PBR28<br />

was displaced by PK11195 or 6-fluoro-PBR28. Finally,<br />

modelisation of the PET data using the “simplified-reference-tissue-model”<br />

showed increased<br />

binding potential (BP) in comparison to the BP<br />

of [ 11 C]PK11195 measured in the same model<br />

(2.0±0.6 versus 1.1±0.2). Immunohistochemical<br />

analyses correlate with PET-imaging and showed<br />

strong activation of microglia in and around the<br />

lesion.<br />

Conclusions: Dynamic µPET studies in rats demonstrate<br />

the potential of 6-[ 18 F]fluoro-PBR28 to<br />

image neuroinflammation.<br />

Acknowledgements: Supported in part by the EC -<br />

FP6-project DiMI (LSHB-CT-2005-512146) and<br />

EMIL (LSH-2004-503569).<br />

References:<br />

1. Chauveau F et al; Eur J Nucl Med Mol Imag 35: 2304-<br />

2319 (2008)<br />

2. Dollé F et al; Curr Med Chem 16: 2899-2923 (2009)<br />

3. Briard E et al. J Med Chem 51: 17-30 (2008)<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

P-084<br />

poStEr<br />

INFECTION and INFLAMMATION


WarSaW, poland May 26 – 29, 2010<br />

P-085 Neuroinflammation is increased in the brain of ageing corpulent (JCR:LA-cp) rats: a positron<br />

emission tomography study<br />

198<br />

Boutin H. (1) , Drake C. (1) , Denes A. (1) , Mccoll B. (1) , Prenant C. (1) , Brown G. (1) , Kassiou M. (2) , Herholz K. (2) , Rothwell N. (2) .<br />

(1) University of Manchester, United Kingdom<br />

(2) University of Sydney, Australia<br />

herve.boutin@manchester.ac.uk<br />

Introduction: Despite intense research and development<br />

of several animal models, drugs efficient in<br />

preclinical model of cerebral ischaemia so far have<br />

failed when reaching clinical trial [1] . One striking<br />

feature of the animal models is the lack of co-morbidities<br />

and risk factors when compared to clinical<br />

set-up, in which patients have atherosclerosis, high<br />

blood pressure, chronic and/or acute inflammation<br />

due to chronic inflammatory diseases and infections.<br />

Inflammation and neuroinflammation in particular<br />

are known aggravating factors of stroke outcome [2] .<br />

Here we investigate the impact of known risk factors<br />

of stroke such as obesity and atherosclerosis in<br />

JCR:LA-cp (corpulent) rats on neuroinflammation as<br />

measured by TSPO expression in activated microglia.<br />

Methods: Neuroinflammation was assessed with<br />

[ 18 F]DPA-714 by PET in Lean (control: Cp/?)) and<br />

JCR:LA-cp (corpulent: Cp/Cp) rats PET imaging. 2<br />

groups of rats were scanned at 9 months or 12 and<br />

15 months of age (n=4 per group). PET images were<br />

co-registered with a MRI template [3] for analysis<br />

and automatic segmentation performed for userindependent<br />

ROI determination [4,5] . Euthanasia was<br />

performed at 9 months and 15 months of age 3 to 7<br />

days after PET imaging to assess various neuroinflammation<br />

biomarkers (GFAP, IBA1, VCAM) by<br />

immunohistochemistry (IHC).<br />

Results: Our results show an increase (+38%) in<br />

neuroinflammation in the brain of corpulent rats<br />

when compared to control. In older rats (12 and<br />

15 months old), neuroinflammation was even further<br />

increased but in both lean and corpulent rats.<br />

imaging life<br />

Neuroinflammation, as quantified by PET, was<br />

mainly localised in peri-ventricular and thalamic<br />

areas of the brains. IHC for microglial activation<br />

confirmed the PET data in 15 months old animals<br />

(Figure below).<br />

Conclusions: Our data show here the importance<br />

of including risk and co-morbidity factors in preclinical<br />

models of stroke as they have a significant<br />

impact on neuroinflammation. This study shows<br />

the crucial role of molecular imaging with the<br />

high translational value of PET imaging to investigate<br />

such paradigms. A more detailed study of<br />

the localisation of neuroinflammation as localised<br />

by PET and IHC is now ongoing. Further investigation<br />

either in animals<br />

with spontaneous<br />

stroke or with induced<br />

cerebral ischaemia will<br />

be required to compare<br />

the impact of co-morbidity<br />

and risk factors<br />

in this animal model.<br />

Acknowledgement: Prof. N. Rothwell and Dr H. Boutin<br />

are funded by MRC. This study was carried out<br />

within the EC-FP6 project DiMI (LSHB-CT-2005-<br />

512146) framework.<br />

References:<br />

1. Dirnagl U. (2006) J Cereb Blood Flow Metab. 26:1465-<br />

1478;<br />

2. McColl B.W. et al. (2009) Neuroscience. 158:1049-<br />

1061;<br />

3. Schwarz A.J. et al. (2006) Neuroimage. 32:538-550;<br />

4. Maroy R. et al. (2008) IEEE Trans.Med.Imaging. 27:342-<br />

354;<br />

5. Maroy R. et al. (2010) J Nucl Med (submitted).


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

Detection of inflammatory diseases by NIRF imaging with specific probes targeting<br />

leukotriene receptor CysLT 1 R<br />

Busch C. (1) , Passon M. (1) , Lehmann F. (2) , Socher I. (1) , Kaiser W.A. (1) , Hilger I. (1) .<br />

(1) University Hospital Jena, IDIR, Jena, Germany<br />

(2) DYOMICS GmbH, Jena, Germany<br />

Corinna.Busch@med.uni-jena.de<br />

Introduction: Leukotriene synthesis occurs early<br />

in inflammatory processes and plays a major role<br />

for the recruitment of leukocytes to the inflamed<br />

region (1). A key representative of these eicosanoids,<br />

leukotriene D4 (LTD4), shows high affinity<br />

to G-protein coupled cysteinyl leukotriene<br />

receptor 1 (CysLT 1 R). Detection of CysLT 1 R by<br />

molecular imaging (2) with near-infrared (NIR)<br />

fluorophores could be a suitable diagnostic tool<br />

for early identification of inflammatory processes.<br />

Aim of this study was the design and characterization<br />

of NIRF-based contrast agents specifically<br />

targeting CysLT 1 R in order to develop future diagnostic<br />

tools for inflammatory diseases, particularly<br />

in early stages.<br />

Methods: Polyclonal rabbit CysLT 1 R antibody<br />

(CysLT 1 R*DY-734) or polyclonal rabbit-IgG<br />

(IgG*DY-734) as well as the corresponding Fab<br />

fragments (Fab-CysLT 1 R*DY-734, Fab-IgG*DY-<br />

734) were bound to activated NHS ester of NIRfluorophore<br />

DY-734 (Dyomics, Jena, Germany).<br />

Probes were characterized by determining dye/protein<br />

ratios. After verification of CysLT 1 R expression<br />

(PCR, flow cytometry) in HL-60 cells, binding<br />

of the synthesized probes in vitro was assessed by<br />

flow cytometry. In vivo, an ear edema was induced<br />

in mice (3), and NIR fluorescence was measured<br />

with whole body imaging system Maestro2.2 after<br />

i.v.-probe administration. Ex vivo biodistribution<br />

studies were performed.<br />

Results: Flow cytometry proved binding of CysLT 1 R*DY-<br />

734 and IgG*DY-734 to CysLT 1 R-expressing HL-60. In<br />

vivo, all probes revealed stronger binding to the edematous<br />

than to the corresponding healthy region. 6<br />

h post injection, specific CysLT 1 R*DY-734 and Fab-<br />

CysLT 1 R*DY-734 demonstrated 1.9 and 1.2 fold higher<br />

binding than IgG*DY-734 and Fab-IgG*DY-734,<br />

respectively. Investigation of isolated organs revealed<br />

that full length IgG´s are eliminated via liver, while<br />

Fab fragments additionally accumulated in kidney.<br />

Conclusions: A novel NIRF-based probe specifically targeting<br />

Leukotriene D 4 receptor CysLT 1 R allows detection<br />

of inflammatory processes in ear edema-induced<br />

mice in early stages.<br />

Acknowledgement: The present investigations were<br />

supported by the “Deutsche Forschungsgemeinschaft”<br />

within the DFG program Hi 689/6-1.<br />

References:<br />

1. Hui Y, Funk C. Cysteinyl leukotriene receptors. Biochem.<br />

Pharmacol. 2002;64:1549-57.<br />

2. Weissleder R, Mahmood U. Molecular Imaging.<br />

Radiology 2001;219:316-33.<br />

3. Kurnatowska I, Pawlikowski M. Anti-inflammatory<br />

effects of somatostatin analogs on zymosaninduced<br />

earlobe inflammation in mice:<br />

comparison with dexamethasone and ketoprofen.<br />

Neuroimmunomodulation 2001;9:119-24.<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

P-086<br />

poStEr<br />

INFECTION and INFLAMMATION


200<br />

WarSaW, poland May 26 – 29, 2010<br />

P-087 PET imaging of Hypoxia by 18 F-Fluoromisonidazole ([ 18 F]FMISO) to detect early stages of<br />

experimental arthritis<br />

Fuchs K. (1) , Grießinger C. (1) , Fischer K. (1) , Mannheim J. (1) , Wiehr S. (1) , Judenhofer M. (1) , Reischl G. (2) , Röcken M. (3) ,<br />

Pichler B. (1) , Kneilling M. (3) .<br />

(1) Laboratory for Preclinical Imaging and Imaging Technology of the Werner Siemens-Foundation, Department of Radiology, Eberhard- Karls University,<br />

Tübingen, Germany<br />

(2) Radiopharmacy, Department of Radiology, Eberhard- Karls University, Tübingen, Germany<br />

(3) Department of Dermatology, Eberhard- Karls University, Tuebingen, Germany University Hospital Tübingen - Department of Radiology, Tübingen, Germany<br />

Kerstin.Fuchs@med.uni-tuebingen.de<br />

Introduction: Early detection of autoimmune diseases<br />

such as rheumatoid arthritis is essential for<br />

early interventional anti-inflammatory treatment<br />

to prevent cartilage- and bone deterioration. Hypoxia<br />

can induce angiogenesis via stabilization of<br />

the transcription factor hypoxia inducible factor<br />

(HIF)-1α/2α in resident and infiltrating cells by induction<br />

of pro-angiogenic mediators. Also, hypoxia<br />

is hypothesized to be one of the early indicators of<br />

inflammation. The aim of our study was to examine<br />

initial phases of hypoxia-induced angiogenesis and<br />

inflammation in rheumatoid arthritis, by in vivo<br />

small animal PET, even before clinical symptoms<br />

or histological joint inflammation are detectable.<br />

Therefore, we used the hypoxia tracer 18 F-Fluoromisonidazole<br />

([ 18 F]FMISO), which selectively accumulates<br />

in hypoxic tissue, and [ 18 F]Fluordesoxyglucose<br />

([ 18 F]FDG).<br />

Methods: We induced arthritis in BALB/c mice via<br />

intraperitoneal injection of serum containing autoantibodies<br />

against glucose-6 phosphate-isomerase<br />

(GPI). Mice underwent [ 18 F]FMISO-, or [ 18 F]FDG<br />

- PET investigations, 6 - 52 hours after induction<br />

of GPI-arthritis. Additionally, we performed H&Estaining,<br />

Western Blot (HIF-1α/2α) and real-time<br />

PCR analysis of gene expression patterns (bFGF,<br />

VEGF, IL-1β, TNF, IL-6, and COX-2) in joint tissue<br />

6 and 12 hours after initiation of arthritis.<br />

Results: Starting 6 hours after induction of GPI-arthritis<br />

we detected an enhanced [ 18 F]FMISO uptake<br />

in arthritic joints compared to healthy joints. Differences<br />

in [ 18 F]FMISO uptake between arthritic<br />

and healthy joints reached a level of significance 13<br />

hours after induction of GPI-arthritis. In contrast<br />

to [ 18 F]FMISO, no increase in [ 18 F]FDG-uptake was<br />

detectable at these early time points (6-13 hours).<br />

Comparable to the in vivo [ 18 F]FDG-PET data, no<br />

histological visible signs of arthritis were examined<br />

in H&E-stained slices of arthritic joint tissue. In line<br />

with the in vivo [ 18 F]FMISO-PET-data, RT-PCR analysis<br />

performed 6 hours after GPI-serum injection<br />

showed a 7.5-fold enhanced expression of HIF-2α<br />

mRNA. Interestingly, mRNA-levels of pro-angiogenic<br />

imaging life<br />

and pro-inflammatory mediators such as bFGF and<br />

VEGF, TNF, IL-6, and COX-2 were not elevated 6h<br />

after induction of GPI-arthritis. Starting 54h after<br />

induction of GPI-arthritis we detected significant<br />

differences in [ 18 F]FDG uptake in arthritic ankles,<br />

and a 6.5-1550 fold enhanced expression NF- k B induced<br />

genes such as COX-2, IL-6, IL-1β and, TNF.<br />

Conclusions: Non invasive in vivo examination of<br />

hypoxia-induced angiogenesis using [ 18 F]FMISO<br />

is a powerful tool to detect initial phases of angiogenesis<br />

in autoimmune diseases such as rheumatoid<br />

arthritis even before joint inflammation becomes<br />

detectable by other methods.


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

Intracellular [ 64 Cu]PTSM and extracellular [ 64 Cu]DOTA-antibody labelling of ovalbuminspecific<br />

Th1 cells for in vivo PET investigations of Th1 cell trafficking in OVA-specific lung<br />

inflammation<br />

Griessinger C.M. (1) , Wiehr S. (1) , Bukala D. (1) , Kesenheimer C. (1) , Röcken M. (1) , Ehrlichmann W. (3) , Reischl G. (3) , Pichler B. (1) ,<br />

Kneilling M. (2) .<br />

(1) Laboratory for Preclinical Imaging and Imaging Technology of the Werner Siemens-Foundation, Department for Radiology, Eberhard Karls University of<br />

Tübingen, Germany<br />

(2) Department for Dermatology, Eberhard Karls University of Tübingen, Germany<br />

(3) Radiopharmacy, Eberhard Karls University of Tübingen, Germany<br />

Christoph.Griessinger@med.uni-tuebingen.de<br />

Introduction: T helper cells play an important role<br />

in the development of autoimmune diseases. For detailed<br />

in vivo analysis of the migration properties of<br />

Th1 cells, high sensitive imaging modalities, such<br />

as small animal PET are powerful tools. So far basic<br />

migration properties like kinetics, homing, and<br />

sites of T cell proliferation in animal models for autoimmune<br />

diseases are still poorly understood. The<br />

aim of our study was to establish new T cell labelling<br />

strategies to gain new insights in Th1 cell trafficking<br />

in vivo using small animal PET. In our studies Th1<br />

cells were in vitro labelled intracellularly with the<br />

lipophilic tracer [ 64 Cu]PTSM or extracellularly with<br />

[ 64 Cu]DOTA-linked antibodies prior to injection<br />

into diseased mice and tracking by small animal PET.<br />

Methods: To investigate whether intracellular [ 64 Cu]<br />

PTSM labelling or extracellular [ 64 Cu]DOTA-antibody<br />

labelling impair ovalbumin (OVA)-specific<br />

Th1 cells, we analysed cell viability and functionality.<br />

OVA-T cell receptor (TCR) transgenic CD4+ T<br />

cells were isolated from spleen and lymph nodes of<br />

DO.11.10 mice and cultured together with irradiated<br />

antigen presenting cells (APC), Oligo 1668 peptide,<br />

anti-IL-4, and IL-2 for 12-14 days. 10 6 OVA-Th1 cells<br />

were labelled with 0.7 MBq [ 64 Cu]PTSM for 3 hours<br />

or with 0.7 MBq radiolabelled OVA-TCR-specific<br />

antibody (KJ1-26), which was linked to [ 64 Cu] via<br />

the chelator DOTA, for 0.5 hours. Th1 cell viability<br />

was assessed by trypan blue staining after incubation<br />

with increasing amounts of activity. Specific<br />

Th1 cell functioning was analyzed through interferon-gamma<br />

(ELISA) levels in supernatants of specific<br />

activated OVA-Th1 cells (T cells + irradiated APC +<br />

OVA peptide). In vivo T cell migration was investigated<br />

in an animal model for OVA-induced lung<br />

inflammation. Mice were sensitized with OVA (i.p.)<br />

and challenged intranasally twice after four weeks<br />

to induce OVA-specific lung inflammation. A total<br />

of 10 7 [ 64 Cu]PTSM or [ 64 Cu]DOTA-KJ1-26 antibody<br />

labelled OVA-Th1 cells were injected i.p. into diseased<br />

and healthy mice. Static PET-scans in combination<br />

with CT, biodistribution, and autoradiography<br />

were performed 24 and 48 hours after OVA-Th1<br />

cell transfer.<br />

Results: In vitro investigations revealed an activity<br />

dependent impairment of OVA-Th1 viability and<br />

functionality after [ 64 Cu]PTSM or [ 64 Cu]DOTA-<br />

KJ1-26 antibody labelling. After a time period of 24<br />

hours post labelling, the cell viability sunk to 80%<br />

and functionally was decreased by 20% compared<br />

to unlabelled control cells. Analyzing OVA-specific<br />

Th1 cell migration in the mouse model for lung inflammation,<br />

we detected an accumulation of [ 64 Cu]<br />

PTSM labelled OVA-Th1 cells in lung tissue and the<br />

thymus already 24h after the final challenge. Detection<br />

of [ 64 Cu]DOTA-KJ1-26 antibody labelled OVA-<br />

Th1 cells was even possible for up to 48 hours. In<br />

vivo PET data were further confirmed by ex vivo<br />

biodistribution and autoradiography. Compared to<br />

[ 64 Cu]PTSM labelled OVA-Th1 cells we gained a<br />

more defined distribution of extracellularly [ 64 Cu]<br />

DOTA-KJ1-26 labelled cells in the peritoneum (the<br />

site of injection), draining lymphatic tissue and the<br />

sites of OVA-specific lung inflammation. Furthermore<br />

we detected a significant accumulation of<br />

OVA-Th1 cells in the omentum majus.<br />

Conclusions: Both [ 64 Cu]-based labelling methods<br />

cause an impairment of Th1 cells viability and functionality.<br />

However, both labelling strategies are applicable<br />

for in vivo PET-imaging, revealing a detection<br />

limit of 500 OVA-Th1 cells in draining lymph<br />

nodes and sites of OVA-specific lung inflammation<br />

over a time period of 48 hours.<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

P-088<br />

poStEr<br />

INFECTION and INFLAMMATION


202<br />

WarSaW, poland May 26 – 29, 2010<br />

P-089 [ 18 F]DPA-714, [ 18 F]PBR111 and [ 18 F]FEDAA1106: Radiosyntheses on a TRACERLAb FX-FN<br />

synthesizer<br />

Kuhnast B. , Damont A. , Demphel S. , Le Helleix S. , Boisgard R. , Tavitian B. , Dollé F. .<br />

CEA, Orsay, France<br />

bertrand.kuhnast@cea.fr<br />

Introduction: Neuroinflammation is involved in<br />

acute and chronic neurological disorders through<br />

the activation of microglial cells or the recruitment<br />

of peripheral macrophages. Both microglia and<br />

macrophage activation results in the notable overexpression<br />

on the outer mitochondrial membranes<br />

of the so-called translocator protein (TSPO 18 kDa),<br />

supporting today extensive efforts in the design of<br />

radioligands for the in vivo imaging of this pharmacological<br />

target by Positron Emission Tomography<br />

[1]. Of particular interest are DPA-714 [2], PBR111<br />

[3] and FEDAA1106 [4], three ligands belonging to<br />

different chemical classes (the pyrazolo[1,5-a]pyrimidineacetamides,<br />

the imidazo[1,2-a]pyridineacetamides<br />

and the N-benzyl-N-(2-phenoxyaryl)acetamides,<br />

respectively) but all designed for a<br />

labelling with the positron-emitter fluorine-18 via a<br />

tosyoxy-for-fluorine nucleophilic aliphatic substitution.<br />

Production of [ 18 F]DPA-714, [ 18 F]PBR111 and<br />

[ 18 F]FEDAA1106 on the advanced, commercially<br />

available, automated module TRACERLab TM FX-FN<br />

synthesizer is presented.<br />

Methods: The automated process implemented on<br />

the TRACERLab TM FX-FN synthesizer involves: (A)<br />

the preparation of the K[ 18 F]F-Kryptofix ® 222 complex<br />

in two heating steps, first at 60°C for 7 min<br />

under a stream of N 2 and then at 120°C under reduced<br />

pressure for 5 min, followed by (B) reaction<br />

of K[ 18 F]F-Kryptofix ® 222 with the appropriate tosylprecursors<br />

(4 to 5 mg) at 165°C for 5 min in DMSO<br />

(0.7 mL), then (C) SepPak ® Plus Alumina N cartridge<br />

pre-purification after dilution of the reaction mixture<br />

with HPLC solvent (4 mL) and finally (D) semipreparative<br />

HPLC purification. HPLC purifications<br />

were performed on a X-Terra® column for [ 18 F]DPA-<br />

714 (Solvent : NH 4 OAc 0.1M pH 10 / MeCN : 60/40<br />

(v/v), Flow rate : 5 mL/min, Rt = 11-12 min), on a<br />

Symmetry ® C-18 column for [ 18 F]PBR111 (Solvent :<br />

H 2 O / MeCN / PicB7 : 60/40/2 (v/v/v), Flow rate : 5<br />

mL/min, Rt = 14-15 min) and on a Symmetry ® C-18<br />

column for [ 18 F]FEDAA1106 (Solvent : H 2 O / MeCN<br />

/ TFA : 60/40/0.1 (v/v/v), Flow rate : 5 mL/min, Rt =<br />

21-22 min). The HPLC-collected fraction containing<br />

pure [ 18 F]DPA-714, [ 18 F]PBR111 or [ 18 F]FEDAA1106<br />

imaging life<br />

were automatically formulated using a SepPak ® Plus<br />

C-18 cartridge ((i) HPLC-collected fraction dilution<br />

and loading on the cartridge, (ii) cartridge washing<br />

with 10 mL of water, (iii) cartridge elution with 2 mL<br />

of EtOH, and (iv) final dilution with 8 mL of saline).<br />

The process was programmed on the TRACERLab TM<br />

FX-FN synthesizer in one single “method” divided<br />

in three “time-lists”.<br />

Results: Starting from a 37.0 GBq cyclotron-produced<br />

[ 18 F]fluoride batch, 6.7 to 8.5 GBq (18-23%<br />

non decay corrected yields) of [ 18 F]DPA-714 or [ 18 F]<br />

PBR111 or [ 18 F]FEDAA1106, > 99% radiochemically<br />

pure and ready-to-inject, were obtained within 50-<br />

60 min. The overall decay corrected radiochemical<br />

yield reached up to 30%. Specific radioactivities<br />

ranged from 74 to 222 GBq/µmol (2-6 Ci/µmol).<br />

Conclusions: Radiosynthesis of [ 18 F]DPA-714, [ 18 F]<br />

PBR111 and [ 18 F]FEDAA1106 has been successfully<br />

implemented on a TRACERLab TM FX-FN<br />

synthesizer.<br />

Acknowledgements: Supported in part by the EC -<br />

FP6-project DiMI (LSHB-CT-2005-512146) and<br />

EMIL (LSH-2004-503569).<br />

References:<br />

1. Dollé F et al; Curr Med Chem 16: 2899-2923 (2009)<br />

2. Damont A et al; J label Compds Radiopharm 51: 286-<br />

292 (2008)<br />

3. Dollé F et al; J label Compds Radiopharm 51: 435-439<br />

(2008)<br />

4. Zhang MR et al; J Med Chem 49: 2735-2742 (2006)


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

In vivo near-infrared fluorescence imaging of lung matrix metalloproteinases in an acute<br />

cigarette smoke-induced airway inflammation model in different mice strains<br />

Perez-Rial S., Del Puerto-Nevado L., Giron-Martinez A., Gonzalez-Mangado N., Peces-Barba G. .<br />

Instituto de Investigacion Sanitaria-Fundacion Jimenez Diaz-CIBERES, Madrid, Spain<br />

sperezr@fjd.es<br />

Introduction: Recent studies have suggested that<br />

pulmonary macrophage-derived metalloproteases<br />

(MMPs) are the critical mediators of cigarette smoke<br />

exposure (CSE)-induced emphysema, but it has never<br />

been seen via imaging techniques [1] . The purpose<br />

of this study was to visualize and quantify acute pulmonary<br />

inflammation by real-time near-infrared<br />

fluorescence (NIRF)-mediated molecular imaging<br />

[2] in the lung tissue evaluating MMPs activity in response<br />

to CSE in mice strains with different susceptibility<br />

to develop smoking-induced emphysema.<br />

Methods: To accomplish this task, susceptible<br />

(C57BL/6j) and resistant (129S2/SvHsd) mice [3]<br />

were exposed to acute CSE using a whole-body exposition.<br />

24h after CSE MMPs activity was assessed<br />

via optical imaging system by a MMPs-sensitive activatable<br />

fluorescence probe in order to characterize<br />

the distinctive profile of CSE-induced acute inflammation.<br />

Furthermore MMPs protein levels in the<br />

lung tissue were analyzed by Western blot analysis.<br />

Results: In vivo semiquan- in vivo lung imaging<br />

titative optical imaging<br />

analysis of MMPs activity<br />

in the lung revealed<br />

increased acute-CSE-associated<br />

MMPs activity in<br />

C57BL/6j but not in 129S2/<br />

SvHsd. Only in susceptible<br />

mice most important<br />

MMPs protein levels were<br />

significantly increased in the lung tissue of smokers<br />

compared with the non-smokers group.<br />

Conclusions: Optical imaging via NIRF offers a simple,<br />

effective, and rapid technique for noninvasive<br />

monitoring and semiquantitative analysis of lung<br />

inflammation and MMPs expression. We are able to<br />

distinguish between susceptible and resistant mice<br />

strains in terms of the profile of MMPs activity in<br />

the early stages of pulmonary disease. The results of<br />

our study suggest that mechanisms underlying different<br />

susceptibilities to CSE can be detected at the<br />

beginning of the disorder. They produce different<br />

response to acute tobacco smoke.<br />

Acknowledgement: This work is supported in part by the<br />

Spanish “Ministerio de Ciencia e Innovación” (SAF2008-<br />

05412-C02- 02) and CIBERES (CB06/06/0009).<br />

References:<br />

1. Churg A et al; Am J Respir Crit Care Med. 167(8):1083-9<br />

(2003)<br />

2. Haller J et al; J Appl Physiol. 104(3):795-802 (2008)<br />

3. Morris A et al; J Pharmacol Exp Ther. 327(3):851-62<br />

(2008)<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

P-090<br />

poStEr<br />

INFECTION and INFLAMMATION


204<br />

WarSaW, poland May 26 – 29, 2010<br />

P-091 Development and pre-clinical evaluation of a novel class of 18 F labelled PET ligands for<br />

evaluation of PBR/TSPO in the brain<br />

Trigg W. , Ahmad R. , Arstad E. , Avory M. , Hirani E. , Jones P. , Khan I. , Luthra S. , Morisson-Iveson V. , O’shea D. ,<br />

Passmore J. .<br />

GE Healthcare, Amersham, United Kingdom<br />

william.trigg@ge.com<br />

Introduction: The peripheral benzodiazepine receptor<br />

(PBR; otherwise known as TSPO (18kDa)) is a<br />

well-established target for imaging activated microglial<br />

cells and macrophages in inflammatory diseases<br />

such as MS, Alzheimer’s disease and a wide range<br />

of both peripheral and brain diseases 1 . Starting from<br />

a core tetracyclic indole pharmacophore 2 , which displayed<br />

high affinity for the PBR, we have designed<br />

a series of molecules to assess the SAR around the<br />

pharmacophore and to determine the best site to introduce<br />

the radiolabel.<br />

Methods: Compounds have been assessed for PBR<br />

affinity in a radioligand binding assay and in a range<br />

Compound (Ki)<br />

[ 11C]PK11195 (1.24nM)<br />

[ 18F]AH114011 (0.37nM)<br />

[ 18F]AH114629 (0.40nM)<br />

imaging life<br />

Initial brain<br />

uptake (%ID/g)<br />

OB @ 2 min<br />

(%ID/g)<br />

* Whole brain value not calculated<br />

** [3H]PK11195 used for autoradiography studies<br />

OB @ 30<br />

min<br />

(%ID/g)<br />

2:30 Striatum<br />

ratio<br />

OB : Striatum<br />

ratio<br />

(30 min)<br />

Metabolism<br />

Profile - % Parent<br />

in brain at 60<br />

min p.i.<br />

FNA Model lesion<br />

: non lesion ratio<br />

(in vitro)<br />

0.28-0.48* 0.42 0.21 3.20 2.10 100 2.8**<br />

0.32 0.39 0.31 1.73 2.07 96.0 2.3<br />

0.42 0.51 0.28 4.75 3.50 97.6 NT<br />

of in vitro ADME assays. Following on from the in<br />

vitro assessments, promising compounds were radiolabelled<br />

and assessed in vivo for biodistribution<br />

and metabolism profiles. The compounds with the<br />

most appropriate profiles were assessed using autoradiography<br />

in the Facial Nerve Axotomy (FNA)<br />

model described by Banati et al. 3<br />

The corresponding data for the archetypal PBR ligand<br />

PK11195 was generated for comparison.<br />

Results: The olfactory bulb (OB) has high expression<br />

of PBR and is used to measure specific uptake,<br />

with the striatum used as a low expression area for<br />

comparative purposes. Key data obtained are summarised<br />

in the table below.<br />

Conclusions: Preclinical evaluation of the 18F labelled<br />

tetracyclic indole class of ligands for PBR<br />

shows that this class of molecules represents a<br />

promising class of PET ligands that should be further<br />

evaluated.<br />

References:<br />

1. Cagnin et al., Neurotherapeutics, Vol. 4, No. 3, 443-452<br />

(2007)<br />

2. Okubo et al., Bioorg Med. Chem. 12(2):423-438 (2004)<br />

3. Banati et al., J. Neurocytol. 26:77-82. (1997)


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

Local administration of adeno-associated virus into the mammary gland ductules<br />

Hamm J. (1) , Cojoca R. (1) , Iezzi M. (2) , Mautino A. (1) , Turco E. (1) , Silengo L. (1) , Musiani P. (2) , Forni G. (1) .<br />

(1) University of Turin, Italy<br />

(2) University of Chieti Pescara, Italy<br />

jorg_hamm@yahoo.com<br />

Introduction: Adeno-associated virus (AAV) is a<br />

single-stranded DNA parvovirus with charateristics<br />

that render it attractive for gene therapy applications<br />

(1). More than 14 AAV serotypes have<br />

been characterized which differ in efficiency of<br />

infection and tissue tropisms. Cell entry seems to<br />

occur by AAV receptor mediated endocytosis, the<br />

receptors recognised by the capsid proteins vary in<br />

different serotypes and might be an determinant<br />

for tissue tropism. The DNA remains mainly extrachromosomal,<br />

does not replicate, and is unable to<br />

produce infectious particles, features desirable for<br />

gene therapy vectors. In the majority of the applications<br />

AAV2 vector is used for gene transfer experiments,<br />

but other serotypes have been explored<br />

recently because they can offer superior transduction<br />

efficiencies or different cell tropisms. Furthermore,<br />

neutralising antibodies against AAV2<br />

are frequently found in human sera and can compromise<br />

efficiency of gene transfer. AAV9 has been<br />

reported to allow a robust expression in heart and<br />

muscle, but other expression sites have also been<br />

indicated.<br />

Methods: AAV9 vectors expressing luciferase as a reporter<br />

gene driven by the cytomegalo virus promoter<br />

(AAV9-CMV-luc) or by the murine EF1alpha promoter<br />

(AAV9-EF1a-luc) were administered locally into<br />

the mammary gland network by intraductal injection<br />

(2) into BALB/c female mice. Expression profile and<br />

body distribution were determined by in vivo and<br />

ex vivo optical imaging, and by histological analysis.<br />

Results: AAV9-CMV-luc migrates from the injected<br />

gland to ipsilateral glands, but not to contralateral<br />

glands. Luciferase expression is observed predominantly<br />

in muscle cells and is stable for months. The<br />

observed expression of luciferase in the ipsilateral<br />

lymph nodes indicate that the virus might use the<br />

lymphatic system to reach all of the five ipsilateral<br />

mammary glands.<br />

Conclusions: AAV9 vectors appear to be well suited<br />

for targeted, long-lasting gene expression in muscle<br />

tissue. Mammary epithelia cells are apparently not<br />

infected efficiently, not even after site specific injection<br />

into the duct of mammary glands.<br />

References:<br />

1. Büning et al.; J Gene Med 2008; 10: 717–733.<br />

2. Murata et al.; Cancer Res. 2006 Jan 15;66(2):638-45.<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

P-092<br />

poStEr<br />

TARGETED THERAPY


206<br />

WarSaW, poland May 26 – 29, 2010<br />

P-093 Copper-64- and Gallium-68- NODAGA-conjugated bombesin antagonists as new PET tracers<br />

Mansi R. , Dumont R. , Jamous M. , Tamma M. , Nicolas G. , Fani M. , Cescato R. , Reubi J.C. , Weber W. , Maecke H. .<br />

Nuclear Medicine, Freiburg, Germany<br />

rosalba.mansi@uniklinik-freiburg.de<br />

Introduction: The gastrin-releasing peptide or<br />

bombesin receptor (GRPr) is frequently overexpressed<br />

in several human cancers including prostate,<br />

breast, small cell lung and pancreatic cancer<br />

[1,2]. Specific radiolabeled ligand for this receptor<br />

offer new opportunities for the diagnosis and<br />

treatment of these diseases. Many bombesin antagonists<br />

have shown high affinity for the GRPr<br />

[3]. In this study, we evaluated the potential of a<br />

statine-based bombesin antagonist, conjugated to<br />

NODAGA through a polyethyleneglycol spacer, to<br />

specifically target GRPr expressing cancer cells.<br />

We determine the effect of two different radioisotopes<br />

Ga-68 and Cu-64 on the tumor targeting efficacy<br />

and in vivo pharmacokinetics. Moreover we<br />

evaluated the PET imaging properties of the two<br />

radiotracers.<br />

Methods: The peptide assembling and NODAGAconjugation<br />

were accomplished on solid phase. The<br />

conjugate MJ5 was radiolabeled with Ga-68 and<br />

Cu-64 and the radioconjugates were evaluated in<br />

vitro and in vivo in tumor-bearing nude mice, using<br />

the GRP-receptor positive prostate carcinoma<br />

cell line PC-3. To study the bombesin antagonistic<br />

properties of MJ5 cells were treated either with 10<br />

nmol/L bombesin, or with 1 µmol/L MJ5 or, with<br />

10 nmol/L bombesin in the presence of a 100-fold<br />

excess MJ5 for 30 min at 37°C. GRPr internalization<br />

was studied by immunofluorescence.<br />

Results: The immunofluorescence assays confirmed<br />

the strong antagonist properties of the conjugates: MJ5<br />

failed to induce significant internalisation of GRPr and<br />

when given at a concentration of 1 µmol/L together with<br />

10 nmol/L bombesin, MJ5 was able to antagonize bombesin-induced<br />

receptor internalisation. Biodistribution<br />

studies revealed high and specific uptake of both conjugates<br />

in PC-3 tumors and in GRPr positive tissues such<br />

as pancreas and intestine. The Ga-68-MJ5 biodistribution<br />

data showed a good tumor uptake at 1h (4.54±0.52%<br />

I.A./g) and good tumor to kidney and tumor to blood<br />

ratios (4.1 and 10.9 respectively). These promising data<br />

prompted us to investigate the biodistribution of MJ5<br />

with a longer lived radionuclide. The tumor uptake of<br />

the Cu-64-MJ5 was 3.7 times higher than the Ga-68labeled<br />

analog at 1h (16.80±3.22% I.A./g) and remained<br />

imaging life<br />

high at 4h (13.97±3.68% I.A./g). At 4 h higher tumor to<br />

kidney (8.6), tumor to blood (36.2) and tumor to pancreas<br />

(18.2) ratios were achieved than for Ga-68-MJ5 at<br />

1 h. The PET/CT images demonstrated exellent visualization<br />

of the PC3 tumors by Ga-68- and Cu-64 MJ5.<br />

Conclusions: The labeling with the two radionuclides<br />

distinctly influences the pharmakokinetics of the radiopeptides<br />

in particular in regard to the uptake in<br />

the tumor and receptor positive organs. The high<br />

tumor uptake and the good tumor to background<br />

ratio of the Cu-64 labeled analog at 4h warrant clinical<br />

testing of this probe for PET imaging in humans.<br />

Acknowledgement: We acknowledge COSTD38 and<br />

the Swiss National Science Foundation for the financial<br />

support.<br />

References:<br />

1. R.Markwalder, J.C. Reubi, Cancer Res., 155 (1999) 1152.<br />

2. J.C. Reubi, M. Korner, et al., Eur.J.Nucl.Med.Mol.Imaging,<br />

31 (2004) 803<br />

3. R.T. Jensen, D.H. Coy, Trends Pharmacol. Sci., 12 (1991) 13


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

A novel indocyanine green nanoparticle probe for non invasive fluorescence imaging in vivo<br />

Mérian J. (1) , Navarro F. (1) , Josserand V. (2) , Texier I. (1) .<br />

(1) CEA LETI MINATEC, Grenoble, France<br />

(2) CRI INSERM U823, France<br />

juliette.merian@cea.fr<br />

Introduction: Fluorescence imaging, a non invasive,<br />

non ionizing and sensitive technology, is opening a<br />

new era in medical application such as image-guided<br />

surgery. Indocyanine Green (ICG), the only Near InfraRed<br />

optical contrast agent approved by the FDA<br />

[1], is encapsulated in lipidic nanoparticles (LNP).<br />

Using LNP as nanocargo presents advantages such as<br />

improvement of the dye chemical stability, biocompatibility<br />

and passive targeting, as well as suitable<br />

properties for fluorescence detection of tumors [2].<br />

Methods: LNP are composed of a lipid core, stabilized<br />

by phospholipids and pegylated surfactants and are<br />

dispersed in aqueous buffer. ICG (Infracyanine) is<br />

incorporated into the lipid mixture as a concentrated<br />

solution in ethanol. Solvent is evaporated and this<br />

oily phase is mixed with aqueous phase before sonication<br />

is performed at 40°C for a whole 5min period<br />

(VCX750 Ultrasonic processor). Particle size, size<br />

distribution, and surface charges are evaluated by<br />

dynamic light scattering (DLS). Optical properties<br />

Nano ICG<br />

Free ICG<br />

characterizations are performed using visible absorbance<br />

and fluorescence spectrometries. MTT assay<br />

is performed using 3T3 fibroblasts to assess LNP cytotoxic<br />

index. The in vivo distribution of ICG-loaded<br />

LNP is investigated in Nude mice bearing xenografted<br />

sub-cutaneous TS/Apc tumors using a NIR<br />

whole animal imaging device (Fluobeam 800), and<br />

compared to that of free ICG in aqueous solution.<br />

Results: ICG loading does not modify the size (30nm<br />

diameter, one population) and the polydispersity index<br />

(from 0.11 to 0.13) of the LNP, but decreases<br />

the zeta potential, which becomes more negative<br />

(-5.8±2.5mV for nude LNP, versus -18.2±2.1mV for<br />

ICG loaded LNP). The dye encapsulation within<br />

LNP induces a 16nm red-shift of its absorption and<br />

emission (maximum at 820nm); as well as an improvement<br />

of its fluorescence quantum yield and a<br />

longer lifetime in comparison to free dye in water.<br />

LNP carrier is well tolerated in vitro (IC50~1 mg/mL<br />

of lipids). Furthermore, ICG loaded LNP injection<br />

in Nude mice implanted with subcutaneous Ts/Aps<br />

tumors, shows a better fluorescence signal in tumor<br />

site in comparison of free ICG at the same dose.<br />

Conclusions: LNP vectorization provides ICG a<br />

modification of its pharmacokinetics in addition<br />

to an improvement of its optical properties. These<br />

characteristics are suitable for long term and sensitive<br />

in vivo imaging required for efficient tumor<br />

detection. ICG loaded LNP, thanks to their low size,<br />

takes advantages of the Enhanced Permeability and<br />

Retention (EPR) effect and are passively accumulated<br />

with time into the tumor.<br />

Acknowledgement: J.Mérian1, F. Navarro 1 , V. Josserand 2 ,<br />

I. Texier 1 : 1CEA LETI Minatec, 2 CRI-INSERM U823<br />

References:<br />

1. Frangioni, J.V. In vivo near infrared fluorescence<br />

imaging. Curr. Opinion Chem. Biol. 7, 2003:626-634<br />

2. Texier, I. Goutayer, M. and al Journal of Biomedical<br />

Optics, 14(5), 2009: 054005<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

P-094<br />

poStEr<br />

TARGETED THERAPY


WarSaW, poland May 26 – 29, 2010<br />

P-095 Evaluation and optimization of the concept of an antibody directed enzyme-prodrug therapy<br />

using noninvasive imaging technologies<br />

208<br />

Napp J. (1) , Dullin C. (2) , Krewer B. (3) , Von Hof J.M. (3) , Schmuck K. (3) , Mathejczyk J. (4) , Hartung F. (4) , Stühmer W. (4) , Tietze L. F. (3) ,<br />

Alves F. (4) , Pardo L. A. (4) .<br />

(1) MI for exp Medicine, Göttingen, Germany<br />

(2) University Medicine Göttingen, Germany<br />

(3) University Göttingen, Germany<br />

(4) MPI for exp Medicine, Göttingen, Germany<br />

jnowako1@gwdg.de<br />

Introduction: Relatively low selectivity to tumor versus<br />

normal cells challenges virtually all cancer chemotherapies.<br />

Site-specific activation of prodrugs in<br />

tumors is one strategy to achieve high efficacy and<br />

specificity of treatment, decreasing toxicity in normal<br />

tissues. Here, we present the design and validation<br />

of an Antibody Directed Enzyme-Prodrug Therapy<br />

(ADEPT) in which an antibody against Eag1 is used<br />

to carry the drug-activating enzyme, ß-galactosiase<br />

(ß-gal) to the tumor tissue. Eag1 (ether-à-go-go1)<br />

voltage-gated potassium channel has been chosen as<br />

a tumor-specific target since this plasma-membrane<br />

protein is easily accessible to extracellular interventions<br />

and Eag1 is aberrantly expressed (>75%) in tumors<br />

from diverse origin but basically not detected<br />

in healthy tissue outside the central nervous system.<br />

Methods: Two monoclonal anti-Eag1 antibodies,<br />

mAb62 and mAb56, a humanized 56 antibody, as<br />

well as a single chain fragment of the mAb62, scFv62,<br />

were tested for their feasibility to deliver the drugactivating<br />

enzyme ß-gal to the tumor. Near infrared<br />

fluorescence (NIRF) imaging was used to study<br />

the biodistribution and binding characteristics of<br />

the anti-Eag1 antibodies in vivo in a subcutaneous<br />

Eag1-expressing MDA-MB-435S tumor model in<br />

nude mice. Fluorescence intensity, lifetime and location<br />

of Cy5.5 labeled antibodies in vivo was measured<br />

with the time-domain NIRF imager, Optix<br />

MX2 (ART, Advanced Research Technologies; Canada),<br />

at certain time points. Fluorescence lifetime<br />

(the average time during the molecule stays in its excited<br />

state) was used to discriminate between nonspecific<br />

and probe-derived signals. Distribution of<br />

the fluorescent probe in tumor sections ex vivo was<br />

further investigated by the Odyssey infrared imaging<br />

system (LI-COR Biosciences, Germany) as well<br />

as by NIRF microscopy (Axiovert 200M, Carl Zeiss,<br />

Germany). For the tumor-specific activation of the<br />

prodrug, the mAb62 was conjugated to ß-gal, resulting<br />

in 62-gal. The ß-gal activity of the 62-gal conjugate<br />

and its ability to bind to the Eag1 epitope were<br />

tested in vitro on Eag1-expressing MDA-MB-435S<br />

cells and control AsPC-1 cells using a colorimetric<br />

CPRG (chlorophenolred-ß-D-galactopyranoside)<br />

imaging life<br />

assay. The ß-gal activity in mice was analyzed by<br />

NIRF imaging using the fluorescent activatable probe,<br />

DDAOG (9H-(1,3-dichloro-9,9-dimethylacridin-2-one-<br />

7-yl) β-d-galactopyranoside) by NIRF imaging.<br />

Results: All tested antibodies targeting Eag1 bound<br />

specifically to MDA-MB-435S tumors with maximal<br />

intensity peaks at 24-48 h after application; fluorescence<br />

was still detectable for at least 1 week in vivo.<br />

We confirmed specific binding of the antibodies to<br />

the tumors by ex vivo NIRF imaging of tumors isolated<br />

from mice injected with fluorescently labeled<br />

antibodies 24 h prior to section, as well as by NIRF<br />

microscopy of tumor slices. Moreover, we show that<br />

monoclonal antibodies against Eag1, but not the<br />

scFv62 fragment, resulted in strong fluorescence<br />

signals in the area over liver, detectable for at least 4<br />

days in vivo. Similar fluorescence signals over liver<br />

were also observed in tumor-bearing mice injected<br />

with Cy5.5-labeled control IgGĸ2B, confirming that<br />

the liver signals did not result from Eag1-mediated<br />

binding of those antibodies to cells within liver.<br />

Furthermore, we show that the 62-gal conjugate<br />

specifically binds in vitro to Eag1-expressing MDA-<br />

MB-435S, but not to control Eag1-non expressing<br />

AsPC-1 cells and that 62-gal possess high ß-gal activity,<br />

when bound to MDA-MB-435S cells. Moreover,<br />

24 h after application of the 62-gal to the tumor<br />

bearing mice we detected ß-gal activity in vivo over<br />

the tumor area.<br />

Conclusions: Here, we successfully applied NIRF<br />

imaging to evaluate anti-Eag1 antibodies as tools for<br />

a novel concept of targeted cancer therapy. Since in<br />

vivo 62-gal 1) specifically binds to Eag1-expressing<br />

tumors and 2) shows measurable ß-gal activity at the<br />

tumor site, this conjugate can further be applied in<br />

the ADEPT for specific activation of cytotoxic prodrugs<br />

at the tumor site.


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

A novel [ 18 F] PET imaging agent for the epidermal growth factor receptor<br />

Pisaneschi F. (1) , Shamsaei E. (1) , Nguyen Q.D. (1) , Glaser M. (2) , Robins E. (2) , Kaliszczak M. (1) , Smith G. (1) , Spivey A.C. (1) ,<br />

Aboagye E.O. (1) .<br />

(1) Imperial College London, London, United Kingdom<br />

(2) Medical Diagnostic Discovery (part of GE Healthcare), United Kingdom<br />

f.pisaneschi@imperial.ac.uk<br />

Introduction: The Epidermal Growth Factor Receptor<br />

(EGFR/c-ErbB1/HER1) is overexpressed in<br />

many cancers including breast, ovarian, endometrial<br />

and non-small cell lung cancer. 1, 2 An EGFR specific<br />

imaging agent could facilitate clinical evaluation of<br />

primary tumours and/or metastases.<br />

Methods: We designed and synthesized a small array<br />

of fluorine containing compounds based on<br />

a 3-cyanoquinoline core. 3 The compounds were<br />

screened in vitro for their affinity in cell-free EGFR<br />

autophosphorylation assay and for their activity in<br />

the inhibition of the EGFR tyrosine kinase in EGFR<br />

overexpressing A431 cell lines.<br />

Results: A lead compound, incorporating 2’-fluoroethyl-1,2,3-triazole<br />

was selected for evaluation as a<br />

radioligand based on its high affinity for EGFR kinase<br />

(IC 50 = 1.81 ± 0.18 nM), good cellular potency<br />

(IC 50 = 21.97 ± 9.06 nM), low lipophilicity and good<br />

metabolic stability. ‘Click’ labelling 4 afforded [ 18 F]-<br />

2’-fluoroethyl-1,2,3-triazole derivative in 7% end<br />

of synthesis (EOS) yield from aqueous fluoride in a<br />

total synthesis time of 3 h and >99% radiochemical<br />

purity. The compound showed good stability in vivo<br />

and a 4-fold higher uptake in A431 tumour xenografts<br />

relative to muscle. Furthermore, the radiotracer<br />

could be visualized in A431 tumour bearing mice<br />

by small animal PET imaging (NUV 60 = 0.13±0.02).<br />

Conclusions: The novel [ 18 F]-imaging agent constitutes<br />

a promising radiotracer for further evaluation<br />

for imaging of EGFR status.<br />

Acknowledgement: This work was funded by Cancer<br />

Research UK programme grant C2536/A7602 and<br />

UK Medical Research Council core funding grant<br />

U.1200.02.005.00001.01.<br />

References:<br />

Bazley, L. A.; Gullick, W. J. Endoncr. Relat. Cancer 2005, 12,<br />

S17.<br />

Marmor, M. D.; Skaria, K. B.; Yarden, Y. Int. J. Radiat. Oncol.<br />

Biol. Phys. 2004, 58, 903.<br />

Wissner, A.; Overbeek, E.; Reich, M. F.; Floyd, M. B.; Johnson,<br />

B. D.; Mamuya, N.; Rosfjord, E. C.; Discafani, C.; Davis, R.;<br />

Shi, X.; Rabindran, S. K.; Gruber, B. C.; Ye, F.; Hallett, W. A.;<br />

Nilakantan, R.; Shen, R.; Wang, Y.-F.; Greenberger, L. M.; Tsou,<br />

H.-R. J. Med. Chem. 2003, 46, 49.<br />

Glaser, M.; Årstad, E. Bioconjugate Chem. 2007, 18, 989.<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

P-096<br />

poStEr<br />

TARGETED THERAPY


210<br />

WarSaW, poland May 26 – 29, 2010<br />

P-097 Structural methods in subcellular image analysis<br />

Dietlmeier J. , Whelan P.F. .<br />

Centre for Image Processing and Analysis, Dublin, Ireland<br />

julia.dietlmeier@ieee.org<br />

Introduction: From a computer vision perspective<br />

subcellular imaging is an extremely complex and dynamic<br />

environment as can be inferred from Fig.1(a).<br />

Mitochondria form an important category of membrane<br />

enclosed, on average 200nm large organelles<br />

which reside inside every living cell. Mitochondrial<br />

morphology is crucial to the understanding of<br />

apoptosis mechanisms and the subsequent development<br />

of therapies targeting age- and cancer-related<br />

diseases[1],[2]. There is a high demand in automated<br />

segmentation which can provide an objective quantitative<br />

information in a reasonable time frame[2].<br />

However, the state-of-the-art is dominated by manual<br />

tools. Early attempts to automate the segmentation<br />

are based on the machine-learning framework[3].<br />

Methods: Instead of pursuing the machine-learning<br />

pathway we approach the segmentation and localization<br />

problem from the structural point of view which<br />

also has an appealing theoretical aspect. Here, we<br />

apply the well developed and widely acknowledged<br />

framework of Mathematical Morphology. Our multistage<br />

segmentation targets (i) specific challenges<br />

of image acquisition such as low contrast, non-uniform<br />

illumination and speckle noise; (ii) structural<br />

challenges such as cluttered scene, clustered and deformed<br />

mitochondria and deformed cristae.<br />

Results: We outline our preliminary results by examining<br />

Fig.1. The first and the key step is localization<br />

which is based on the analysis of mitochondrial<br />

morphology. After extracting localiza tion markers<br />

we apply a seeded region grow algorithm which fills<br />

the inner-membrane space of each organelle, as can<br />

be seen in Fig.1(e). Gap-free control boundary is<br />

currently achieved by using morphological thickening<br />

and thinning operators on the binary image. In<br />

order to separate clustered mitochon-dria we apply a<br />

seeded watershed algorithm. The results of extracted<br />

sha-pes are provided in Fig.1(f)-(g). We verify that<br />

our segmentation works well on small-scale images<br />

containing a small number of mitochondria as can<br />

be seen in Fig.1(f)-(l). The other technique to obtain<br />

mitochondrial contour is recon-struction-by-dilation.<br />

Here, we correlate the localization markers with<br />

extracted contours in order to yield correct result.<br />

imaging life<br />

Fig.1 (a) TEM image of STS-treated DU-145 prostate cell. (b). Example<br />

of two clustered mitochondria. (c)-(l) Selected segmentation results.<br />

Conclusions: The major challenge is firstly to extract<br />

the mitochondrial shape within reasonable<br />

accuracy. Secondly, we attempt to expand our detection<br />

approach to a larger scale problem such as<br />

the whole cell image. We plan to validate this on a<br />

larger representative mitochondria image database<br />

and in doing so compare our approach to existing<br />

machine-learning algorithms.<br />

Acknowledgement: This research was supported by<br />

NBIP Ireland funded under the Higher Education<br />

Authority PRTLI Cycle 4, co-funded by the Irish<br />

Government and the European Union - Investing in<br />

your future. Special thanks to Royal College of Surgeons<br />

in Ireland (RCSI) for sample preparation and<br />

acquisition of TEM images. We would like to thank<br />

American Society for Cell Biology (ASCB) and Nature<br />

Publishing Group3 for the permission to use<br />

selected micrographs.<br />

References:<br />

1. Perkins GA et al; Methods in Enzymology. 456:29–52<br />

(2009)<br />

2. Sun MG et al; Nature Cell Biology. 9:1057–1065 (2007)<br />

3. Narasimha R et al; Pattern Recognition. 42:1067–1079<br />

(2009)


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

In vivo-post mortem multimodal image registration in a rat glioma model<br />

Dubois A. , Boisgard R. , Jego B. , Lebenberg J. , Hérard A. S. , Dollé F. , Lebon V. , Delzescaux T. , Tavitian B. .<br />

CEA-I2BM, Orsay, France<br />

albertine.dubois@cea.fr<br />

Introduction: Histological staining techniques can<br />

identify the regulation of specific biomarkers in<br />

small animal models of glioma and provide indicators<br />

of cellular dysfunction. The correlation of in<br />

vivo imaging signal changes and molecular indicators<br />

of tissue damage in histological brain sections<br />

represents an important means of understanding the<br />

cellular mechanisms responsible for these changes<br />

under pathological conditions. This requires an accurate<br />

image registration that can compensate the<br />

distortions that occur in the brain during the extraction,<br />

fixation, and staining process. Here, we present<br />

the preliminary results obtained by applying an<br />

overall registration strategy for the fusion of in vivo<br />

PET and MRI data with post mortem brain images in<br />

a rat 9L-glioma model.<br />

Methods: Experiments were conducted on male<br />

Wistar rats. Twelve days after intrastriatal injection<br />

of 9L rat glioma cells, we acquired T2-weighted MRI<br />

and 60 min dynamic PET using [18F]DPA-714 [1]<br />

for in vivo evaluation of peripheral benzodiazepine<br />

receptor (PBR) expression. Following in vivo imaging,<br />

animals were euthanized. The entire brains were<br />

cut into 20 µm-thick coronal sections and processed<br />

for H&E staining and PBR immunohistochemistry.<br />

A blockface photograph was also recorded prior to<br />

each section. The overall co-registration strategy relied<br />

on using the blockface photographs as an intermediate<br />

reference, onto which the in vivo MRI and<br />

PET images and the digitized histo- and immunohistochemical<br />

brain sections were registered separately<br />

and superimposed so as to obtain in vivo-post<br />

mortem registration [2;3].<br />

Results: By way of feasibility study, the co-registration<br />

strategy was only performed on one rat’s right<br />

hemisphere. As illustrated in Figure 1, after each registration<br />

task, both the external contours, the outer<br />

edges of the cortex and inner structures such as the<br />

corpus callosum, the hippocampus, the striatum and<br />

even the tumor were correctly superimposed whatever<br />

the modality.<br />

Conclusions: We obtained promising registration<br />

results of T2-weighted MRI and [18F]DPA-714 PET<br />

with post mortem brain volumes. Once results from<br />

additional animals had been provided, our approach<br />

could be used to evaluate, quantify and compare<br />

tumor volume, pharmacokinetic and physiological<br />

parameters (e.g. ligand differential uptake areas<br />

Fig1: Fusion of (A) PET (rainbow)<br />

and MRI (black and<br />

white), (B) Blockface (black<br />

and white) and H&E (pink)<br />

volumes, (C) MRI (black<br />

and white) and blockface<br />

volume (rainbow), (D) MRI<br />

(black and white) and H&E<br />

volume (pink) and (E) PET<br />

(rainbow) and H&E volume<br />

(black and white)<br />

within the tumor) measured from in vivo MRI and<br />

PET data with those derived from corresponding<br />

post mortem histo- and immunohistochemistry.<br />

References:<br />

1. Damont A et al; J label Compds Radiopharm. 51 (7):<br />

286-292 (2008)<br />

2. Dauguet J et al; J Neurosci Methods. 164 (1): 191–204<br />

(2007)<br />

3. Lebenberg J et al; Neuroimage. In press (2010)<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

P-098<br />

poStEr<br />

MI DATA ANALYSIS METHODS


212<br />

WarSaW, poland May 26 – 29, 2010<br />

P-099 Feasibility and success of Independent Component Analysis of resting state fmri data from<br />

the rat<br />

Jonckers E. , Van Auderkerke J. , Van Der Linden A. , Verhoye M. .<br />

Bio-Imaging Lab, Sint Lenaarts, Belgium<br />

elisabeth.jonckers@ua.ac.be<br />

Introduction: Resting state fMRI is used with growing<br />

interest with the purpose to spot age1 or pathology2<br />

induced abnormalities in functional connectivity<br />

(FC) in the human brain. Only recently resting state<br />

FC measurements were acquired in rats3, opening the<br />

potential of assessing FC in the rat brain in several<br />

imaging life<br />

Fig 1: 3 Components resulting from ICA of rat resting state data. Colors representing z-values, higher z-values<br />

meaning a higher convergence between time course of the voxel and mean time course for the component.<br />

neuropathologies (e.g. schizophrenia). The human<br />

resting state networks are identified using a seed-based<br />

approach or independent component analysis (ICA)4.<br />

Since ICA analysis of resting state fMRI of rodents is<br />

up to now not published, we performed a pilot study,<br />

implementing the technique in rodents and testing<br />

the reproducibility of the ICA outcome.<br />

Methods: A group of 5 male rats was imaged 4 times.<br />

On 2 time points with one week in between 2 consecutive<br />

resting state data were acquired using single<br />

shot gradient echo EPI. Rats were anesthetized with<br />

Medetomidine. Imaging was done on a 9.4T scanner<br />

using TR 2s and TE 16ms. 12 slices of 1 mm were<br />

acquired with a FOV of (3x3) cm and matrix size of<br />

128x128. ICA was implemented on the dataset using<br />

GIFT (Group ICA of fMRI toolbox), working in matlab2008.<br />

The number of components for the ICA of<br />

the individual subjects or group (5 rats x 4 repetitions)<br />

was set at 15.<br />

Results: The outcome of our ICA analysis, clearly<br />

revealed different brain regions within 10 of the 15<br />

components. Figure 1 demonstrates 3 components of<br />

clearly functional different cortical regions.<br />

Each ICA component was evaluated over the different<br />

time points. Mean components (for the 5 animals)<br />

were very reproducible both for measurements on the<br />

same day as measurements with one week in between.<br />

Also single subject components were similar over time.<br />

Although the differences are bigger between measurements<br />

with one week in between, indicating that there<br />

could be an influence of scanner variability and physiological<br />

state of the animal.<br />

Conclusions: This study implementing ICA on rat<br />

resting state data proves the usability of the technique<br />

for studying FC in the rat brain in a reproducible<br />

way. This paves the way to use this tool for assessing<br />

FC changes under different circumstances<br />

such as both physiological changes and pathologies.<br />

Implementing the technique in animal models<br />

can help to unravel the underlying processes.<br />

References:<br />

1. Damoiseaux JS et al (2008) Cereb Cortex. 18(8), 1856-<br />

1864.<br />

2. Greicius MD et al (2004) Proc Natl Acad Sci U S A. 101(13),<br />

4637-4642.<br />

3. Pawela CP et al (2008) Magn Reson Med. 59(5), 1021-1029.<br />

4. Beckmann CF et al (2005) Philos Trans R Soc Lond B<br />

Biol Sci. 360(1457), 1001-1013.


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

A fast and robust acquisition scheme for CEST experiments<br />

Longo D. , Cittadino E. , Terreno E. , Aime S. .<br />

University of Torino, Italy<br />

dario.longo@unito.it<br />

Introduction: Chemical Exchange Saturation Transfer<br />

(CEST) agents are a new class of MRI contrast<br />

agent based on the selective irradiation of a mobile<br />

proton pool in slow/intermediate exchange with<br />

water [1] . Long irradiation pulses at the resonant frequency<br />

(∆ω) are needed to obtain a complete saturation<br />

of the CEST pool and a correct saturation transfer<br />

(ST%) quantification requires the acquisition<br />

of many images at different frequency offsets (Zspectrum),<br />

thus making a CEST experiment a timeconsuming<br />

procedure depending on the number of<br />

sampled frequency offsets [2] . A trade-off between<br />

the number of frequency offsets and the accuracy of<br />

ST% determination has to be found in order to distinguish<br />

the characteristic ST% time course for individual<br />

voxels. The development of fast and robust<br />

methods are therefore mandatory to correctly detect<br />

CEST molecules in Molecular Imaging applications.<br />

In this work a computational procedure has been<br />

developed to evaluate the accuracy of ST% quantification<br />

as a function of the number of frequency offsets<br />

centered around the resonant frequency of the<br />

mobile proton pool with different levels of noise and<br />

of B 0 inhomogeneities. We also proposed a different<br />

acquisition procedure, by acquiring first a complete<br />

Z-spectrum followed by the sampling of a limited<br />

number of frequency offsets and then replacing<br />

these points in the first Z-spectrum, thus improving<br />

the temporal resolution of CEST contrast quantification,<br />

without sacrifying the ST% accuracy.<br />

Methods: Z-spectra were simulated by using a twopool<br />

model solving the Bloch equation (T 1A = 2 s,<br />

T 2A = 0.1 s, T 1B = 1 s, T 2B = 0.015 s, ∆ω = 4.2 ppm, k ex<br />

= 160 s -1 , 30 mM diacest agent, saturation pulse 3<br />

μT for 5 s). Gaussian noise was added both in the<br />

signal intensities and in the frequency axis and zero<br />

shifts were introduced in the range 0.1-1 ppm. Zspectra<br />

were interpolated by smoothing spline (with<br />

zero shift correction) by different number of points<br />

around the resonant frequency (with step of 0.1 ppm)<br />

both at positive (∆ω) and negative offsets (-∆ω): i) 7<br />

points, ii) 5 points, iii) 3 points, iv) 3 points with 0.2<br />

ppm step, v) 1-(∆ω/-∆ω) (ON/OFF scheme) with<br />

and vi) without zero-shift correction, compared to<br />

a full Z-spectrum in the range ±10 ppm with steps<br />

of 0.1 ppm. In in vivo CEST images were acquired<br />

on a 7 Tesla Bruker spectrometer with a complete<br />

Z-spectrum (single-shot RARE spin-echo, TR 6 s,<br />

centric encoding, MTX 64, saturation pulse: 3 μT<br />

for 5 s) followed, upon injection of a CEST agent by<br />

the dynamic acquisition of only 3 points around the<br />

resonant frequency (∆ω = 4.2 ppm) for 30 min (50<br />

scans, temporal resolution: 36 s).<br />

Results: The ON/OFF scheme with and without zero-shift<br />

correction showed the highest mean ST errors,<br />

up to 2 % and 5%, respectively and with high<br />

standard deviation values. The method iv) with 3<br />

points spaced of 0.2 ppm, in comparison with the<br />

more accurate but also time consuming i) and ii)<br />

methods, displayed mean ST errors lower than 1%,<br />

but with a drastic reduction in acquisition time. In<br />

in vivo the continous acquisition of only 3 frequency<br />

offsets around the resonant frequency and the correction<br />

for zero shifts by using the pre-contrast Zspectrum<br />

allowed to evaluate the ST% time course<br />

with a temporal resolution of few seconds and to<br />

discriminate between enhancing and not-enhancing<br />

pixels in kidneys.<br />

Conclusions: The acquisition of 3 points spaced of 0.2<br />

ppm around the resonant frequency of the mobile proton<br />

pool resulted to be sufficient enough to provide a robust<br />

and accurate ST% quantification, with a fast temporal<br />

resolution by using a modified Z-spectrum acquisition.<br />

Acknowledgement: Economic support from regional<br />

government ( NanoIGT project - CIPE 2007) and<br />

EC-FP7 project ( ENCITE: FP7-HEALTH-2007A).<br />

References:<br />

1. Ward KM, et al.; J Magn Reson. 2000; 143: 79<br />

2. Liu G, et al.; Magn Reson Med. 2009; 61: 399<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

P-100<br />

poStEr<br />

MI DATA ANALYSIS METHODS


214<br />

WarSaW, poland May 26 – 29, 2010<br />

P-101 Binding potential estimation in 11C-PE2I PET brain striatal images: impact of partial volume<br />

correction under segmentation errors<br />

Maroy R. , Dusch E. , Comtat C. , Leroy C. , Trebossen R. .<br />

CEA, Orsay, France, Metropolitan<br />

renaud.maroy@cea.fr<br />

Introduction: Numerous methods have been proposed<br />

for the correction of the Partial Volume Effect<br />

(PVE) that hampers striatal PET studies. While<br />

their quantification accuracy is known for most,<br />

their ability to recover correct pharmacokinetic parameters<br />

in case of segmentation errors is less clear.<br />

The work proposes to compare the binding potential<br />

estimations using 4 distinct Time Activity Curve<br />

(TAC) estimation methods.<br />

Methods: A phantom based on the Zubal phantom<br />

of a 11C-PE-2I PET exam with kinetics generated<br />

using the simplified tissue model of Lammertsmaa 1<br />

(R1=1.28, k2=.09, BP=17) was analytically simulated<br />

on a ECAT HRRT (Siemens, 2.4mm intrinsic<br />

resolution). PET images were reconstructed with<br />

1.2�1.2�1.2mm 3 voxels and 20 frames using both<br />

RM-OP-OSEM 2 , which compensate for PVE, and<br />

OP-OSEM.<br />

Twenty realizations of caudate segmentations containing<br />

errors ranging between 40% volume underestimation<br />

(-40%) and 40% volume overestimation<br />

imaging life<br />

(40%) were defined 3 . Four TAC estimation methods<br />

were compared for the caudate: the mean TAC measurement<br />

in the standard OP-OSEM image (ME) and<br />

in the RM-OP-OSEM image (MRO), the Geometric<br />

Transfer Matrix method (GTM) and an improved<br />

GTM 3 method using voxel selection (GTM20). The<br />

R1, k2 and binding potential (BP) estimated with<br />

the estimated TACs using 1 were compared based<br />

on the Apparent Recovery Coefficient (ARC).<br />

Results: Unlike k2 and R1,<br />

the BP is globally underestimated<br />

by the methods.<br />

The lowest performances<br />

are obtained without correction,<br />

using ME. The<br />

best ARC for the TACs (Fig<br />

1.A) and R1 (Fig 1.C) are<br />

achieved using MRO, but<br />

the use of MRO leads to<br />

suboptimal BP (Fig 1.D),<br />

which is the model parameter<br />

of major interest. The<br />

best ARC for BP and k2<br />

(Fig 1.B) are achieved using<br />

GTM20, which provides<br />

better TACs than GTM,<br />

leading also to better ARC<br />

than GTM.<br />

Conclusions: Our improved GTM method with voxel<br />

selection achieves the TAC estimation in terms of<br />

binding potential and k2 recovery.<br />

References:<br />

1. A A Lammertsma et al. Neuroimage, 1996<br />

2. F C Sureau et al. JNM, 2008<br />

3. R Maroy et al. SNM, 2009


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

Automated quantification scheme based on an adapted probabilistic atlas based<br />

segmentation of the brain basal nuclei using hierarchical structure-wise registration<br />

Maroy R. (1) , Leroy C. (1) , Douaud G. (2) , Trebossen R. (1) .<br />

(1) CEA, Orsay, France, Metropolitan<br />

(2) Radcliffe Hospital, Oxford, United Kingdom<br />

renaud.maroy@cea.fr<br />

Introduction: The work<br />

proposes: 1) a new basal<br />

ganglia segmentation<br />

method based on probabilistic<br />

atlas construction<br />

and compared to<br />

manual segmentation,<br />

2) a validation method<br />

for segmentation of structures<br />

using a co-acquired<br />

PET image.<br />

Methods: T1 MRI (T1) and<br />

18F-PEII PET images were<br />

acquired in 12 healthy<br />

adults and co-registered.<br />

The PET images, acquired<br />

on the HRRT PET system,<br />

were reconstructed with<br />

both the AWOSEM (NoPSF) and the 3D-OP-<br />

OSEM PSF (PSF) methods. The basal nuclei were<br />

segmented manually in a knowledge base (KB) of<br />

24 T1 and in the 12 acquired subjects T1 (IS). A<br />

probabilistic atlas adapted to IS was constructed<br />

using a structure-wise hierarchical registration of<br />

each KB image on IS. Segmentation was obtained<br />

simply through atlas probability maximization.<br />

A GTM correction was applied on the NoPSF<br />

using as spatial domain 1) the manually drawn<br />

structures or 2) the automatically drawn structures,<br />

with A) ROI defined as in [Frouin, 2002]<br />

or B) with adequate selection of 20% (GTM20)<br />

of the corresponding spatial domain voxels.<br />

The estimated values were compared for 1)+A)<br />

and 2)+B) to the mean TAC in PSF, considered<br />

as the reference, inside manually drawn<br />

eroded regions using the estimation error (E).<br />

Results: GTM20 performed significantly better<br />

than GTM for both the manual and automated<br />

delineation. Besides, the gain using together the<br />

automated segmentation and GTM20 (E1)+A)/<br />

E2)+B)) was 6.1 for the caudate, 3.6 for the globus<br />

pallidus, 4.6 for the putamen and 1.4 for the<br />

thalamus for an increased reproducibility.<br />

Conclusions: We have proposed a quantification<br />

scheme using an adapted probabilistic atlas based<br />

segmentation of the basal nuclei and a partial volume<br />

correction method that enhances both the<br />

precision and the reproducibility of the PVE corrected<br />

measures.<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

P-102<br />

poStEr<br />

MI DATA ANALYSIS METHODS


216<br />

WarSaW, poland May 26 – 29, 2010<br />

P-103 VHISTdiff - comparing workflow histories (VHIST/VINCI)<br />

Vollmar S. , Hüsgen A. , Sué M. , Nock J. , Krais R. .<br />

Max Planck Institut for Neurological Research, Köln, Germany<br />

vollmar@nf.mpg.de<br />

Introduction: VHIST [1] has been developed to<br />

document workflows in multi-modality imaging<br />

(however, it is not at all limited to this field). The<br />

VHIST file format is PDF compatible and it can be<br />

displayed with any PDF viewer (“human readable”).<br />

However, VHIST files also contain structured information<br />

on each workflow step (embedded XML)<br />

suitable for automated processing. In one current<br />

project, we are integrating VHIST into VINCI [2],<br />

our package for visualization and analysis of tomographical<br />

data. This enables us to natively create<br />

VHIST files which can store meta data of original<br />

file formats, filter operations, “image arithmetics”<br />

and registration parameters.<br />

Methods: An interesting application of VHIST files<br />

is the comparison of workflow histories belonging<br />

to similar but subtly different image data, e.g. image<br />

data of one subject which has been aquired by<br />

the same scanner but which has been processed in a<br />

different way (type and order of filter steps; thresholding;<br />

application of masks; different types of normalization,<br />

different reconstruction parameters;<br />

different approaches when converting raw data). For<br />

this kind of comparison we are currently developing<br />

a new tool, VHISTdiff, which allows to automatically<br />

find corresponding workflow steps and report<br />

the differences.<br />

The figure depicts a graphical representation of the<br />

“macroscopic” differences between two VHIST files<br />

(A.vhist, B.vhist) as they were found by the VHISTdiff<br />

algorithm: each tree represents the workflow<br />

history of one image. Dotted lines denote corresponding<br />

workflow steps, white text marks workflow<br />

steps that do not have a pendant. VHIST files<br />

can contain other VHIST files: in this example, the<br />

registration step of the A-tree contains file C.vhist,<br />

and likewise D.vhist is contained in the other registration<br />

step. The report of differences between<br />

corresponding workflow steps (e.g. different filter<br />

parameters) are not part of this particular graphical<br />

representation which has been rendered fully automatically<br />

using Graphviz [3].<br />

VHISTdiff ’s algorithm uses some ideas of the traditional<br />

UNIX diff command for comparison of text<br />

imaging life<br />

files. However, the concept has been generalized to<br />

tree structures: VHISTdiff uses Dijkstra’s algorithm<br />

[4] to optimize a cost function which favours association<br />

strings of similar workflow steps.<br />

Outlook: First results of this new tool look promising.<br />

We will investigate strategies to define cost<br />

functions that try to be useful for a wide range of<br />

real-world use cases.<br />

References:<br />

1. VHIST, http://www.nf.mpg.de/vhist<br />

2. VINCI, http://www.nf.mpg.de/vinci<br />

3. The Graphviz software package, http://www.graphviz.org<br />

4. Dijkstra, E. W. (1959). “A note on two problems in<br />

connexion with graphs”. Numerische Mathematik 1:<br />

269–271


<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

Fast matrix-free method for fluorescence imaging<br />

Zacharopoulos A. (1) , Garofalakis A. (2) , Ripoll J. (1) , Arridge S. (3) .<br />

(1) FORTH IESL , Heraklion, Greece<br />

(2) CEA, I2BM , France<br />

(3) UCL CS, United Kingdom<br />

azacharo@iesl.forth.gr<br />

Introduction: In non-contact Fluorescence Molecular<br />

Tomography, the large number of measurements<br />

poses a challenge since the reconstruction methods<br />

used rely on the inversion of a derivative operator<br />

and the explicit formulation and storage of this operator<br />

in a matrix is generally<br />

not feasible. We test a matrixfree<br />

method that addresses the<br />

problems of large data sets and<br />

reduces the computational cost<br />

and memory requirements<br />

for the reconstructions. More<br />

specifically we challenged<br />

the Matrix-Free method with<br />

in-vivo measurements from<br />

mice where fluorescence tubes<br />

of different but controlled<br />

concentrations are inserted,<br />

to assess the quantification<br />

performance of the method.<br />

Methods: Our approach initially presented in [1], is<br />

based on a formulation of the diffusion approximation<br />

for the fluorescence case and the TOAST Image<br />

Reconstruction in Optical Tomography FEM<br />

package, [2] and uses a matrix free formulation.<br />

The explicit calculation and storage of the Jacobian<br />

is avoided by replacing it by a vector times matrix<br />

operator and a vector times adjoint matrix operator.<br />

Since we wanted to assess the quantification properties<br />

of the matrix-free method we aquired in-vivo<br />

measurements using a balb/c mouse where capillary<br />

of Alexa Fluor 647 of controlled concentrations<br />

a)60.5uM, b)22uM, c)8.5uM, d)2.2uM and e)0.9 uM.<br />

was inserted hypodermically in the ventral side. For<br />

the reconstructions, a slab-like geometry of dimensions<br />

41mm x 41mm x 12mm was used with 5733<br />

nodes and 4800 voxel-elements. For the optical<br />

parameters we assumed μa =0.06 mm-1 and μs =1.<br />

mm-1 for the 615nm excitation and μa =0.05 mm-1<br />

and μs =1.5 mm-1 for the 700nm emission wavelengths.<br />

We set 1458 detectors positions by sampling<br />

the images, which for the 36 sources created<br />

two vectors of 48285 measurements each.<br />

Results: The results for the five different experiments<br />

are presented as iso-surfaces in 3D in the<br />

figure where the white light image of the mouse<br />

is rendered in the background as a reference. The<br />

average time for the reconstruction was 50sec, in a<br />

Pentium 2Ghz machine with 2GB of memory.<br />

Conclusions: Using the Matrix-free method we<br />

managed to reconstruct for the positions of the<br />

fluorescent tubes and a relative quantification accuracy<br />

and sensitivity. More specifically the algorithm<br />

demonstrated an almost linear response between<br />

the controlled concentrations and the reconstructed<br />

ones.<br />

Acknowledgement: This work was funded by the<br />

EC Seventh Framework Grant, FMT-XCT, grant<br />

201792.<br />

References:<br />

1. Zacharopoulos A, et al (2009) Opt. Express , 17, 3042-<br />

-3051.<br />

2. Arridge R S (1999) Inverse Problems, 15, 41--93 .<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

P-104<br />

poStEr<br />

MI DATA ANALYSIS METHODS


P-105<br />

218<br />

WarSaW, poland May 26 – 29, 2010<br />

TOPIC: Cancer Biology<br />

PET and MRI studies applied on characterization of Fisher/F98 rat glioma model<br />

Valtorta S. (1) , Ronchetti F. (2) , Lo Dico A. (3) , Politi L.S. (4) , Masiello V. (5) , Matarrese M. (5) , Zara G. (6) , Zenga F. (7) , Scotti G. (4) ,<br />

Mauro A. (2) , Moresco R.M. (5) .<br />

(1) Università degli Studi di Milano; IBFM-CNR, Segrate, Italy<br />

(2) Istituto Auxologico Italiano, Italy<br />

(3) San Raffaele Scientific Institute; IBFM-CNR; L.A.T.O., Italy;<br />

(4) San Raffaele Scientific Institute, Italy;<br />

(5) San Raffaele Scientific Institute; IBFM-CNR; Università degli Studi di Milano-Bicocca, Italy;<br />

(6) Università di Torino, Italy;<br />

(7) Università di Brescia, Italy<br />

silvia.valtorta@unimi.it<br />

Introduction: Preclinical brain tumor models<br />

have provided a wealth of information on the<br />

biology, imaging and experimental therapeutics<br />

of brain tumors [1],[2] . The aim of our study is<br />

characterized Fisher/F98 rat glioma model using<br />

Positron Emission Tomography (PET) and<br />

Magnetic Resonance (MR) analysis to set up an<br />

experimental model useful to study the efficacy<br />

of new colloidal vectors for chemotherapy.<br />

Methods: Syngenic rat brain-glioma models<br />

(Fisher/F98) was obtained by stereotactic (x=2;<br />

y=5; z=3) implantation of different cell concentrations<br />

(10 2 , 10 3 , 10 4 and 10 5 ). To monitor tumor<br />

growth progression, rats underwent once a week<br />

Gadolinium enhanced T1-MRI studies followed<br />

by [ 18 F]FDG PET studies, starting from 7 days<br />

after surgery. A group of animals performed also<br />

[ 18 F]FAZA PET studies to evaluate regional tissue<br />

hypoxia. To improve quantification, PET and<br />

MRI images were fused using PMOD 2.7 software.<br />

Max radiotracers uptake was calculated for tumor,<br />

frontal cortex, cerebellum and background using<br />

region of interest (ROI) analysis. Radioactivity<br />

concentration values expressed in MBq/g were<br />

then transformed into percentage of injected dose<br />

per gram of tissue (%ID/g). Moreover, histological<br />

analysis of proliferation, apoptosis, differentiation,<br />

neoangiogenesis and hypoxia markers were<br />

performed.<br />

Results: Mean survival time of rats injected with<br />

10 4 and 10 5 cells was nine days. One week after<br />

surgery, MRI revealed a rapid growth that reached<br />

0.11 cm 3 mean tumour volume. Animals injected<br />

with 10 3 and 10 2 cells showed a mean survival time<br />

of 18 and 24 days respectively. In rats injected with<br />

10 3 cells, tumor was revealed 14 days after surgery<br />

at MRI and [ 18 F]FDG PET and successively tumors<br />

rapidly increased. Disease course in 10 2 cells<br />

injected rats was slower. Tumors were characterized<br />

by high [ 18 F]FDG uptake and hypoxic subareas<br />

which only partially overlapped. Hypoxic<br />

imaging life<br />

areas were mainly localized in correspondence<br />

to Gd-enhanced regions whereas hyper glucose<br />

metabolic areas were localized in the outer part<br />

of the tumors. At histological analysis tumoral<br />

masses showed an infiltrative pattern of growth<br />

and moderate neoangiogenesis. Tumors obtained<br />

at animals death showed diffused necrotic areas.<br />

HIF1 was clearly expressed by glial and neuronal<br />

cells in oedematous and hypoxic areas.<br />

Conclusions: Our study indicates that Fisher/<br />

F98 rat glioma model reproduce s the characteristic<br />

of aggressiveness of human glioblastoma.<br />

Tumor was characterized by high glucose<br />

metabolism and by hypoxic sub-areas. The concentration<br />

of 10 2 cells permits to better monitor<br />

disease onset and progression and to plan<br />

experiments to test new anti-neoplastic therapies<br />

efficacy.<br />

Acknowledgement: This work was supported in part<br />

by grants from: Regional AIRC 2008 n°6278 and<br />

FIRB-CNR n°RBIP06M8ZA.<br />

References:<br />

1. Spaeth N et al; Eur J Nucl Med Mol Imaging. 33:673-<br />

682 (2006)<br />

2. Brioschi AM et al; J Nanoneurosci. 1:65-74 (2009)


TOPIC: Cancer Biology<br />

<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

Non-invasive “E2F sensing” system for monitoring DNA damage alteration induced by BCNU<br />

Monfared P. (1) , Rudan D. (1) , Viel T. (1) , Waerzeggers Y. (1) ,Hadamitzky M. (1) , Schneider G. (1) , Rapic S. (1) ,Neumaier B. (1) , Backes H. (1) ,<br />

Winkeler A. (1) , Jacobs A.H.. (1,2).<br />

(1) Max Planck Institute for Neurological Research, Cologne,<br />

(2) European Institute for Molecular Imaging (EIMI), University of Muenster, Germany<br />

Parisa.Monfared@nf.mpg.de<br />

Introduction: Imaging transcriptional regulation of<br />

endogenous genes in living animals using noninvasive<br />

imaging techniques provide a clear perception<br />

of normal and cancer-related biological processes.<br />

Radiolabeled reporter probes and PET imaging can<br />

be translated into human studies in the near future.<br />

In our preliminary study, the endogenous expression<br />

of E2F, a gene that affects several important<br />

biological processes, has been imaged in vivo with<br />

bioluminescence imaging. A retroviral vector, Cis-<br />

E2F/LUC-IRES-TKEGFP, was generated by placing<br />

the reporter genes under control of an artificial<br />

cis-acting E2F-specific enhancer element. Following<br />

retroviral transduction of tumor cells in established<br />

xenografts, DNA damage induced alteration<br />

of E2F transcriptional activity, which correlated<br />

with the expression of E2F-dependent downstream<br />

genes as assessed by biolumencsnce imaging.<br />

Aim: To verify whether the cis-reporter system<br />

(Cis-E2F/LUC-IRES-TKEGFP) is sufficiently sensitive<br />

to image endogenous transcriptional gene<br />

regulation by [ 18 F] FHBG PET imaging.<br />

Methods: U87dEGFR-E2F-LITG cells were injected<br />

subcutaneously in nude mice and the<br />

development of the tumours was followed by<br />

Multimodal imaging. Two weeks after implantation,<br />

bioluminescence imaging, FLT and<br />

FHBG were performed before and 24h after<br />

treatment with low and high doses of BCNU.<br />

Results: Here, we validate the utility of [ 18 F]FLT-<br />

PET to image proliferation rate of the tumor in response<br />

to DNA damage and compare with [ 18 F]<br />

FHBG uptake which is associated with E2F transcriptional<br />

activity. In keeping with in vitro findings,<br />

after 24 h post-treatment, low dose of BCNU<br />

induced an increase in [ 18 F]FHBG uptake as compared<br />

to non-treated mice. However, with high<br />

dose of BCNU [ 18 F]FHBG uptake decreased in E2F<br />

xenografts. [ 18 F]FLT accumulation in E2F xenografts<br />

decreased with low and high doses of BCNU<br />

at 24 and 48 hours. Quantitative changes in tumor<br />

[ 18 F]FLT uptake were associated with decreased<br />

tumor proliferation and tumor [ 18 F]FHBG uptake<br />

correlated with transcriptional gene regulation of<br />

E2F in response to DNA damage<br />

Conclusions: We show the utility of [ 18 F]FHBG-<br />

PET to image DNA damage induced by BCNU and<br />

shown the tumor-specific activity of E2F. We propose<br />

that these types of reporter systems, will allow<br />

a detailed insight into the kinetics of cell cycle<br />

control and for the development of new cell cycle<br />

targeted molecular therapies.<br />

Acknowledgement: This work is supported in part<br />

by the EC-FP6 European DiMI, (LSHC-CT-2004-<br />

503569) LSHB-CT-2005-512146 and Clinigene<br />

(LSHB-CT-06-018933).<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

P-106<br />

poStEr<br />

LATE BREAKING


P-107<br />

220<br />

WarSaW, poland May 26 – 29, 2010<br />

TOPIC: Neuroimaging<br />

Multi-tracer PET imaging of a mouse model of Alzheimer´s disease to assess microglial<br />

activation related to ageing and anti-inflammatory treatment<br />

Rapic S. (1) , Backes H. (1) , Viel T. (1) , Hadamitzky M. (1) , Monfared P. (1) , Rudan D. (1) , Vollmar S. (1) , Neumaier B. (1) , Hoehn M. (1) , Van der<br />

Linden A. (2) , Heneka M.T. (3) , Jacobs A.H. (1,4)<br />

(1) Max Plank Institute for Neurological Research, Cologne, Germany<br />

(2) Bio-Imaging Lab, University of Antwerp, Belgium<br />

(3) Department of Neurology, University of Bonn, Germany<br />

(4) European Institute for Molecular Imaging (EIMI)<br />

Sara.Rapic@nf.mpg.de<br />

Introduction: Alzheimer´s disease (AD), the leading<br />

cause of dementia in elderly people, is characterized<br />

by the deposition amyloid-β plaques in the<br />

brain surrounded by activated microglia. Activated<br />

microglia play an important role in amyloid clearance<br />

but also secrete pro-inflammatory molecules<br />

that lead to chronic neuroinflammation, and toxins<br />

that trigger cell death cascades in neurons, both<br />

eventually resulting in neurodegeneration. The<br />

nuclear peroxisome proliferator-activated receptor<br />

gamma (PPAR-γ) plays an important role in inflammatory<br />

processes. Agonists that bind and activate<br />

PPAR-γ, suppress inflammation by inhibiting the<br />

expression of pro-inflammatory molecules at transcriptional<br />

level. Determination of the contribution<br />

of neuroinflammation to the progression of AD can<br />

provide new insights in the development of therapy.<br />

Aim: To assess with in vivo positron emission tomography<br />

(PET) whether microglial activation is correlated<br />

with age in APP/PS1 transgenic mice and wildtype<br />

mice and if anti-inflammatory treatment with a<br />

PPAR-γ agonist can reduce neuroinflammation.<br />

Methods: Multi-tracer micro-PET imaging was performed<br />

in 13 months old APP Swe /PS1 dE9 transgenic<br />

mice (n = 5) and C57BL/6J wild type mice (n = 7).<br />

Dynamic imaging was performed after intravenous<br />

injection of [ 11 C]PK11195, a radioligand for the<br />

translocator protein-18 kDa which is upregulated in<br />

activated microglia and [ 18 F]FDG for the quantification<br />

of cerebral glucose metabolic rate. [ 18 F]Fluoride<br />

bone imaging was performed after every scan for coregistration<br />

purposes. The animals were imaged at<br />

baseline and treated for 4 weeks with a PPAR-γ agonist.<br />

Right after treatment and 4 weeks off treatment,<br />

all animals were investigated again using the same<br />

imaging protocol to assess the effect of the therapy.<br />

Results: Preliminary results show that uptake of<br />

[ 11 C]PK11195 is upregulated in transgenic mice<br />

compared to wild-type mice in the cortex (SUV<br />

of 75.15 and 57.20 respectively), thalamus (SUV<br />

of 78.90 and 63.63 respectively) and cerebellum<br />

(SUV of 82.12 and 65.47 respectively). Uptake of<br />

imaging life<br />

[ 18 F]FDG in transgenic mice is elevated as well,<br />

compared to wild-type mice, in the cortex (SUV<br />

of 162.10 and 106.78 respectively), thalamus (SUV<br />

of 172.07 and 115.33 respectively) and cerebellum<br />

(SUV of 166.50 and 114.46 respectively). Data were<br />

corrected for the total injected dose (ID). First image<br />

processing of the data obtained after treatment<br />

shows decreased brain retention of both tracers in<br />

transgenic mice.<br />

Conclusions: The higher uptake of [ 11 C]PK11195 in<br />

the brain of transgenic mice compared to wild-type<br />

mice indicates the presence of activated microglia,<br />

suggesting neuroinflammation of the Alzheimer<br />

brain. Reduction of [ 11 C]PK11195 brain retention<br />

after treatment with the PPAR-γ agonist reflects its<br />

anti-inflammatory effect.<br />

With the help of animal models for Alzheimer’s disease<br />

and in vivo molecular imaging, longitudinal detection of<br />

microglial cells is made possible and may be of vital importance<br />

in defining their role in the progression of the<br />

disease and in the development of therapeutic strategies.<br />

Acknowledgement: This work is supported in part by<br />

EC-FP6-project and DiMI LSHB-CT-2005-512146.


��������� ����� ������������� �������� ���������� ���� �����<br />

������������������������������������������������������<br />

������������������������������������������������������������<br />

�������� ���� ���������� ��������� ������������� �����<br />

���������������<br />

�<br />

• ������������������������������������������������������������<br />

��������������������������<br />

• ����������������������������������������������������������<br />

����������������������������������<br />

• ��������������������������������������������������<br />

• ������� ����� ��������� ����� ���� ������� �������� �����<br />

����������������������<br />

• ������������������������������������������������������<br />

• ���� ����������� ��������� �������� ����������� ��� ���������<br />

������������������������<br />

�<br />

��������������<br />

���������������������������������������������������������������������<br />

����������������������������������������������������������������������������������<br />

���������������������������������������<br />

�������������������������������������������������������������������������������<br />

���<br />

����<br />

����


index


INDEX


ANIMASCOPE<br />

High TECHNOLOGY for High TRANSPARENCY<br />

• High Technology for High Transparency<br />

o Non invasive imaging technologies<br />

o Small animals Preclinical imaging<br />

o Multimodality imaging services (Ultrason, MRI, Optical, PET, SPECT, CT Scan)<br />

• Reduce, Refine, Replace<br />

o Quantitative, Precise, Predictive<br />

o Reduces experimental Artifacts<br />

o Longitudinal Studies - Reduce Animal Usage (minus 70-80%)<br />

o Reduction and Refinement of animal works are Cost and Time-effective on R&D<br />

• Bridge the gap between preclinical and clinical research<br />

o Drug Delivery and Liberation (drug to target)<br />

o Efficacy, Safety, Pathology (desiderable and undesirable effects)<br />

o Ligand-Target interaction (receptors, enzymes, transporters) including Dose<br />

Occupancy relationships<br />

o Dynamic Biodistribution and Kinetics<br />

• One stop shop for :<br />

o biology,<br />

o cardiovascular,<br />

o cell therapy, gene therapy, immunization,<br />

o musculoskeletal,<br />

o neurology, nephrology,<br />

o oncology,<br />

o reproduction, development,<br />

o nutrition (nutraceuticals, dietary supplements and<br />

functional food).<br />

Contact : Daniel CHRISTIAEN<br />

ZI EUREKALP 38660 St Vincent de Mercuze (F)<br />

d.christiaen@animascope.eu<br />

Tel +33 (0)476 97 94 87<br />

www.animascope.eu<br />

Give to your study a new look


Index<br />

A<br />

Abasolo I. 126<br />

Abiraj K. 77<br />

Aboagye E.O. 209<br />

Adamczak J. 64<br />

Aelvoet S. A. 63<br />

Agarwal A. 41<br />

Agin V. 148<br />

Ahmad R. 204<br />

Aigner L. 62, 81, 169<br />

Aime S. 49, 79, 114,<br />

171,174, 176, 213<br />

Allard S. 90<br />

Alves F. 128, 208<br />

Anderson S. 155<br />

Arena F. 49<br />

Arridge S. 217<br />

Arstad E. 204<br />

Ashitate Y. 55<br />

Aswendt M. 171<br />

Audrain H. 189<br />

Autio A. 54, 130<br />

Avory M. 204<br />

Awde A. 160<br />

B<br />

Baatenburg De Jong R.J.<br />

119<br />

Backes H. 134, 219, 220<br />

Baekelandt V. 63, 85<br />

Barnard P. 77<br />

Barré L. 78<br />

Barré W. 35<br />

Barrio J. 157<br />

Bauernfeind A. 155<br />

Baulieu J. L. 157<br />

Bauwens M. 116<br />

Beaufils E. 157<br />

Bednar B. 89<br />

Behe M. 117<br />

Béhé M. 52<br />

Bellard E. 127<br />

Bender D. 189<br />

Benoit D. 155<br />

Benyó Z. 37<br />

Berardi F. 151<br />

Bergner F. 190<br />

Bergström J. P. 78<br />

Bernhardt P. 52<br />

Bernsen M. 118, 163,<br />

166,167, 168<br />

Berthezene Y. 65<br />

Berti R.P. 172<br />

Beynel A. 191<br />

Bhalla R. 183<br />

Bijster-Marchand M. 118<br />

Bilski M. 192<br />

Biserni A. 90, 91<br />

<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

Blanc A. 53<br />

Blankevoort V. 158<br />

Blockx I. 169<br />

Blossier A. 197<br />

Boehm-Sturm P. 64<br />

Bogaert-Buchmann A. 147<br />

Bogner W. 115<br />

Bohmer M. 103<br />

Boisgard R. 152, 160,<br />

191,197, 202, 211<br />

Bonetto F. 161<br />

Bonnet C. 182<br />

Bonzom S. 43<br />

Borelli M. 84<br />

Bormans G. 85, 170<br />

Boschi F. 42<br />

Bos P. 167<br />

Botnar R. 97<br />

Botta M. 48<br />

Boussel L. 68<br />

Boutin H. 198<br />

Brepoels L. 30<br />

Brown G. 198<br />

Brüggemann C. 171<br />

Brzezinski K. 184<br />

Bukala D. 201<br />

Burdinski D. 44<br />

Buron F. 182<br />

Busch C. 199<br />

Bzyl J. 72, 122<br />

C<br />

Cabello J. 184<br />

Calandrino R. 42<br />

Calderan L. 42<br />

Calotti F. 120<br />

Camus V. 157<br />

Cannet E. 68<br />

Carelli S. 56<br />

Carlsen H. 100<br />

Carpenter A. 36<br />

Carroll X. 155<br />

Casteels C. 33<br />

Catanzaro V. 79, 176<br />

Caveliers V. 29, 136<br />

Cemazar M. 127<br />

Cescato R. 206<br />

Céspedes M. V. 126<br />

Chapotot L. 191<br />

Chattopadhyay A. 151<br />

Chauveau F. 65<br />

Chazalviel L. 148<br />

Cheung P. 164<br />

Choe J. G. 153<br />

Choi H. S. 55<br />

Cho T. H. 65<br />

Christofori G. 52<br />

Ciana P. 90, 91<br />

Cibiel A. 173<br />

Ciobanu L. 160<br />

Cittadino E. 79, 114,<br />

174,213<br />

Clark C. 36<br />

Clerici M. 84, 165<br />

Clerici M. S. 56<br />

Cojoca R. 205<br />

Coleman P. 89<br />

Coll J. L. 139<br />

Comtat C. 214<br />

Contag C.H. 21<br />

Cordula D. 140<br />

Cottet M. 135<br />

Couillaud F. 83<br />

Crosby 61<br />

Cruz Ricondo L. J. 161<br />

Cuhlmann S. 100<br />

Curtis A. 41<br />

Custers E. 73<br />

D<br />

D´Alessandria C. 87<br />

D’Ambrosio D. 42<br />

Damont A. 197, 202<br />

Dastrù W. 114<br />

Debeissat C. 83<br />

Debyser Z. 63, 85<br />

Deckers N. 116<br />

Decristoforo C. 178<br />

Defraiteur C. 149<br />

De Geeter F. 101<br />

Degrassi A. 84<br />

Degroot T. 30<br />

De Jong M. 104, 107, 118<br />

Delest B. 180<br />

Deliolanis N. 185, 188<br />

Delli Castelli D. 114<br />

Delpassand E. S. 105<br />

Del Puerto-Nevado L. 203<br />

Del Vecchio S. 143<br />

Delzescaux T. 211<br />

Demello A. 189<br />

Demmer O. 87<br />

Demphel S. 202<br />

Denes A. 198<br />

De Poel M. 118<br />

De Rooij K. 142<br />

Deroose C. 85, 170<br />

De Saint-Hubert M. 30,116<br />

Desco M. 177, 181<br />

De Smet M. 103<br />

De Spirito M. 121<br />

De Swart J. 118<br />

De Vocht N. 57<br />

Devoogdt N. 29, 136<br />

Devos E. 30, 116<br />

Devos H. 101<br />

De Vries A. 73<br />

De Vries I. J. 161<br />

D’huyvetter M. 136<br />

Díaz-Lópeza R. 172<br />

Diependaele A. S. 148<br />

Dietlmeier J. 210<br />

Digilio G. 79, 176<br />

Dijkgraaf I. 87<br />

Dikaiou K. 186, 196<br />

Dilworth J. 77<br />

Doeswijk G. 166<br />

Dollé F. 160, 187, 197<br />

202,211<br />

Douaud G. 215<br />

Douek P. 68<br />

Drake C. 198<br />

Dresch C. 82<br />

Dubois A. 160, 187, 211<br />

Ducongé F. 173, 187<br />

Dullin C. 128, 208<br />

Dumont R. 117, 206<br />

Duong L. T. 89<br />

Dupont D. 147, 187<br />

Durroux T. 135<br />

Dusch E. 214<br />

Duyckaerts C. 179<br />

Dziuk E. 195<br />

Dziuk M. 192<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

E<br />

Ehrlichmann W. 201<br />

Ell P. J. 52<br />

Emond P. 78<br />

Emonds K. 137<br />

Etard O. 148<br />

Euskirchen P. 123<br />

Evans P. 100<br />

Evens N. 85<br />

F<br />

Fani M. 77, 117, 178, 206<br />

Farde L. 75<br />

Farrell E. 167<br />

Farr T. D. 64<br />

Fattal E. 172<br />

Fedeli F. 79<br />

Fekete K. 37<br />

Fekete M. 48<br />

Fenzi A. 42<br />

Fernández Y. 126<br />

Figdor C. 161<br />

Figueiredo S. 174<br />

Finnema S. 75<br />

Fiorini C. 84, 165<br />

Fischer K. 200<br />

Flamen P. 138<br />

Fokong S. 122, 175<br />

Forni G. 205<br />

IndEx


226<br />

WarSaW, poland May 26 – 29, 2010<br />

Fortin P. Y. 83<br />

Franci X. 149<br />

Frangioni J. 55<br />

Freret L. 172<br />

Fuchs K. 200<br />

G<br />

Gadella T.W.J. 60, 61<br />

Gaetjens J. 122<br />

Garcia C. 138<br />

Garcia E. 53<br />

Garnuszek P. 70<br />

Garofalakis A. 187, 217<br />

Gätjens J. 175<br />

Gauberti M. 66, 145, 148<br />

Gee A. 189<br />

Genevois C. 83<br />

Geraldes C. 179<br />

Geraldes C.F. 174<br />

Gerhard A. 32<br />

Gervais P. 152<br />

Geys R. 159<br />

Ghanem G. 138<br />

Gianolio E. 49, 79, 171<br />

Gillet B. 147<br />

Giron-Martinez A. 203<br />

Giżewska A. 195<br />

Glaser M. 209<br />

Glatz J. 188<br />

Gleason A. 89<br />

Glowa B. 70, 146<br />

Glowalla A. 144<br />

Goblet D. 129, 149<br />

Goedhart J. 61<br />

Goethals L. 101<br />

Golzio M. 127<br />

Gombert K. 173<br />

Gonzalez-Mangado N. 203<br />

Gorio A. 56<br />

Gourand F. 78<br />

Grabner G. 115<br />

Gremse F. 72<br />

Griessinger C. 178<br />

Grießinger C. 200<br />

Griessinger C.M. 201<br />

Gringeri C. 176<br />

Groetzinger C. 140<br />

Grote M. 131<br />

Grouls C. 122<br />

Gruber S. 115<br />

Gruell H. 44, 47, 73, 103<br />

Gruetter R. 46<br />

Gsell W. 100<br />

Guedin P. 148<br />

Guenoun J. 166<br />

Guglielmetti C. 162<br />

Guilloteau D. 76, 78, 157<br />

Gulyas B. 78<br />

imaging life<br />

H<br />

Haberkorn U. 102<br />

Hadamitzky M. 123, 134,<br />

219, 220<br />

Haeck J. 118<br />

Haiat G. 172<br />

Halldin C. 75, 78<br />

Hammers A. 156<br />

Hamm J. 205<br />

Hamplova A. 179<br />

Harlaar N. J. 99<br />

Hartung F. 208<br />

Haskard D. 100<br />

Hasselbalch S. G. 35<br />

Haubner R. 117, 178<br />

Heckemann R. A. 156<br />

Heerschap 31<br />

Heerschap A. 161<br />

Heijman E. 103<br />

Helbich T. 115<br />

Heneka M.T. 220<br />

Hennink W. 132<br />

Herance J. R. 126<br />

Hérard A. S. 211<br />

Herholz K. 198<br />

Hernandez L. 43<br />

Hernot S. 136<br />

Herranz F. 177, 181<br />

Herzog E. 99<br />

Higuchi T. 97<br />

Hijnen N. 103<br />

Hilger I. 199<br />

Hillebrands J. L. 99<br />

Himmelreich U. 171<br />

Hink M.A. 61<br />

Hirani E. 204<br />

Hoeben R. 120<br />

Hoehn M. 64, 171, 220<br />

Hogset A. 49<br />

Hommet C. 157<br />

Honer M. 196<br />

Horváth I. 37<br />

Houston G. 168<br />

Hubalewska-Dydejczyk A.<br />

70, 146<br />

Hüsgen A. 216<br />

Huskens J. 44<br />

Hutteman M. 55, 142<br />

Hwang Y. M. 153<br />

I<br />

Ibrahimi A. 30, 63, 85<br />

Iezzi M. 205<br />

Iommelli F. 143<br />

J<br />

Jacobs A.H. 123, 134,<br />

219, 220<br />

Jafurulla M. 151<br />

Jalkanen S. 54<br />

Jamous M. 206<br />

Janssens I. 173<br />

Jaron A. 154<br />

Jassens I. 187<br />

Jego B. 160, 197, 211<br />

Jikeli J. 64<br />

Jiskoot W. 132<br />

Jonckers E. 212<br />

Jones H. 100<br />

Jones P. 204<br />

Joshi A. 36<br />

Josserand V. 139, 207<br />

Jucker M. 34<br />

Judenhofer M. 200<br />

Justicia C. 150<br />

K<br />

Kachelrieß M. 190<br />

Kaijzel E. 106, 119, 120,<br />

124, 125, 132,<br />

142, 158<br />

Kaiser W. A. 199<br />

Kaisin G. 129<br />

Kaliszczak M. 209<br />

Kallur T. 64<br />

Kaminski G. 192, 195<br />

Kandasamy M. 169<br />

Karczmarczyk U. 70, 146<br />

Karhi T. 130<br />

Karolczak M. 190<br />

Kassiou M. 197, 198<br />

Kaufholz P. 131<br />

Keereweer S. 106, 119,125<br />

Keil B. 52<br />

Keist R. 53, 186<br />

Kepe V. 157<br />

Keramidas M. 139<br />

Kerrebijn J. 119<br />

Kesenheimer C. 201<br />

Kessler H. 87, 128<br />

Keupp J. 103<br />

Khan I. 204<br />

Kiessling F. 72, 122, 175<br />

Klaubert D. 90<br />

Klohs J. 186<br />

Kneilling M. 200, 201<br />

Knetsch P. 178<br />

Knödgen E. 123<br />

Knudsen G. M. 35, 156<br />

Knuuti J. 71<br />

Kobylecka M. 193<br />

Komm B. 91<br />

Kossodo S. 89<br />

Kotek G. 163, 166, 167<br />

Krais R. 216<br />

Krams R. 100<br />

Krautkramer M. 36<br />

Krenning E.P. 107<br />

Krestin G. 163, 166, 167,<br />

168<br />

Krewer B. 208<br />

Krolicki L. 193<br />

Krucker T. 41<br />

Kruttwig K. 171<br />

Krzanowski M. 70, 146<br />

Kubicek V. 179<br />

Kuhnast B. 187, 202<br />

Kumar-Singh S. 159<br />

Kunikowska J. 193<br />

Kuppen P. 55, 142<br />

Kuśnierz-Cabala B. 146<br />

Kyrgyzov I. 43<br />

L<br />

Labate V. 121<br />

Lacasta C. 184<br />

Lacivita E. 151<br />

Laget M. 135<br />

Lahoutte T. 29, 101, 136<br />

Laine J. 71<br />

Laitinen I. 71<br />

Lamberton F. 148<br />

Langereis S. 44, 103<br />

Lapp R. 190<br />

Lazar A. 179<br />

Lebenberg J. 211<br />

Lebon V. 211<br />

Lederle W. 72, 122<br />

Le Helleix S. 202<br />

Lehel S. 35<br />

Lehmann F. 199<br />

Lehmann S. 53<br />

Lemaire C. 129<br />

Le Masne Q. 43<br />

Lenda-Tracz W. 194<br />

Leopoldo M. 151<br />

Lepetit-Coiffé M. 83, 88<br />

Leppänen P. 71<br />

Leroy C. 214, 215<br />

Lettieri A. 143<br />

Levrey O. 43<br />

Liang J. 164<br />

Libani I. V. 56<br />

Libani I.V. 165<br />

Li G. 164<br />

Linhart V. 184<br />

Lin S. A. 89<br />

Li R. 155<br />

Liu Z. 175<br />

Llosá G. 184<br />

Lo Dico A. 218<br />

Long N. 189<br />

Longo D. 213<br />

López M. 126


López M.E. 126<br />

Löwik C.W.G.M. 55, 106,<br />

119, 120, 124, 125,<br />

132, 142, 158<br />

Lub J. 73<br />

Lucignani G. 56, 84, 165<br />

Lui R. 56, 165<br />

Luoto P. 130<br />

Luthra S. 204<br />

Luxen A. 129, 149<br />

M<br />

Mackeyev Y. 133<br />

Mackiewicz N. 187<br />

Macrez R. 145<br />

Madaschi L. 56<br />

Madsen K. 35<br />

Maecke H. 52, 77, 117, 206<br />

Maggi A. 90, 91<br />

Maier F. C. 34<br />

Maina T. 107<br />

Mainini F. 114<br />

Maitrejean S. 43<br />

Mali A. 130<br />

Mangues R. 126<br />

Mannheim J. 34, 200<br />

Mansfield J. 41<br />

Mansi R. 52, 206<br />

Marengo M. 42<br />

Marfia G. 56<br />

Markelc B. 127<br />

Marner L 35<br />

Maroy R. 214, 215<br />

Marra F. 56<br />

Marsouvanidis I.P. 107<br />

Martelli C. 84, 165<br />

Martin A. 152<br />

Martinelli J. 48<br />

Martins A. 179<br />

Masiello V. 218<br />

Massberg S. 26<br />

Matarrese M. 218<br />

Máthé D. 37<br />

Mathejczyk J. 208<br />

Mathejczyk J.E. 128<br />

Mathis G. 135<br />

Maubert E. 145<br />

Maulucci G. 121<br />

Maurin M. 70<br />

Mauro A. 218<br />

Mautino A. 205<br />

Mazurek A. 195<br />

Mccoll B. 198<br />

Meijering E. 166<br />

Mele M. 121<br />

Menchise V. 79, 176<br />

Mercier F. 129<br />

Mérian J. 207<br />

<strong>5th</strong> <strong>EuropEan</strong> <strong>MolEcular</strong> <strong>IMagIng</strong> <strong>MEEtIng</strong> – EMIM2010<br />

Merli D. 56<br />

Messeguer A. 150<br />

Meyer A. 131<br />

Meyer Y. 180<br />

Mezzanotte L. 120<br />

Michelini E. 120<br />

Mieog J. 142<br />

Mieog S. 55<br />

Mikołajczak R. 70, 146<br />

Miller P. 74, 189<br />

Miranda S. 126<br />

Mol I. 106, 119, 124, 125<br />

Monfared P. 123, 134, 219,<br />

220<br />

Montagne A. 66, 145<br />

Moonen C. 83, 88<br />

Moreira J.N. 174<br />

Moresco R. M. 218<br />

Morfin J.F. 179<br />

Morisson-Iveson V. 204<br />

Mortelmans L. 30, 116,<br />

137, 170<br />

Moser E. 115<br />

Mottaghy F. 30, 116, 137<br />

Mronz M. 190<br />

Musiani P. 205<br />

Muylle K. 138<br />

N<br />

Napolitano R. 79<br />

Napp J. 128, 208<br />

Narula J. 95<br />

Nataf S. 65<br />

Navarro F. 207<br />

Nekolla S. 97<br />

Neumaier B. 134, 219, 220<br />

Nguyen H. H. P. 169<br />

Nguyen Q. D. 209<br />

Nicolaij K. 47<br />

Nicolas G. 206<br />

Nicolay K. 73<br />

Niessen W. 166<br />

Nighoghossian N. 65<br />

Noack P. 180<br />

Nock B.A. 107<br />

Nock J. 216<br />

Ntziachristos V. 99, 185,<br />

188<br />

Nuyts J. 137<br />

O<br />

Odenthal J. 34<br />

Odoardi F. 51<br />

Oh S. Y. 153<br />

Oikonen V. 130<br />

Oliver J. F. 184<br />

Opalinska M. 70, 146<br />

Orset C. 66, 145, 148<br />

O’shea D. 204<br />

Ottobrini L. 56, 84, 165<br />

P<br />

Pach D. 146<br />

Palmowski M. 122<br />

Panieri E. 121<br />

Pani G. 121<br />

Pardo L. A. 208<br />

Paris J. 129<br />

Park E. 153<br />

Park K. W. 153<br />

Park S. 155<br />

Passmore J. 204<br />

Passon M. 199<br />

Pauli J. 128<br />

Pawlak D. 193<br />

Peces-Barba G. 203<br />

Peleman C. 29<br />

Pereson S. 159<br />

Perez-Rial S. 203<br />

Perkuhn M. 72<br />

Perrone R. 151<br />

Pesenti E. 84<br />

Pestourie C. 173<br />

Peterson J. 89<br />

Petoud S. 45, 182<br />

Petrik M. 178<br />

Pichler B. 34, 178, 200, 201<br />

Pickarski M. 89<br />

Pietzsch H.J. 178<br />

Pijarowska J. 154<br />

Pikkemaat J. 44<br />

Pinker K. 115<br />

Pirozzi G. 143<br />

Pisaneschi F. 209<br />

Pisani E. 172<br />

Planas A. 150<br />

Plenevaux A. 149<br />

Plenge E. 166<br />

Pluchino S. 80<br />

Podgajny Z. 192, 195<br />

Politi L. S. 218<br />

Ponsaerts P. 162<br />

Pontecorvo M. 36<br />

Praet J. 162<br />

Prenant C. 198<br />

Pucci B. 172<br />

Q<br />

Que I. 106, 120, 124, 132<br />

Quesson B. 83<br />

R<br />

Rafecas M. 184<br />

Rainone V. 84<br />

Rajopadhye M. 89<br />

Rakowski T. 70<br />

Rando G. 91<br />

Rangaraj N. 151<br />

Rangger C. 178<br />

Raphael B. 43<br />

Rapic S. 134, 219, 220<br />

Ratering D. 186<br />

Raynaud J.S. 145<br />

Razansky D. 99, 188<br />

Reder S. 97<br />

Reischl G. 34, 200, 201<br />

Renard P.Y. 180<br />

Resch-Genger U. 128<br />

Reubi J. C. 52, 77<br />

Reubi J.C. 206<br />

Reumers V. 63<br />

Reutelingsperger C. 116<br />

Rexin A. 140<br />

Riccardi P. 155<br />

Richard J.A. 180<br />

Righini C. 139<br />

Riou A. 65<br />

Ripoll J. 186, 217<br />

Rix A. 72, 122<br />

Robillard M. 47<br />

Robins E. 209<br />

Röcken M. 200, 201<br />

Roda A. 120<br />

Roivainen A. 54, 71, 130<br />

Rojas S. 126<br />

Romanowicz G. 36<br />

Romieu A. 180<br />

Ronchetti F. 218<br />

Rosell Y. 177<br />

Rossin R. 103<br />

Rothwell N. 198<br />

Rubio M. 145<br />

Rudan D. 123, 134, 219,<br />

220<br />

Rudin M. 39, 53, 186<br />

Ruiz-Cabello J. 177, 181<br />

Russo M. 84<br />

Rutten E. 138<br />

<strong>EuropEan</strong> SocIEty for <strong>MolEcular</strong> <strong>IMagIng</strong> – <strong>ESMI</strong><br />

S<br />

Saanijoki T. 54, 130<br />

Saha K. 36<br />

Salerno M. 179<br />

Salinas B. 177, 181<br />

Salvatore M. 143<br />

Sanches P. 103<br />

Saraste A. 97<br />

Sawall S. 190<br />

Saxena R. 151<br />

Sbarbati A. 42<br />

Schäfers M. 98<br />

Schellenberger E. 69<br />

Schibli R. 77<br />

Schibli R. 53, 196<br />

IndEx


228<br />

WarSaW, poland May 26 – 29, 2010<br />

Schmid A. 34<br />

Schmidt D. 155<br />

Schmuck K. 208<br />

Schneider G. 134, 219<br />

Schober A. 72<br />

Schulze A. 131<br />

Schulz P. 140<br />

Schulz R. 72, 188<br />

Schwaiger M. 97<br />

Schwartz Jr S. 126<br />

Scotti G. 218<br />

Sebrie C. 147<br />

Sée V. 24, 25<br />

Sersa G. 127<br />

Shade C. 182<br />

Shamsaei E. 209<br />

Sherif H. 97<br />

Sigovan M. 68<br />

Sihver W. 131<br />

Sijbers J. 159<br />

Silengo L. 205<br />

Silvola J. 71<br />

Sims-Mourtada J. 105, 133<br />

Sipilä H. 54, 71<br />

Siquier K. 197<br />

Siquier-Pernet K. 160<br />

Skovronsky D. 36<br />

Slütter B. 132<br />

Smith G. 209<br />

Snoeks T. 106, 119, 124,<br />

125, 158<br />

Socher I. 199<br />

Soema P. 132<br />

Solevi P. 184<br />

Soria G. 150<br />

Sowa-Staszczak A. 70, 141,<br />

146<br />

Spadaro M. 114<br />

Spinelli A. E. 42<br />

Spivey A. C. 209<br />

Srinivas M. 161<br />

Stadelbauer A. 115<br />

Stein J. 38<br />

Stiller D. 34<br />

Stompor T. 70, 146<br />

Strijkers G. 96<br />

Stühmer W. 208<br />

Stuker F. 186, 196<br />

Suárez L. 126<br />

Sué M. 216<br />

Suilamo S. 130<br />

Sułowicz W. 146<br />

Sur C. 89<br />

Suzenet F. 179, 182<br />

Svarer C. 156<br />

Swinnen J. 137<br />

Szalus N. 192, 195<br />

Szigeti K. 37<br />

imaging life<br />

T<br />

Takano A. 78<br />

Tamma M. 206<br />

Tamma M. L. 77<br />

Tannous B. 185<br />

Tapfer A. 97<br />

Taruttis A. 99<br />

Tatsi A. 107<br />

Tauber C. 157<br />

Taulier N. 172<br />

Tavitian B. 152, 160, 173,<br />

187, 191, 197,<br />

202, 211<br />

Tchouate Gainkam O 136<br />

Tei L. 48<br />

Teissie J. 127<br />

Teräs M. 130<br />

Terreno E. 114, 174, 213<br />

Texido G. 84<br />

Texier I. 207<br />

Thézé B. 152, 173, 191<br />

Thonon D. 129, 149<br />

Tiemann K. 103<br />

Tietze L. F. 128, 208<br />

Toelen J. 63, 85<br />

Torres E. 114<br />

Toth E. 179, 182<br />

Tousseyn T. 30<br />

Trabattoni D. 84<br />

Trattnig S. 115<br />

Trebossen R. 214, 215<br />

Trigg W. 204<br />

Trinquet E. 135<br />

Tsapis N. 172<br />

Tudela R. 150<br />

Turco E. 205<br />

Tworowska I. 105, 133<br />

U<br />

Uijtterdijk A. 167<br />

Urbach W. 172<br />

V<br />

Vahrmeijer A. 55, 142<br />

Vainio J. 130<br />

Vainio P. 130<br />

Valtorta S. 218<br />

Van Auderkerke J. 212<br />

Van Beek E. 158<br />

Van Beers B.E. 28<br />

Van Broeckhoven C. 159<br />

Van Buul G. 167<br />

Van Dam G. M. 99<br />

Van Den Haute C. 63<br />

Vandeputte C. 85<br />

Van Der Heiden K. 100<br />

Van Der Linden A. 57,<br />

138, 150, 159, 162,<br />

169,212, 220<br />

Van Der Vorst J. 55, 142<br />

Van De Velde C. 55, 142<br />

Vandormael S. 138<br />

Van Driel P. 106, 119, 125<br />

Van Duijnhoven S. 47<br />

Vaneycken I. 29, 136<br />

Vanhoutte G. 159<br />

Vanhove C. 101<br />

Van Laere K. 33, 85, 170<br />

Van Osch G. 167<br />

Van Santvoort A. 170<br />

Van Tiel S. 163, 168<br />

Van Weerden W. 137<br />

Van Weeren L. 61<br />

Varrone A. 75<br />

Vats D. 186, 196<br />

Venel Y. 157<br />

Veraart J. 159<br />

Verbruggen A. 30, 85, 116<br />

Vercouillie J. 157<br />

Verfaillie C. 170<br />

Verhaar J. 167<br />

Verhoye M. 169, 212<br />

Vermaelen P. 170<br />

Verwijnen S. 118<br />

Viel T. 123, 134, 219, 220<br />

Vilar R. 189<br />

Villette S. 182<br />

Vivien D. 66, 145, 148<br />

Vollmar S. 216, 220<br />

Von Hof J. M. 208<br />

Vreys R. 162, 169<br />

Vuillemard C. 152<br />

W<br />

Waerzeggers Y. 219<br />

Wang H. 196<br />

Watson J. 90<br />

Weber C. 72<br />

Weber W. 27, 117, 206<br />

Wehrl H. F. 34<br />

Weidl E. 97<br />

Weigelin B. 161<br />

Weinans H. 167<br />

Wester H.J. 87, 97, 178<br />

Whelan P.F. 210<br />

Wiart M. 65<br />

Wicki A. 52<br />

Wiedenmann B. 140<br />

Wiehr S. 34, 200, 201<br />

Wielopolski P. 163, 167, 168<br />

Wilczynski G. 67<br />

Wild D. 52<br />

Wilson L. J. 133<br />

Winkeler A. 160, 219<br />

Woff E. 138<br />

Wolfs E. 170<br />

Wouters L. 149<br />

Wurdinger T. 185<br />

X<br />

Xavier C. 29, 136<br />

Xie B. 106, 119, 125<br />

Y<br />

Yang E. 164<br />

Yang X. 164<br />

Yared W. 89<br />

Ylä-Herttuala S. 71<br />

Young A. 148<br />

Yudina A. 88<br />

Z<br />

Zacharopoulos A. 217<br />

Zannetti A. 143<br />

Zara G. 218<br />

Zeebregts C. J. 99<br />

Zenga F. 218<br />

Zheng J. 160<br />

Zhong P. 164<br />

Ziegler S. 40<br />

Zwier J. 135


230<br />

WarSaW, poland May 26 – 29, 2010<br />

<strong>ESMI</strong>/DiMI management office<br />

c/o MPI for neurological research<br />

Gleueler Str. 50<br />

50931 Köln<br />

T ++49 2 21 4 78 8 72 44<br />

++49 2 21 4 78 8 79 60<br />

E office@e-smi.eu<br />

Layout: Doris Kracht<br />

Binding and Printing: Hundt Druck GmbH<br />

imaging life

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!