25.02.2013 Views

2011 - ExMI – Experimental Molecular Imaging - RWTH

2011 - ExMI – Experimental Molecular Imaging - RWTH

2011 - ExMI – Experimental Molecular Imaging - RWTH

SHOW MORE
SHOW LESS

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

Chair of <strong>Experimental</strong> <strong>Molecular</strong> <strong>Imaging</strong><br />

Faculty of Medicine<br />

<strong>Imaging</strong><br />

Pathophysiology<br />

Down to the<br />

<strong>Molecular</strong> Level<br />

Director<br />

Univ.-Prof. Dr. med. Fabian Kießling<br />

<strong>RWTH</strong> Aachen University Hospital<br />

Pauwelsstrasse 30, D-52074 Aachen<br />

Helmholtz-Institute for Biomedical Engineering<br />

Pauwelsstrasse 20, D-52074 Aachen<br />

Phone: +49 (0)241 80 80116 (Secretary)<br />

+49 (0)241 80 80117 (Office)<br />

Fax: +49 (0)241 80 33 80116<br />

Email: fkiessling@ukaachen.de<br />

Web: http://www.molecular-imaging.ukaachen.de<br />

Staff<br />

Abou-Elkacem, Lotfi, Dipl.-Biol, PhD student<br />

Al Rawashdeh, Wa’el, MSc, PhD student<br />

Appold, Lia, BSc student<br />

Arns, Susanne, Technical assistant<br />

Bätke, Sarah, MSc, PhD student<br />

Berker, Yannick, Dipl.-Ing. MSc, PhD student<br />

Bölükbas, Ali, MSc student<br />

Bzyl, Jessica, BSc, PhD student<br />

Curaj, Adelina, MD, PhD student<br />

Doleschel, Dennis, Dipl.-Biol, PhD student<br />

Duarte Jorge, Samuel, MSc student<br />

Dueppenbecker, Peter, Dipl.-Ing., PhD student<br />

Ehling, Josef, Dr. med., Postdoc<br />

Fokong, Stanley, MSc, PhD student<br />

Franke, Jochen, MSc student<br />

Fuge, Felix, Dipl.-Chem, PhD student<br />

Gätjens, Jessica, Dr. rer. nat., Group leader<br />

Gebhardt, Pierre, Dipl.-Ing., PhD student<br />

Goldschmidt, Benjamin, Dipl.-Ing., PhD student<br />

Gremse, Felix, Dipl.-Ing., PhD student<br />

Grouls, Christoph, MD Student<br />

Jayapaul, Jabadurai, MSc, PhD student<br />

Koczera, Patrick, MD student<br />

Kunjachan, Sijumon, MSc, PhD Student<br />

Lammers, Twan, PhD, DSc, Group leader<br />

Lederle, Wiltrud, Dr. rer. nat., Group leader<br />

Liu, Zhe, PhD, Postdoc<br />

Mertens, Marianne, MSc, PhD student<br />

Mertens, Natascha, Technical assistant<br />

<strong>2011</strong><br />

Helmholtz-Institute for Biomedical Engineering<br />

<strong>RWTH</strong> Aachen University<br />

Möckel, Diana, Technical assistant<br />

Novak, Mara, Technical assistant<br />

Palmowski, Karin, Dr. med., Postdoc<br />

Palmowski, Moritz, PD Dr. med., Group leader<br />

Rix, Anne, Technical Assistant<br />

Salomon, Andre, Dr.-Ing., Postdoc<br />

Schultz, Volkmar, Dr.-ing., Group leader<br />

Theek, Benjamin, MSc, student<br />

Tiemann, Birgit, Administrative assistant<br />

Truhn, Daniel, Dipl.-Phys., MSc, PhD student<br />

Weiler, Marek, Technical Assistant<br />

Weissler, Bjoern, Dipl.-Ing., PhD Student<br />

Wu, Zhuojun, MSc, PhD student<br />

Yokota Rizzo, Larissa, MSc, PhD student<br />

35


<strong>Experimental</strong> <strong>Molecular</strong><br />

<strong>Imaging</strong><br />

36<br />

Helmholtz-Institute for Biomedical Engineering<br />

<strong>RWTH</strong> Aachen University<br />

Introduction<br />

The Chair of <strong>Experimental</strong> <strong>Molecular</strong> <strong>Imaging</strong> (<strong>ExMI</strong>) at the<br />

Helmholtz-Institute for Biomedical Engineering (HIA) at<br />

<strong>RWTH</strong> Aachen University focuses on the development and<br />

evaluation of novel imaging methods and contrast agents<br />

to characterize and treat cancer and cardiovascular disorders.<br />

This is mainly achieved by vascular targeting and<br />

theranostic approaches, which are developed and tested in<br />

preclinical animal models. In this context, novel biomaterials,<br />

including nanoparticles, polymeric carriers, liposomes<br />

and microbubbles can play a major role. Their delivery and<br />

therapeutic efficacy can be assessed by functional and molecular<br />

imaging, which has become an established tool in<br />

preclinical research.<br />

<strong>ExMI</strong> follows a multimodal approach, based on combinations<br />

of MRI, CT, ultrasound, optical imaging and nuclear<br />

medicine techniques. Image fusion, hybrid imaging as well<br />

as multimodal contrast agents are in the research scope.<br />

Usually, the choice of imaging strategies and contrast agents<br />

follows the biological and medical problem to be investigated.<br />

Furthermore, we intend to better connect preclinical<br />

and clinical research, and to translate novel surrogate markers<br />

and (molecular) diagnostics into clinical evaluation.<br />

Currently, the department consists of five research groups,<br />

working on the biological mechanisms of tumor development<br />

and angiogenesis, on the synthesis of novel imaging<br />

probes, on theranostic concepts to treat cancer and cardiovascular<br />

diseases, on MR-PET hybrid imaging, and on translational<br />

studies to bring functional and molecular ultrasound<br />

to the clinic.<br />

Mechanisms of Tumor<br />

Progression and Metastasis<br />

The group “Mechanisms of Tumor Progression and<br />

Metastasis” aims at elucidating the mechanisms of tumor<br />

growth and progression, with a special focus on the impact<br />

of the tumor microenvironment and the longitudinal in<br />

vivo characterization by non-invasive imaging. Besides tumor<br />

staging and improved diagnosis, further attempts are<br />

directed to monitor therapy effects, including the evaluation<br />

of novel therapeutics. In vivo imaging is complemented<br />

by histological and functional in vitro analyses in order<br />

to validate the data and to identify new potential markers<br />

and targets.<br />

We recently compared different imaging modalities with<br />

respect to the non-invasive assessment of tumor growth<br />

and size in GFP/RFP-expressing colon carcinoma xenografts.<br />

Magnetic resonance imaging (MRI) was proven to<br />

be highly sensitive and accurate for tumor size and growth<br />

assessment. Micro-computed tomography (µCT) provided<br />

an almost similar accuracy. Optical reflectance imaging<br />

(ORI) was as sensitive as MRI, but overestimated the tumor<br />

size. Fluorescent intensity measurements provided more<br />

accurate data of the tumor size than fluorescent area analyses<br />

(Fig. 1) [1] .<br />

[1] Abou-Elkacem et al, Anticancer Res 31: 2907 (<strong>2011</strong>)<br />

<strong>2011</strong><br />

Fig.1. A: Monitoring of tumor growth in s.c. HCT 116-GFP-<br />

RFP xenografts using ORI, MRI and μCT. Representative<br />

images show tumors at day 1, 3 and 15 after inoculation.<br />

B: Correlation of the tumor volumes determined in vivo by<br />

MRI, μCT and ORI with ex vivo caliper data at day 12 and<br />

15 (n=9). From [1] .<br />

A second study aimed at non-invasive apoptosis assessment<br />

in response to anti-angiogenic therapy, using both<br />

optical imaging (Annexin-Vivo) and gamma counting<br />

( 99m Tc-HYNIC-Annexin V). As opposed to immunohistochemistry,<br />

which clearly showed apoptosis induction<br />

after treatment with the multi-tyrosine kinase inhibitor<br />

SU11248, both Annexin probes were found to be unable<br />

to specifically detect apoptosis in vivo, most probably because<br />

of the breakdown of the vasculature and reduced<br />

probe delivery [2] .<br />

Due to the discovery of the erythropoietin-receptor<br />

(EpoR) on tumor cells, the use of erythropoietin (Epo) for<br />

the treatment of cancer-induced anemia is currently under<br />

intense debate. In order to elucidate the role of Epo<br />

and EpoR in lung cancer, a near-infrared probe for fluorescence<br />

mediated tomography (FMT) was developed and<br />

tested for longitudinally assessing EpoR status in lung cancer<br />

xenografts with different EpoR expression levels. As<br />

shown in Fig. 2, the probe was found to be highly specific,<br />

and could accurately determine the EpoR expression level<br />

of the xenografts in vivo, as demonstrated by FMT/µCT<br />

hybrid imaging. These data suggest a high potential of this<br />

newly developed probe for analyzing the EpoR status in<br />

tumors and the role of Epo in lung cancer [3] .<br />

Fig. 2. Specific binding of Cy5.5-labeled Epo (red) to tumors<br />

with different EpoR expression levels (lower in A549<br />

(A); higher in H838 (B)), as demonstrated via in vivo competition<br />

experiments with excess unlabeled Epo (blue).<br />

FMT/μCT hybrid imaging clearly confirmed the stronger<br />

accumulation of Epo-Cy5.5 in H838 (E) as compared to<br />

A549 (D). White arrows indicate tumors, hatched arrows<br />

bone. From [3] .<br />

[2] Lederle et al, EJNMMI Research 1: 26 (<strong>2011</strong>)<br />

[3] Doleschel et al, J Nucl Med (in press)


Probe Design for <strong>Molecular</strong><br />

<strong>Imaging</strong><br />

The chemical group within <strong>ExMI</strong>, which focuses on the development<br />

of novel probes for molecular imaging, has in<br />

the past year made significant progress towards the development<br />

of FMN- and FAD-coated superparamagnetic iron<br />

oxide (USPIO) nanoparticles, which were shown to be highly<br />

suitable for in vivo imaging of tumors and tumor angiogenesis,<br />

as well as for ex vivo labeling endothelial cells [4] .<br />

In addition, USPIO have been incorporated into poly(butyl<br />

cyanoacrylate)-based microbubbles (MB), thereby enabling<br />

the non-invasive visualization of MB destruction using MRI [5] .<br />

These USPIO-containing MB have furthermore been shown<br />

to be useful for mediating and monitoring drug delivery<br />

across the blood-brain-barrier (BBB). This is exemplified by<br />

Fig. 3, showing the deposition of USPIO nanoparticles released<br />

from MB into the brain upon ultrasound-mediated destruction,<br />

as well as extravasation of the blood pool marker<br />

FITC-dextran from systemic circulation across the BBB [6] .<br />

Fig. 3. Mediating and monitoring blood-brain-barrier<br />

(BBB) permeation using USPIO-containing MB. A-B: T2*relaxation<br />

rates in the brains of mice before (A) and 45<br />

min after (B) the application of USPIO-MB plus destructive<br />

ultrasound (US)<br />

pulses, showing<br />

efficient deposition<br />

of released<br />

USPIO across<br />

the BBB. C-D:<br />

Upon combining<br />

USPIO-MB<br />

with US, significantextravasation<br />

of FITCdextran<br />

(green)<br />

into the brain<br />

was observed<br />

(C), which was<br />

not the case for<br />

non-US-treated<br />

controls (D).<br />

Blood vessels<br />

counterstained<br />

in red.<br />

As part of Patim.NRW, USPIO nanoparticles are also being<br />

developed for visualizing the localization, the resorption and<br />

the functionality of cardiovascular implants, such as stents,<br />

shunts and patches. To this end, as part of an initial proof-ofprinciple<br />

study, collagen-based scaffolds have been prepared<br />

which contain different amounts of USPIO, they have been<br />

characterized using TEM and SEM, cell growth onto and into<br />

these scaffolds has been evaluated, and they have been<br />

used to non-invasively monitor their localization in mice. As<br />

shown in Fig. 4, depending on the amount of USPIO incorporated,<br />

the labeled scaffolds could be visualized with high<br />

sensitivity, both ex vivo and in vivo, and USPIO incorporation<br />

did not affect their structural properties [7] . Experiments<br />

evaluating the impact of USPIO labeling on cell growth and<br />

[4] Jayapaul et al, Biomaterials 32: 5863 (<strong>2011</strong>)<br />

[5] Liu et al, Biomaterials 32: 6155 (<strong>2011</strong>)<br />

[6] Koczera et al (in prep)<br />

[7] Mertens et al (in prep)<br />

<strong>2011</strong><br />

Helmholtz-Institute for Biomedical Engineering<br />

<strong>RWTH</strong> Aachen University<br />

aiming to assess the suitability of these matrices for monitoring<br />

scaffold resorption are currently ongoing.<br />

Fig. 4. USPIO-labeled scaffolds. A-C: Collagen-based matrices<br />

were labeled with different amounts of USPIO (A), and<br />

their structural properties were evaluated using scanning<br />

electron microscopy at 20x (B) and 80x (C) magnification.<br />

D-E: Standard (D) and color-coded (E) T2*-imaging of a<br />

USPIO-labeled scaffold (arrow) upon intraperitoneal implantation.<br />

Nanomedicines and theranostics<br />

Efforts in our group primarily focus on the development of image-guided<br />

nanomedicines for treating cancer and inflammatory<br />

disorders. In the past year, significant progress has been in<br />

the development of passively and actively targeted polymers<br />

and micelles for drug targeting to tumors, as well as in the design<br />

and evaluation of various different nanomedicine formulations<br />

for treating rheumatoid arthritis [8]; [9]; [10] .<br />

In addition, we have evaluated how well nanocarrier materials<br />

are able to overcome multidrug resistance (MDR). To<br />

this end, three different doxorubicin-containing nanomedicines<br />

(i.e. liposomes, polymers and micelles) and four different<br />

cell lines were used (i.e. A431, SW620, B16 and CT26;<br />

both in parental and in MDR-form; obtained upon continuously<br />

exposing the cells to IC90 concentrations, and selecting<br />

surviving cells; see Fig. 5A-B). In line with the literature,<br />

nanomedicines suffered less from the expression of MDRconferring<br />

drug efflux pumps than did free doxorubicin, as<br />

confirmed by both cytotoxicity and cell uptake experiments<br />

(Fig. 5C). As opposed to the literature, however, in which<br />

often pharmacologically active carrier materials and/or artificially<br />

resistant (e.g. Pgp-overexpressing) cells are used, the<br />

differences in IC50 between parental and MDR-cells were<br />

much smaller (2-4; vs. 10-100), indicating that the ability of<br />

clinically relevant nanomedicines to overcome physiologically<br />

relevant MDR should not be overestimated [11] .<br />

Additional experiments in our group, performed as part of<br />

the ForSaTum project, have aimed at imaging and analyzing<br />

[8] Huis in ‘t Veld et al, Nanoscale 3: 4022 (<strong>2011</strong>)<br />

[9] Talleli et al, J Control Release 153: 93 (<strong>2011</strong>)<br />

[10] Crielaard et al (in prep)<br />

[11] Kunjachan et al, Eur J Pharm Sci (in press)<br />

<strong>Experimental</strong> <strong>Molecular</strong><br />

<strong>Imaging</strong><br />

37


<strong>Experimental</strong> <strong>Molecular</strong><br />

<strong>Imaging</strong><br />

38<br />

Helmholtz-Institute for Biomedical Engineering<br />

<strong>RWTH</strong> Aachen University<br />

tumor angiogenesis using a combination of in vivo and ex vivo<br />

micro-computed tomography (µCT) and immunohistochemistry<br />

(IHC). To this end, as exemplified in part by Fig.<br />

6, blood vessels in 4 different subcutaneous xenograft tumors<br />

(A431, Calu-6, MLS and A549) were visualized using<br />

both µCT and IHC, and morphological characteristics such<br />

as vessel size, vessel distribution and vessel branching were<br />

analyzed [12] Fig. 5. Overcoming multidrug-resistance (MDR) using nanomedicines.<br />

A-B: Three different doxorubicin (Dox) -containing<br />

nanomedicine formulations (polymers, liposomes and<br />

micelles; B) were used to evaluate to the impact of drug<br />

targeting on overcoming MDR, hypothesizing that nanomedicines<br />

are more useful than free drugs for achieving target<br />

site (nuclear) localization in vitro (A). C: Fluorescence microscopy<br />

studies, confirming that the upregulation of MDRconferring<br />

drug efflux pumps affects the cellular uptake of<br />

free Dox more strongly than that of Dox-containing nanomedicines.<br />

Dox: red. Nuclei: blue. Cell membranes: green.<br />

. In addition, in vivo µCT imaging<br />

was implemented to non-invasively assess<br />

the relative blood volume (rBV) in these 4<br />

tumors models. Upon comparing the results<br />

obtained using µCT to those obtained<br />

using IHC, which still is the golden standard<br />

in (anti-)angiogenesis experiments, it was<br />

found that the results obtained using both<br />

Fig. 6: Visualization of tumor blood vessels in A549 (A,B,E)<br />

and A431 (C,D,F) tumor xenografts using in vivo (A,C) and<br />

ex vivo (B,D) micro-computed tomography and immunohistochemistry<br />

(E-F). Arrows indicate degree of blood vessel<br />

branching. Blood vessels are stained in green, α-SMA in<br />

red and nuclei in blue.<br />

<strong>2011</strong><br />

experimental procedures correlated very<br />

well, with correlation coefficients far above<br />

0.9 and p-values far below 0.05, indicating that<br />

functional in vivo µCT imaging is a highly suitable<br />

modality for non-invasively evaluating tumor<br />

angiogenesis and the vascular response<br />

to anti-angiogenic therapies.<br />

Hybrid <strong>Imaging</strong><br />

Technologies<br />

New system developments: The group Hybrid <strong>Imaging</strong><br />

Technology (HIT) has developed a new detector concept<br />

for simultaneous Positron Emission Tomography (PET) and<br />

Magnetic Resonance <strong>Imaging</strong> (MRI). The team succeeded in<br />

applying this new detector technology in a preclinical PET/<br />

MR scanner (see Fig. 7). The scanner is designed as an insert<br />

for a human 3T MRI system [13] . The system was designed,<br />

built, and tested within the timeframe of the EU FP7 project<br />

HYPERImage and will be further optimized within the<br />

NRW project ForSaTum. The new MR-compatible detector<br />

stack uses analog and digital Silicon PhotoMultiplier (SiPM)<br />

technology which further advances the performance (sub<br />

nano-second coincidence resolution time) of future semiconductor<br />

based PET system. The detector has been developed<br />

in cooperation with the University of Heidelberg<br />

(Prof. Peter Fischer) and the Fondazione Bruno Kessler<br />

(Claudio Piemonte, PhD). The team has developed a new<br />

data acquisition and control environment for this new generation<br />

of digital PET scanners, which enables a purely SW<br />

based data processing and thus the application of new iterative<br />

algorithms, like self normalization and calibration [14] ,<br />

or maximum likelihood crystal identification to improve the<br />

image resolution and sensitivity [15] . A special gamma-transparent<br />

Radio Frequency (RF) coil supporting simultaneous<br />

PET/MR has been developed for imaging of animals with a<br />

size up to rabbits [16] Fig. 7: Left: simultaneously acquired PET-, MR-, and fused<br />

PET plus MR image. Right: preclinical PET/MR system<br />

. The interference investigation using<br />

the new MR-compatible PET modules finally shows no significant<br />

performance degradation of PET and MRI during simultaneous<br />

acquisition.<br />

MR-based attenuation correction for hybrid PET/MRI systems:<br />

Accurate gamma photon attenuation correction<br />

(AC) is essential for quantitative PET/MRI as there is no<br />

simple relation between MR image intensity and attenuation<br />

coefficients. Attenuation maps (µ-maps) can be<br />

[12] Ehling et al (in prep)<br />

[13] Schulz et al, IEEE NSS, J2-05 (<strong>2011</strong>)<br />

[14] Goldschmidt et al, IEEE NSS, MIC18.M-194 (<strong>2011</strong>)<br />

[15] Lerche et al, IEEE NSS, MIC18.M-180 (<strong>2011</strong>)<br />

[16] Truhn et al, Med Phys 38: 3995 (<strong>2011</strong>)


derived by segmenting MR images and assigning attenuation<br />

values to the compartments. Ultra-short echo time<br />

(UTE) sequences have been used to separate cortical bone<br />

and air, and the Dixon technique has enabled distinguishing<br />

soft and adipose tissues. Unfortunately, sequential application<br />

of these sequences is time-consuming and complicates<br />

image registration. Within the HIT we develop new<br />

MR sequences and mathematical techniques to extract<br />

the attenuation of the gamma photon out of the MR and<br />

the PET data [17] . We recently proposed a UTE triple-echo<br />

(UTILE) MR- sequence [18] Fig. 8: MRI pulse sequence diagram of our UTILE sequence<br />

(left) with image processing flowchart (right)<br />

combining UTE sampling for<br />

bone detection and gradient echoes for Dixon water/fat<br />

separation in a single-shot 3D acquisition (TE1/TE2/TE3/<br />

TR = 0.09/1.09/2.09/4.1 ms at 3T), see Fig. 2. Thus the<br />

UTILE MR- sequence enables the generation of MR-based<br />

four-class µ-maps without anatomical priors, yielding results<br />

more similar to CT-based ones than with three-class<br />

segmentation only.<br />

Translational Studies<br />

The group translational studies focuses on the late preclinical<br />

imaging and early translational research for monitoring<br />

of tumor angiogenesis, predominantly in skin, breast,<br />

kidney and liver cancer. In this regard, novel imaging techniques<br />

are being developed and evaluated in preclinical animal<br />

models. The group members are radiologists, biologists<br />

and chemists. The main focus is on functional and molecular<br />

ultrasound imaging using in-house prepared non-specific<br />

as well as targeted ultrasound contrast agents. Additionally,<br />

small animal imaging is being performed using MRI, optical<br />

imaging and PET. To verify in vivo experiments, imaging data<br />

is supplemented with immunohistochemical and other<br />

biological methods.<br />

Breast Cancer <strong>Imaging</strong>: Clinically translatable ultrasound<br />

contrast agents are tested for their potential application on<br />

humans. These contrast agents are well suited for characterizing<br />

and distinguishing breast cancer models with different<br />

angiogenesis and aggressiveness [19] . Further, the<br />

molecular information alone is better suited to discriminate<br />

differently aggressive tumor models compared to functional<br />

information alone. We now focus on examining breast<br />

cancer models during tumor growth and treatment using<br />

molecular and functional ultrasound.<br />

[17] Salomon et al, IEEE TMI 30: 804 (<strong>2011</strong>)<br />

[18] Berker et al, J Nucl Med (in press)<br />

[19] Bzyl et al, Eur Radiol 21: 1988 (<strong>2011</strong>)<br />

[20] Rix et al, Eur J Radiol (in press)<br />

<strong>2011</strong><br />

Helmholtz-Institute for Biomedical Engineering<br />

<strong>RWTH</strong> Aachen University<br />

Liver <strong>Imaging</strong>: In cooperation with<br />

the group of Prof. Trautwein, transgenic<br />

mice which spontaneously develop<br />

liver fibrosis and hepatocellular<br />

carcinoma are being examined using<br />

ultrasound and MRI. The aim of these<br />

studies is to investigate the potential<br />

role of targeted ultrasound contrast<br />

agents for assessment of liver dysplasia<br />

compared to the gold standard<br />

MRI.<br />

Kidney <strong>Imaging</strong>: In cooperation with Prof. Dr. Koesters,<br />

transgenic mice which spontaneously develop renal cell<br />

cancer are being examined using ultrasound and CT. The<br />

aim of these studies is to investigate the potential role of<br />

novel ultrasound contrast agents for the characteritzation<br />

of renal cell cancer.<br />

Development of<br />

Novel <strong>Imaging</strong><br />

Probes and Techniques:<br />

For ultrasoundimaging,<br />

2D and 3D<br />

Doppler and<br />

B-mode based imaging<br />

approaches<br />

have been established<br />

[20] . Novel<br />

tumor specific<br />

contrast agents<br />

which are being<br />

synthesized,<br />

characterized and<br />

tested in vitro and<br />

in vivo.<br />

Acknowledgements<br />

This work was supported by<br />

• European Commission (FP7)<br />

• German Research Foundation (DFG)<br />

• German Federal Ministry of Education and Research<br />

(BMBF)<br />

• HighTech.NRW<br />

• START<br />

• Bracco<br />

Awards<br />

Fig. 9: 3D-Reconstruction of a tumor,<br />

used to facilitate functional and molecular<br />

ultrasound imaging.<br />

• Dennis Doleschel, Jabaduraj Jayapaul, Dr. Zhe Liu:<br />

WMIC Travel Grant<br />

• Felix Gremse: Preis für Wissenstransfer der<br />

Medizinischen Gesellschaft Aachen<br />

• Dr. Twan Lammers: DGBMT-Klee prize for<br />

Biomedical Engineering (2nd place)<br />

• Dr. Twan Lammers: International Journal of<br />

Nanomedicine Early Career Award<br />

<strong>Experimental</strong> <strong>Molecular</strong><br />

<strong>Imaging</strong><br />

39


<strong>Experimental</strong> <strong>Molecular</strong><br />

<strong>Imaging</strong><br />

40<br />

Helmholtz-Institute for Biomedical Engineering<br />

<strong>RWTH</strong> Aachen University<br />

Most relevant Publications:<br />

[1.] Berker Y, Franke J, Salomon A, Palmowski M, Donker H, Temur Y,<br />

Mottaghy F, Kuhl C, Izquierdo D, Fayad Z, Kiessling F, Schulz V. MRbased<br />

attenuation correction for hybrid PET/MRI systems: A fourclass<br />

tissue segmentation technique using a combined ultrashort echo<br />

time (UTE)/Dixon MR sequence. J Nucl Med (in press)<br />

[2.] Bzyl J, Lederle W, Rix A, Grouls C, Tardy I, Pochon S, Siepmann M,<br />

Penzkofer T, Schneider M, Kiessling F, Palmowski M. <strong>Molecular</strong> and<br />

functional ultrasound imaging in differently aggressive breast cancer<br />

xenografts using two novel ultrasound contrast agents (BR55 and<br />

BR38). Eur Radiol <strong>2011</strong>;21:1988-1995<br />

[3.] Crielaard B, Yousefi A, Schillemans J, Vermehren C, Buyens K, Braekmans<br />

K, Lammers T, Storm G. An in vitro assay based on surface plasmon<br />

resonance to predict the in vivo circulation kinetics of liposomes.<br />

J Control Release <strong>2011</strong>;156:307-314<br />

[4.] Doleschel D, Mundigl O, Wessner A, Gremse F, Bachmann J, Rodriguez<br />

A, Klingmüller U, Jarsch M, Kiessling F, Lederle W. Targeted<br />

near-infrared imaging of the erythropoietin receptor in human lung<br />

cancer xenografts. J Nucl Med <strong>2011</strong> (in press)<br />

[5.] Fokong S., Siepmann M., Liu Z., Schmitz G., Kiessling F., Gätjens J. Advanced<br />

characterization and refinement of poly n-butyl cyanoacrylate<br />

(PBCA) microbubbles for ultrasound imaging. Ultrasound Med Biol<br />

<strong>2011</strong>;37:1622-1634<br />

[6.] Gremse F, Grouls C, Palmowski M, Vries A, Gruell H, Das M, Mühlenbruch<br />

G, Akhtar S, Schober A, Kiessling F. Virtual Elastic Sphere<br />

Processing Enables Reproducible Quantification of Vessel Stenosis in<br />

CT and MR Angiographies. Radiology <strong>2011</strong>;260:709-717<br />

[7.] Huis In ‘t Veld R, Storm G, Hennink W, Kiessling F, Lammers T. Macromolecular<br />

nanotheranostics for multimodal anticancer therapy. Nanoscale<br />

<strong>2011</strong>;4:4022-4034<br />

[8.] Jayapaul J, Hodenius M, Arns S, Lederle W, Lammers T, Comba P,<br />

Kiessling F, Gaetjens J. FMN-coated fluorescent iron oxide nanoparticles<br />

for RCP-mediated targeting and labeling of metabolically active<br />

cancer and endothelial cells. Biomaterials <strong>2011</strong>;32:5863-5871<br />

[9.] Kiessling F, Gaetjens J, Palmowski M. Application of <strong>Molecular</strong><br />

Ultrasound for <strong>Imaging</strong> Integrin Expression. Theranostics<br />

<strong>2011</strong>;1:127-134<br />

Team<br />

<strong>2011</strong><br />

[10.] Kiessling I, Bzyl J, Kiessling F. <strong>Molecular</strong> ultrasound imaging and its<br />

potential for paediatric radiology. Pediatr Radiol <strong>2011</strong>;41:176-84<br />

[11.] Lammers T, Aime S, Hennink W, Storm G, Kiessling F. Theranostic<br />

Nanomedicines. Acc Chem Res <strong>2011</strong>;44:1029-1038<br />

[12.] Lammers T, Kiessling F, Hennink W, Storm G. Drug targeting to<br />

tumors: Principles, pitfalls and (pre-)clinical progress. J Control<br />

Release (in press)<br />

[13.] Lederle W, Depner S, Schnur S, Obermueller E, Catone N, Just<br />

A, Fusenig NE, Mueller MM. IL-6 promotes malignant growth of<br />

skin SCCs by regulating a network of autocrine and paracrine cytokines.<br />

Int J Cancer <strong>2011</strong>;128:2803-2814.<br />

[14.] Liehn E, Tuchscheerer N, Kanzler I, Drechsler M, Fraehmos L,<br />

Schuh A, Koenen R, Zandler S, Soehnlein O, Hristov M, Grigorescu<br />

G, Urs A, Leabu M, Bucur I, Merx M, Zernecke A, Ehling J,<br />

Gremse F, Lammers T, Kiessling F, Bernhagen J, Schober A, Weber<br />

C. Double-edged role of the CXCL12-CXCR4 axis in experimental<br />

myocardial infarction. J Am Coll Cardiol <strong>2011</strong>;58:2415-2423<br />

[15.] Liu Z, Lammers T, Ehling J, Fokong S, Bornemann J, Kiessling F,<br />

Gätjens L Iron oxide nanoparticle-containing microbubble composites<br />

as contrast agents for MR and ultrasound dual-modality<br />

imaging. Biomaterials <strong>2011</strong>;32:6155-6163<br />

[16.] Rix A, Lederle W, Siepmann M, Fokong S, Behrendt FF, Bzyl J,<br />

Grouls C, Kiessling F, Palmowski M. Evaluation of high frequency<br />

ultrasound methods and contrast agents for characterising tumor<br />

response to anti-angiogenic treatment. Eur J Radiol <strong>2011</strong> (in<br />

press)<br />

[17.] Salomon A, Goedicke A, Schweizer B, Aach T, Schulz V. Attenuation<br />

maps for PET/MR uing using simultaneous reconstruction. IEEE<br />

Transaction on Medical <strong>Imaging</strong>. <strong>2011</strong>;30:804-813<br />

[18.] Truhn D, Kiessling F, Schulz V. Optimised RF Shielding Techniques for<br />

Simultaneous PET/MR. Med Phys <strong>2011</strong>;38:3995-4000<br />

[19.] Wiessler M, Hennrich U, Pipkorn R, Waldeck W, Cao L, Peter J,<br />

Ehemann V, Semmler W, Lammers T, Braun K. Theranostic cRGD-<br />

BioShuttle Constructs Containing Temozolomide- and Cy7 For NIR-<br />

<strong>Imaging</strong> and Therapy. Theranostics <strong>2011</strong>;1:381-394<br />

[20.] Xiao L, Li J, Brougham D, Fox E, Feliu N, Bushmelev A, Schmidt A,<br />

Mertens N, Kiessling F, Fadeel B, Mathur S. Water-Soluble Superparamagnetic<br />

Magnetite Nanoparticles with Biocompatible Coating<br />

for Enhanced Magnetic Resonance <strong>Imaging</strong> (MRI). ACS Nano<br />

<strong>2011</strong>;5:6315-6324

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!