29.11.2012 Views

Microbubble Symposium: Fabrication, Characterisation and ...

Microbubble Symposium: Fabrication, Characterisation and ...

Microbubble Symposium: Fabrication, Characterisation and ...

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.

<strong>Microbubble</strong> <strong>Symposium</strong>:<br />

<strong>Fabrication</strong>, <strong>Characterisation</strong> <strong>and</strong> Translational<br />

Applications<br />

4 th & 5 th July 2011<br />

Weetwood Hall, Otley Road, Headingley, Leeds LS16 5PS


2 <strong>Microbubble</strong> <strong>Symposium</strong>: <strong>Fabrication</strong>, <strong>Characterisation</strong> <strong>and</strong> Translational Applications<br />

4 th & 5 th July 2011


<strong>Microbubble</strong> <strong>Symposium</strong>: <strong>Fabrication</strong>, <strong>Characterisation</strong> <strong>and</strong> Translational Applications 3<br />

4 th & 5 th July 2011<br />

Contents<br />

Program 4<br />

Abstracts:<br />

Page<br />

Oral presentation – Session 1<br />

<strong>Microbubble</strong> <strong>Fabrication</strong> <strong>and</strong> <strong>Characterisation</strong> 7<br />

Oral presentation – Session 2<br />

<strong>Microbubble</strong> Ultrasound <strong>Characterisation</strong> 13<br />

Oral presentation – Session 3<br />

<strong>Microbubble</strong> Translational Applications 19<br />

Poster presentations 24<br />

Delegate list 46


4 <strong>Microbubble</strong> <strong>Symposium</strong>: <strong>Fabrication</strong>, <strong>Characterisation</strong> <strong>and</strong> Translational Applications<br />

4 th & 5 th July 2011<br />

Monday 4 th July 2011<br />

Program<br />

12:00 – 14.00 Registration <strong>and</strong> buffet lunch<br />

14:00 – 14:15 Stephen Evans Welcome<br />

Session 1: <strong>Microbubble</strong> <strong>Fabrication</strong> <strong>and</strong> <strong>Characterisation</strong> – Chair: Stephen Evans<br />

14:15 – 14:50 Marjorie Longo Physical chemistry of lipid/lipopolymer shelled<br />

models of medical microbubbles<br />

14:50 – 15:10 Klazina Kooiman A novel method to determine the lipid viscosity<br />

<strong>and</strong> distribution of coated microbubbles<br />

15:10 – 15:40 Tea / Coffee<br />

15:40 – 16:15 Eleanor Stride Magnetic microbubbles for localised imaging<br />

<strong>and</strong> drug delivery: development,<br />

characterisation <strong>and</strong> preliminary application in<br />

vivo<br />

16:15 – 16:35 Jonathan McKendry Mechanical properties of surface modified<br />

microbubbles by atomic force microscopy<br />

16:35 – 17:10 Ine Lentacker Update on microbubble <strong>and</strong> ultrasound assisted<br />

drug delivery within the Sonodrugs project<br />

18:00 – 19:30 Poster Session <strong>and</strong> Drinks<br />

19:30 - onwards <strong>Symposium</strong> Dinner


<strong>Microbubble</strong> <strong>Symposium</strong>: <strong>Fabrication</strong>, <strong>Characterisation</strong> <strong>and</strong> Translational Applications 5<br />

4 th & 5 th July 2011<br />

Tuesday 5 th July 2011<br />

Session 2: <strong>Microbubble</strong> Ultrasound <strong>Characterisation</strong> – Chair: Steven Freear<br />

09:00 – 09:35 Vassilis Sboros Single microbubble acoustics<br />

09:35 – 09:55 Verya Daeichin Stimulating the subharmonic response of<br />

ultrasound contrast agents using selfdemodulation<br />

09:55 – 10:30 Robert Eckersley Quantitative contrast-enhanced ultrasound<br />

imaging: pitfalls, challenges, <strong>and</strong> advances<br />

10:30 – 11:00 Tea / Coffee<br />

11:00 – 11:20 Serge Mensah Release <strong>and</strong> characterization of a single bubble<br />

11:20 – 11:55 Paul Campbell Observations of microbubble cavitation under<br />

optical trap control<br />

12:00 – 13:30 Lunch<br />

Session 3: <strong>Microbubble</strong> Translational Applications – Chair: Louise Coletta<br />

13:30 – 14:10 Fabian Kiessling <strong>Microbubble</strong>s for molecular imaging <strong>and</strong><br />

theranostics<br />

14:10 – 14:30 Gemma Marston Imaging the development of functional tumour<br />

vasculature in vivo in preclinical models<br />

14:30 – 14:50 Philippe Trochet VevoCQ software allows advanced<br />

quantification of perfusion kinetics parameters<br />

with consistent, reliable <strong>and</strong> actionable data<br />

14:50 – 15:20 Tea / Coffee<br />

15:20 – 15:55 Sarah Fawcett Our early experience of pre clinical in vivo use of<br />

MicroMarker<br />

15:55 – 16:30 Jane Smith (Bates) Bubbles in the clinical setting<br />

16:30 – 16:45 Sir Alex Markham Closing remarks


6 <strong>Microbubble</strong> <strong>Symposium</strong>: <strong>Fabrication</strong>, <strong>Characterisation</strong> <strong>and</strong> Translational Applications<br />

4 th & 5 th July 2011


<strong>Microbubble</strong> <strong>Symposium</strong>: <strong>Fabrication</strong>, <strong>Characterisation</strong> <strong>and</strong> Translational Applications 7<br />

4 th & 5 th July 2011<br />

Session 1: <strong>Microbubble</strong> <strong>Fabrication</strong> <strong>and</strong> <strong>Characterisation</strong><br />

Oral Presentation<br />

Marjorie Longo<br />

University of California<br />

Title: Physical Chemistry of lipid/lipopolymer shelled models of medical microbubbles<br />

Abstract: In this talk, I will reflect upon my group’s investigation of the richness of physical<br />

chemical behavior in microbubble shells resembling the lipid shells of certain medical<br />

microbubbles. These findings will then be related to our most recent work attempting to<br />

underst<strong>and</strong> <strong>and</strong> engineer the performance of microbubbles. We found that a decrease in<br />

the permeability of the shell to air took place with increasing saturated acyl chain length <strong>and</strong><br />

decreased disordered domain boundary density, as could be described by a modified energy<br />

barrier theory that incorporated a simple accessible area model. The morphology of<br />

microbubble shell collapse structures <strong>and</strong> shed particles depended upon both reduced<br />

temperature <strong>and</strong> area compression rate, a consequence of time-temperature superposition.<br />

Shell monolayer phase behavior <strong>and</strong> phase diagrams were strongly dependent upon<br />

saturated acyl chain length of the major component <strong>and</strong> number of chains in the minor<br />

emulsifier component (mono-acyl vs. di-acyl) <strong>and</strong> included discovery of a stabilised<br />

condensed stoichiometric complex (diC14PC3DSPE-PEG20001). Similarly, the bi-acyl<br />

emulsifier (lipopolymer) conferred on the shells 10X higher resistance to gas permeability, a<br />

consequence of the reduced disordered boundary density in the presence of this condensed<br />

component. These results should shed light upon formulation <strong>and</strong> performance of lipidshelled<br />

medical microbubbles. To demonstrate, I will end the talk briefly with some of the<br />

latest observations with respect to maintaining monodispersity, attachment/detachment of<br />

drug delivery vesicles, <strong>and</strong> microbubbles formed at the “optimum” stoichiometric complex<br />

composition.


8 <strong>Microbubble</strong> <strong>Symposium</strong>: <strong>Fabrication</strong>, <strong>Characterisation</strong> <strong>and</strong> Translational Applications<br />

4 th & 5 th July 2011<br />

Session 1: <strong>Microbubble</strong> <strong>Fabrication</strong> <strong>and</strong> <strong>Characterisation</strong><br />

Oral Presentation<br />

Klazina Kooiman<br />

Thoraxcenter, Erasmus MC<br />

Title: A novel method to determine the lipid viscosity <strong>and</strong> distribution of coated<br />

microbubbles<br />

Abstract: Ultrasound contrast agents consist of gas-filled coated microbubbles (MB) with diameters<br />

between 1 <strong>and</strong> 10 µm. Within an ultrasound field, large differences in responses of similar sized MBs<br />

have been reported1,2. Heterogeneous coating properties have been suggested to be the underlying<br />

cause. Until now, properties of this coating, like viscosity have been studied dynamically using a setup<br />

of vibrating MBs in an ultrasound field. This study focuses on determining the viscosity of the<br />

coating for lipid-coated MBs in a static set-up. For that we used MBs with a C4F10 gas core <strong>and</strong> a<br />

coating mixture of DSPC (59.4 mol %), PEG-40 stearate (35.7 mol%), DSPE-PEG(2000) (4.1 mol%) <strong>and</strong><br />

DSPE-PEG(2000)-biotin (0.8 mol%). MBs were made by sonication3, <strong>and</strong> fluorescent streptavidin was<br />

conjugated4. The viscosity of the lipid coating was determined by measuring the mobility of the<br />

fluorescent lipid using Fluorescence Recovery After Photobleaching (FRAP). Recordings were<br />

acquired before <strong>and</strong> after photobleaching a part of the MB coating. The speed of recovery formed<br />

the input of a Monte Carlo simulation resulting in the diffusion coefficient of the fluorescent lipid.<br />

The surface shear viscosity was analytically derived from the diffusion coefficient. We found a surface<br />

shear viscosity of 8×10-6 kg/s that was independent of the MB size, in line with values reported for<br />

the dilatation viscosity in MB dynamics studies5. In addition, we found heterogeneous lipid<br />

distributions in the coating which varied between MBs (see Figure) which suggests partly<br />

immiscibility of the lipids. In conclusion, this study shows that the static surface shear viscosity of the<br />

coating can be determined in an independent way which can now be used in MB dynamics models.<br />

References:<br />

1 Emmer, M., PhD thesis: The onset of bubble vibration, 2009; 2 Faez, T. et al., IEEE Ultras. Symp. Proc. 2010;<br />

3 Klibanov, A.L. et al., Invest. Radiol., 2004. 39: 187-95; 4 Lindner, J.R. et al., Circulation, 2001. 104: 2107-12; 5 van<br />

der Meer, S.M. et al., J. Acoust. Soc. Am., 2007. 121: 648-56.<br />

Additional authors: Klazina Kooiman *a , Marcia Emmer a , Tom J.A. Kokhuis a,b , Johan G. Bosch a , H.<br />

Martijn de Gruiter c , Martin E. van Royen c , Wiggert A. van Cappellen d , Adriaan B. Houtsmuller c ,<br />

Antonius F.W. van der Steen a,b , Nico de Jong a,b,e<br />

a Biomedical Engineering, c Pathology, d Reproduction <strong>and</strong> Development, Erasmus MC, the Netherl<strong>and</strong>s; b ICIN,<br />

the Netherl<strong>and</strong>s; e Physics of Fluids Group, Univ. of Twente, the Netherl<strong>and</strong>s<br />

Figure. Lipid distribution of coating of MBs recorded by 4Pi microscopy. Two typical views (1 <strong>and</strong> 2) out of a<br />

3D acquisition for a 3.0 µm (a) <strong>and</strong> 7.9 µm (b) MB. DSPE-PEG(2000)-biotin was labelled with fluorescent<br />

streptavidin whereas the other coating components DSPC <strong>and</strong> PEG-40 stearate were not fluorescently labelled.


<strong>Microbubble</strong> <strong>Symposium</strong>: <strong>Fabrication</strong>, <strong>Characterisation</strong> <strong>and</strong> Translational Applications 9<br />

4 th & 5 th July 2011<br />

Session 1: <strong>Microbubble</strong> <strong>Fabrication</strong> <strong>and</strong> <strong>Characterisation</strong><br />

Oral Presentation<br />

Eleanor Stride<br />

University College London<br />

Title: Magnetic microbubbles for localised imaging <strong>and</strong> drug delivery: development,<br />

characterisation <strong>and</strong> preliminary application in vivo<br />

Abstract: Coated microbubbles have now been in clinical use as contrast agents for<br />

ultrasound imaging for several decades. More recently, their use in therapeutic applications,<br />

in particular drug delivery <strong>and</strong> gene therapy, has also become a highly active area of<br />

research. There remain, however, some significant challenges which must be overcome in<br />

order to fully realise the potential of microbubbles in these applications. In particular, the<br />

difficulty in controlling the concentration of microbubbles at a given site <strong>and</strong> in ensuring<br />

sufficient proximity between bubbles <strong>and</strong> target cells, has frequently led to disappointing<br />

results from in vivo studies. Recent work has indicated that incorporating magnetic<br />

nanoparticles into the microbubble coating may provide an effective strategy for<br />

overcoming these challenges, by both enabling the bubbles to be localised using an<br />

externally applied magnetic field <strong>and</strong> enhancing the efficiency of cell uptake [1][2].<br />

Significant improvements in gene transfection efficiency have been demonstrated in vitro.<br />

Further investigation to fully underst<strong>and</strong> the mechanisms of enhancement <strong>and</strong> hence<br />

optimise the delivery protocols is however required <strong>and</strong> the present study is concerned with<br />

the detailed characterisation of the magnetic microbubbles <strong>and</strong> their application for gene<br />

transfection in vivo. The results from optical, acoustic <strong>and</strong> magnetic characterisation will be<br />

presented, including the successful manipulation of suspensions of magnetic bubbles subject<br />

to physiological flow conditions. Videos from high speed imaging used to investigate the<br />

dynamic behaviour of single microbubbles <strong>and</strong> the influence of the magnetic field on their<br />

response to an acoustic field will also be presented <strong>and</strong> compared with theoretical modelling<br />

conducted to support <strong>and</strong> interpret the experimental findings. Finally results demonstrating<br />

in vivo transfection in a mouse model will be presented confirming successful localisation of<br />

the transfection site.<br />

References:<br />

[1] Stride, E., Porter, C., Prieto, A. G., Pankhurst, Q. (2009) Ultrasound in Medicine <strong>and</strong> Biology, 35, 861-868; [2]<br />

Vlaskou, D., Mykhaylyk, O., Pradhan, P., Bergemann, C., Klibanov, A. L., Hensel, K., Schmitz, G., <strong>and</strong>, C. (2010)<br />

Human Gene Therapy, 21, 1429-1430


10 <strong>Microbubble</strong> <strong>Symposium</strong>: <strong>Fabrication</strong>, <strong>Characterisation</strong> <strong>and</strong> Translational Applications<br />

4 th & 5 th July 2011<br />

Session 1: <strong>Microbubble</strong> <strong>Fabrication</strong> <strong>and</strong> <strong>Characterisation</strong><br />

Oral Presentation<br />

Jonathan McKendry<br />

University of Leeds<br />

Title: Mechanical properties of surface modified microbubbles by atomic force microscopy<br />

Abstract: Atomic force microscopy (AFM) has been used to investigate the mechanical<br />

properties of phospholipid coated microbubbles <strong>and</strong> to quantify their stiffness. The<br />

mechanical properties were investigated using tipless AFM cantilevers to compress<br />

microbubbles attached to a gold surface in aqueous conditions. The phospholipid<br />

microbubbles were produced by microfluidic flow focusing <strong>and</strong> were found to have stiffness<br />

of 25 mNm -1 . The attachment of a streptavidin coating increased the microbubble stiffness<br />

by a factor of 30 to around 750 mNm -1 . Further, estimation of the frequency response based<br />

on values of stiffness obtained by force spectroscopy seem reasonable in comparison with<br />

those of an uncoated bubble <strong>and</strong> a PEG coated Bracco SonoVue BR14, suggesting that it may<br />

provide useful information in the development of novel microbubble coatings. It is hoped<br />

that the combination of AFM for measuring properties coupled with protein attachment for<br />

frequency response <strong>and</strong> targeting will lead to new design rules for the formulation of<br />

therapeutic microbubble contrast agents.<br />

Figure 1: Schematic showing a microbubble attached to a gold substrate via a biotin-streptavidin bridge <strong>and</strong><br />

being indented by a tipless AFM cantilever.<br />

Additional authors: Mr J. E. McKendry 1 , Dr C. Grant 1 Dr P. L. Coletta 2 , Dr J. A. Evans 3 , Prof S.<br />

D. Evans 1<br />

1. School of Physics <strong>and</strong> Astronomy, University of Leeds, Leeds, LS2 9JT; 2. Leeds Institute of Molecular<br />

Medicine, Wellcome Trust Brenner Building, St James’s Hospital, Leeds, LS9 7TF; 3. Leeds Institute of Genetics,<br />

Health <strong>and</strong> Theraputics, Worsley Building, University of Leeds, Leeds, LS2 9JT


<strong>Microbubble</strong> <strong>Symposium</strong>: <strong>Fabrication</strong>, <strong>Characterisation</strong> <strong>and</strong> Translational Applications 11<br />

4 th & 5 th July 2011<br />

Session 1: <strong>Microbubble</strong> <strong>Fabrication</strong> <strong>and</strong> <strong>Characterisation</strong><br />

Oral Presentation<br />

Ine Lentacker<br />

Ghent University<br />

Title: Update on microbubble <strong>and</strong> ultrasound assisted drug delivery within the Sonodrugs<br />

project.<br />

Abstract: The Sonodrugs project is an EU FP7 collaborative research project (NMP-4-LA-<br />

2008-213706) consisting of 14 partners. The objective of this project is to develop new drug<br />

carriers which can be activated for local drug release using focused ultrasound. One part of<br />

the project focuses on the design of pressure sensitive drug carriers (microbubbles) which<br />

can be imaged <strong>and</strong> locally activated with ultrasound. Another part focuses on thermosensitive<br />

drug carriers which can be activated by local heating using focused ultrasound<br />

under MRI image guidance.<br />

In this presentation we would like to give an overview of the results on microbubble <strong>and</strong><br />

ultrasound based drug delivery within the project. Firstly, we will present some data on the<br />

co-administration of drugs with polymer <strong>and</strong> lipid microbubbles using doxorubicin (DOX) <strong>and</strong><br />

Evans Blue as model drugs (1). These results clearly show enhanced drug delivery <strong>and</strong><br />

ultrasound induced extravasation in vitro <strong>and</strong> in vivo. In a second part, we discuss our work<br />

on the development of a pressure sensitive liposome loaded microbubble. We succeeded in<br />

coupling DOX containing liposomes (Doxil®-like) onto lipid microbubbles. In vitro data<br />

showed that this system is very efficient to enable DOX release <strong>and</strong> enhance DOX delivery<br />

upon ultrasound exposure. This was confirmed with a cell viability assay which showed that<br />

ultrasound exposure of DOX-liposomes attached to microbubbles resulted in a 3-fold higher<br />

toxicity compared to the DOX-liposomes alone (2). We further optimized this concept to<br />

obtain an easy to use, sterile <strong>and</strong> non-immunogenic formulation <strong>and</strong> proved that even very<br />

low DOX-liposome doses are sufficient to promote tumor cell killing after microbubble<br />

loading <strong>and</strong> ultrasound exposure (3). Furthermore this self-assembled liposome loaded<br />

microbubble system was characterized with the Br<strong>and</strong>aris microscope, showing expansion<br />

only behaviour of liposome loaded microbubbles even at low acoustic pressures. In vivo<br />

experiments with this new drug carrier are currently ongoing.<br />

References:<br />

(1) Böhmer M. et al. Focused ultrasound <strong>and</strong> microbubbles for enhanced extravasation. Journal of Controlled<br />

Release 2010, 148 (1), 18-24; (2) Lentacker I., et al. Design <strong>and</strong> evaluation of doxorubicin containing<br />

microbubbles for ultrasound triggered doxorubicin delivery: cytotoxicity <strong>and</strong> mechanisms involved. Molecular<br />

Therapy 2010, 18(1), 101-108; (3) Geers B., et al. Self-assembled liposome loaded microbubbles: The missing<br />

link for safe <strong>and</strong> efficient ultrasound triggered drug delivery. Journal of Controlled Release 2011, 152(2), 249-<br />

256.<br />

Additional authors: I. Lentacker 1 , B. Geers 1 , S. C. De Smedt 1 , M. Mühlmeister 2 , K. Tiemann 2 , Y.<br />

Luan 3 , N. De Jong 3 , J.-M. Escoffre, A. Bouakaz, M. Böhmer 5 , S. Vulto 5 , C. F. Sio 5<br />

1 Ghent Research Group on Nanomedicines, Ghent University, Belgium; 2 Westfälische Wilhelms-Universität<br />

Münster, Germany; 3 Erasmus MC Rotterdam, The Netherl<strong>and</strong>s.; 4 Université Francois Rabelais, Tours, France.<br />

5 Philips Research Europe, Eindhoven, The Netherl<strong>and</strong>s.


12 <strong>Microbubble</strong> <strong>Symposium</strong>: <strong>Fabrication</strong>, <strong>Characterisation</strong> <strong>and</strong> Translational Applications<br />

4 th & 5 th July 2011


<strong>Microbubble</strong> <strong>Symposium</strong>: <strong>Fabrication</strong>, <strong>Characterisation</strong> <strong>and</strong> Translational Applications 13<br />

4 th & 5 th July 2011<br />

Session 2: <strong>Microbubble</strong> Ultrasound <strong>Characterisation</strong><br />

Oral Presentation<br />

Title: Single microbubble acoustics<br />

Vassilis Sboros<br />

University of Edinburgh<br />

Abstract: Ultrasound microbubble (MB)-enhanced imaging is currently applied in the clinic<br />

for heart <strong>and</strong> liver diagnosis. The potential use of quantifying microvascular flow has been<br />

researched for over 20 years. More recent research has explored novel applications of MB<br />

technologies such as molecular imaging, enhancement of cell porosity, thrombolysis <strong>and</strong><br />

drug/gene delivery. The slow progress of the field may be attributed to the lack of high<br />

quality experimental data that enable a thorough underst<strong>and</strong>ing of MB behaviour under<br />

realistic ultrasound fields <strong>and</strong> in realistic in vivo experimental settings, which in turn will<br />

facilitate translation of research into novel in vivo tools.<br />

The necessity for investigating the acoustics of single MBs stems from the lack of a single or<br />

a predictable distribution of their acoustic responses. In other words investigations of MB<br />

clouds are limited in providing information on the individual scatter components, thus<br />

making difficult the comparison of experimental <strong>and</strong> theoretical data, but also an<br />

assessment of the performance of signal processing algorithms. Single MB acoustics<br />

measurements have provided high quality data that may advance MB theory <strong>and</strong> signal<br />

processing research.<br />

With the help of accurate calibration of MB scatter it is possible to observe <strong>and</strong> study<br />

physical phenomena such as resonance, the onset of transient cavitation, MB cracking, the<br />

different contributions of the shell, gas <strong>and</strong> environment including narrow tubing, <strong>and</strong> the<br />

various decay mechanisms.<br />

In addition pulse sequences available in commercial scanners can be assessed accurately. It<br />

is possible to capture large sample sizes of signal distributions <strong>and</strong> enable thorough signal<br />

processing analysis without the prerequisite of a model MB behaviour. It has been found<br />

that current pulse sequences, such as amplitude modulation, have a suboptimal operation as<br />

they exploit a small proportion of MBs, while they successfully cancel linear tissue echoes at<br />

low mechanical index imaging.<br />

In conclusion single MB acoustic measurements offer high quality data for the development<br />

of signal processing, improve the theoretical underst<strong>and</strong>ing of MB behaviour under well<br />

controlled experimental conditions, <strong>and</strong> can efficiently characterise different MB types,<br />

which is useful for the development of MB technologies.


14 <strong>Microbubble</strong> <strong>Symposium</strong>: <strong>Fabrication</strong>, <strong>Characterisation</strong> <strong>and</strong> Translational Applications<br />

4 th & 5 th July 2011<br />

Session 2: <strong>Microbubble</strong> Ultrasound <strong>Characterisation</strong><br />

Oral Presentation<br />

Verya Daeichin<br />

Thoraxcenter, Erasmus MC<br />

Title: Stimulating the subharmonic response of ultrasound contrast agents using selfdemodulation<br />

Abstract: Subharmonic (SH) emission from ultrasound contrast agent (UCA) is of interest since this is<br />

produced only by the UCA <strong>and</strong> is free of artifacts produced in harmonic imaging modes. In this work<br />

we study the use of self-demodulation (S-D) signal as a means of microbubble excitation at the SH<br />

frequency in order to enhance the SH emission of UCA. The S-D wave is a low-frequency signal<br />

produced by nonlinear propagation of an ultrasound wave in the medium <strong>and</strong> it can be<br />

approximated by the second time derivative of the squared envelope of the transmit pulse. A diluted<br />

population (1:10000) of Definity UCA (Bristol Myers Squibb, Boston, MA, USA) was insonified by a 10<br />

MHz single element transducer focused at 76mm. The scattered signals were collected by another 10<br />

MHz transducer. The transmitted 10 MHz wave with a low acoustic pressure of 50 kPa (peak<br />

negative) had a burst length of 6 <strong>and</strong> 20 cycles which was multiplied with the envelope function: E( t<br />

) = exp[- ( 2t/T)^2M] where T is the nominal duration of the pulse <strong>and</strong> the integer M determines the<br />

rise <strong>and</strong> decay time of the envelope (see figure below). The center frequency of the S-D signal is<br />

changing from low frequencies (around 1 MHz) towards the transmitted frequency (10 MHz) by<br />

modifying the envelope function from Gaussian (M=1) to rectangular (M=15). The amplitude of S-D<br />

signal is at its maximum 40 dB below the fundamental transmit amplitude. In order to look at the<br />

true enhancement of SH emission in frequency domain <strong>and</strong> knowing that the S-D wave is located at<br />

the beginning <strong>and</strong> the end of the received signal, the analysis was performed in a time window<br />

discarding 3 periods at 10 MHz from the beginning <strong>and</strong> the end of the 20 cycle burst (see figure<br />

below). For both 6 <strong>and</strong> 20 transmitted cycles, the SH response is increased up to 20 dB because of<br />

the S-D stimulation when M is equal to 15 (rectangular envelope). A suitable design of the envelope<br />

of the transmit excitation to generate a S-D signal at the SH frequency can enhance the SH emission<br />

of UCA. This study suggests the SH imaging is feasible with low cycle-number transmit burst <strong>and</strong> low<br />

acoustic pressure (~50 KPa).<br />

Normalized Scattered Pressure (dB)<br />

0<br />

-20<br />

-40<br />

-60<br />

-80<br />

-100<br />

-120<br />

Spectrum of the signal scattered by the UCA<br />

-0.5<br />

M = 1<br />

0 0.5 1 1.5 2 2.5 3<br />

x 10 5<br />

5 10 15 20 25<br />

Frequency (MHz)<br />

Influence of the S-D signal on the spectrum of the signal scattered by UCA insonified by a 20cycle<br />

burst of 10 MHz.<br />

This research was supported by the Center for Translational Molecular Medicine <strong>and</strong> the Netherl<strong>and</strong>s Heart Foundation (PARISK).<br />

1<br />

0.5<br />

0<br />

-1<br />

1<br />

0.5<br />

0<br />

M = 3<br />

-0.5<br />

Excitation Burst<br />

0 0.5 1 1.5 2 2.5 3<br />

x 10 5<br />

-1<br />

1<br />

0.8<br />

0.6<br />

0.4<br />

M = 0.215<br />

0<br />

-0.2<br />

-0.4<br />

-0.6<br />

-0.8<br />

-1


<strong>Microbubble</strong> <strong>Symposium</strong>: <strong>Fabrication</strong>, <strong>Characterisation</strong> <strong>and</strong> Translational Applications 15<br />

4 th & 5 th July 2011<br />

Session 2: <strong>Microbubble</strong> Ultrasound <strong>Characterisation</strong><br />

Oral Presentation<br />

Robert Eckersley<br />

Imperial College<br />

Title: Quantitative contrast-enhanced ultrasound imaging: pitfalls, challenges, <strong>and</strong> advances.<br />

Abstract: Ultrasound imaging of microbubble contrast agents provides the potential to<br />

obtain quantitative information relating to blood supply <strong>and</strong> tissue function. This technique<br />

is showing great promise for diagnosis <strong>and</strong> monitoring clinical conditions such as<br />

cardiovascular diseases <strong>and</strong> cancer, with considerable potential benefits in terms of patient<br />

care.<br />

A key challenge of this technique is the existence of significant variations in the imaging<br />

results, <strong>and</strong> the lack of underst<strong>and</strong>ing regarding their origin.<br />

In this presentation the potential sources of variability in the quantification of tissue<br />

perfusion based on microbubble contrast-enhanced ultrasound images will be presented<br />

<strong>and</strong> discussed. These are divided into the following three categories: (i) factors relating to<br />

the set up of the ultrasound scanner, including transmission power, focal depth, dynamic<br />

range, signal gain <strong>and</strong> ultrasound frequency, (ii) factors relating to the patient, which include<br />

body physical differences, physiological interaction with the microbubbles, propagation <strong>and</strong><br />

attenuation through tissue, <strong>and</strong> tissue motion, <strong>and</strong> (iii) factors relating to the microbubbles,<br />

including the type of bubbles <strong>and</strong> their stability, preparation, injection technique <strong>and</strong><br />

dosage.<br />

It has been shown that the factors in each category can significantly affect the imaging<br />

results <strong>and</strong> contribute to the variations observed. How these factors influence quantitative<br />

imaging is explained <strong>and</strong> possible methods for reducing such variations are discussed.<br />

Additional authors: R. J. Eckersley 1 , E. Stride 4 , H. Mulvana 1 , T. Gauthier 3 , A. K. P. Lim 1 , D. O.<br />

Cosgrove 1 , <strong>and</strong> M.-X. Tang 2<br />

1 Imaging Sciences Department, Faculty of Medicine, Imperial College London. 2 Department of Bioengineering,<br />

Imperial College London. 3 Department Experimental Medicine <strong>and</strong> Toxicology, Imperial College London. 4<br />

Department Mechanical Engineering, University College London.


16 <strong>Microbubble</strong> <strong>Symposium</strong>: <strong>Fabrication</strong>, <strong>Characterisation</strong> <strong>and</strong> Translational Applications<br />

4 th & 5 th July 2011<br />

Session 2: <strong>Microbubble</strong> Ultrasound <strong>Characterisation</strong><br />

Oral Presentation<br />

Title: Release <strong>and</strong> characterization of a single bubble<br />

Serge Mensah<br />

CNRS<br />

Abstract: During hyperbaric decompression (diving or hyperbaric medicine), both the absolute<br />

ambient <strong>and</strong> the absolute inspired pressures are reducing; (5-100 µm) bubbles may be generated<br />

from pre-existing gas nuclei (0.1-5 µm). An accurate monitoring of the size <strong>and</strong> of the density of the<br />

bubble (including the nuclei) population will provide a valuable means to underst<strong>and</strong> the nucleation<br />

<strong>and</strong> growth processes in various supersaturated tissues. In this aim, an ultrasonic characterization<br />

method based on a dual frequency technique [1] applied on a free single bubble is tested.<br />

In order to design <strong>and</strong> evaluate the accuracy of a free single bubble sizing technique operating in<br />

water, bubbles of different diameters tethered on a wire have been set free by using high amplitude<br />

pressure waves. Then, two ultrasound focused waves impinge simultaneously on each microbubble:<br />

a low frequency chirp (20 kHz


<strong>Microbubble</strong> <strong>Symposium</strong>: <strong>Fabrication</strong>, <strong>Characterisation</strong> <strong>and</strong> Translational Applications 17<br />

4 th & 5 th July 2011<br />

Session 2: <strong>Microbubble</strong> Ultrasound <strong>Characterisation</strong><br />

Oral Presentation<br />

Title: Observations of microbubble cavitation under optical trap control<br />

Paul Campbell<br />

University of Dundee<br />

Abstract: <strong>Microbubble</strong>s are held up as a potential next generation 'theranostic' agent -<br />

exhibiting clear therapeutic characteristics in terms of their ability to permeabilize tissue <strong>and</strong><br />

individual cell membranes for the purposes of either drug delivery or to induce distinct<br />

bioeffects, but also retaining a critical diagnostic capability whilst in their intact state. We<br />

have been studying microbubbles at Dundee for several years now, initially using optical trap<br />

control to isolate single microbubbles for precision observation at distinct proximities from<br />

individual cells. More recently, we have attempted to seed spatially controlled free bubble<br />

cavitation events, for the purposes of achieving enhanced transfection in vitro, via the laser<br />

ignition of optically trapped nanoparticles.<br />

This talk will thus serve as an introduction to the optical trap approach to cavitation in<br />

general, so that others might easily replicate our procedures, <strong>and</strong> will also provide an<br />

overview of the main results that we have achieved for both ultrasonically activated shelled<br />

microbubbles, as well as free microbubbles generated via laser ignition of optically trapped<br />

nanoparticles.


18 <strong>Microbubble</strong> <strong>Symposium</strong>: <strong>Fabrication</strong>, <strong>Characterisation</strong> <strong>and</strong> Translational Applications<br />

4 th & 5 th July 2011


<strong>Microbubble</strong> <strong>Symposium</strong>: <strong>Fabrication</strong>, <strong>Characterisation</strong> <strong>and</strong> Translational Applications 19<br />

4 th & 5 th July 2011<br />

Session 3: <strong>Microbubble</strong> Translational Applications<br />

Oral Presentation<br />

Title: <strong>Microbubble</strong>s for molecular imaging <strong>and</strong> theranostics<br />

Fabian Kiessling<br />

University of Aachen<br />

Abstract: Ultrasound is one of the workhorses in clinical cancer diagnosis. In particular, it is<br />

routinely used to characterize lesions in liver, urogenital tract, head <strong>and</strong> neck <strong>and</strong> soft<br />

tissues. <strong>Microbubble</strong>s were introduced as intravascular contrast agents <strong>and</strong> can be detected<br />

with superb sensitivity <strong>and</strong> specificity using contrast specific imaging modes. By conjugating<br />

biomolecules to the microbubble surface molecular ultrasound imaging becomes feasible.<br />

In this talk our experiences in fabricating <strong>and</strong> using microbubbles for targeted imaging <strong>and</strong><br />

for theranostics are reported. Examples will be given for the use of soft <strong>and</strong> PBCA-based<br />

hard-shell MBs for tumour characterisation <strong>and</strong> therapy response monitoring. For example,<br />

it will be shown that the assessment of the VEGFR2-expression on the tumour<br />

vascularisation is better suited for the discrimination of low <strong>and</strong> high aggressive breast<br />

cancers compared to functional information about the relative tumour blood volume. On the<br />

other h<strong>and</strong>, it will also be shown that the normalisation of the molecular ultrasound data to<br />

the relative blood volume often is necessary to distinguish between change in vessel density<br />

<strong>and</strong> change in molecular marker expression on the vasculature. However, the use of<br />

microbubbles is not restricted to ultrasound. By incorporating dyes <strong>and</strong> superparamagentic<br />

iron oxide nanoparticles into the microbubble shell multimodal diagnostics can be achieved.<br />

In this context, we will show that these microbubbles can excellently be detected by MRI<br />

<strong>and</strong> that their disintegration by ultrasound leads to a changed MR contrast. It will further be<br />

shown that contrast-enhanced ultrasound mediated vascular permeation can favourably be<br />

monitored with these multimodal probes. In summary, microbubbles are versatile diagnostic<br />

<strong>and</strong> theranostic agents, which can be used not only for ultrasound but also for optical<br />

imaging <strong>and</strong> MRI.


20 <strong>Microbubble</strong> <strong>Symposium</strong>: <strong>Fabrication</strong>, <strong>Characterisation</strong> <strong>and</strong> Translational Applications<br />

4 th & 5 th July 2011<br />

Session 3: <strong>Microbubble</strong> Translational Applications<br />

Oral Presentation<br />

Gemma Marston<br />

University of Leeds<br />

Title: Imaging the development of functional tumour vasculature in vivo in preclinical models<br />

Abstract:<br />

Background<br />

Angiogenesis is essential for tumours to grow beyond a few millimetres in size <strong>and</strong> to metastasise,<br />

making it an attractive target for both cancer imaging <strong>and</strong> therapeutic intervention. Contrast<br />

enhanced high-frequency ultrasound (CE HFUS) enables longitudinal studies in mouse models <strong>and</strong><br />

can be used to investigate both qualitative <strong>and</strong> quantitative changes in tumour vasculature including<br />

tumour blood flow <strong>and</strong> perfusion. We have developed CE HFUS protocols to characterise the<br />

development of functional tumour vasculature in vivo in a murine xenograft model of colorectal<br />

cancer.<br />

Method<br />

10 female CD1 Nu/Nu mice implanted with SW480 human colorectal cancer cell xenografts were<br />

imaged with CE HFUS 7 times between day 7 <strong>and</strong> day 28 of tumour growth using the VisualSonics<br />

Vevo 770 system. At each time point a single bolus of microbubbles was injected using a syringe<br />

driver into the tail vein during 2D CE HFUS imaging, with the wash-in data loops captured prior to 3D<br />

CE HFUS imaging.<br />

Results<br />

Time intensity curves were generated post-acquisition <strong>and</strong> used to determine perfusion kinetics <strong>and</strong><br />

vascular data. Tumour vascular space <strong>and</strong> tumour volume were calculated from 3D CE HFUS images.<br />

This showed that the percentage tumour vascular space decreased significantly as the tumour<br />

volume increased. Furthermore, smaller tumours had both faster rates of blood flow <strong>and</strong> significantly<br />

higher maximum levels of vascular perfusion than larger tumours.<br />

Conclusion<br />

This study shows that CE HFUS is a powerful tool for analysis of tumour vascular development in vivo<br />

<strong>and</strong> provides a relatively non-invasive method to assess development of anti-angiogenic therapies<br />

<strong>and</strong> response to treatment in pre-clinical trials.


<strong>Microbubble</strong> <strong>Symposium</strong>: <strong>Fabrication</strong>, <strong>Characterisation</strong> <strong>and</strong> Translational Applications 21<br />

4 th & 5 th July 2011<br />

Session 3: <strong>Microbubble</strong> Translational Applications<br />

Oral Presentation<br />

Philippe Trochet<br />

VisualSonics<br />

Title: VevoCQ Software allows advanced quantification of perfusion kinetics parameters with<br />

consistent, reliable <strong>and</strong> actionable data<br />

Abstract: VevoCQ is a powerful addition to nonlinear contrast imaging functionality on the<br />

Vevo® 2100 imaging system. This software allows for the study of contrast uptake kinetics as<br />

well as late phase targeted enhancement. It provides advanced curve fitting algorithms for<br />

quantitative assessment of perfusion parameters as well as color-coded parametric images<br />

useful for qualitative assessment of the spatial distribution of the same parameters. The<br />

software is a post-processing tool which can be used on numerous tissues, organs <strong>and</strong> tumor<br />

models, including subcutaneous tumors, abdominal organs <strong>and</strong> hind limb muscles.


22 <strong>Microbubble</strong> <strong>Symposium</strong>: <strong>Fabrication</strong>, <strong>Characterisation</strong> <strong>and</strong> Translational Applications<br />

4 th & 5 th July 2011<br />

Session 3: <strong>Microbubble</strong> Translational Applications<br />

Oral Presentation<br />

Sarah Fawcett<br />

University of Cambridge<br />

Title: Our early experience of pre clinical in vivo use of MicroMarker – a) sonoporation for in<br />

vivo gene delivery <strong>and</strong> b) contrast enhanced imaging in tumours after anti-vascular therapy<br />

Abstract: We have attempted to use MicroMarker mediated sonoporation for in vivo<br />

delivery of DNA. Using this approach I will present our pilot data in which we demonstrate<br />

sustained expression of a gene reporter, several weeks after DNA injection into the skin.<br />

In addition we have started to investigate the potential for contrast enhanced ultrasound<br />

imaging for monitoring tumour response to anti-vascular agents <strong>and</strong> comparing these data<br />

with those obtained from DCE-MRI imaging.


<strong>Microbubble</strong> <strong>Symposium</strong>: <strong>Fabrication</strong>, <strong>Characterisation</strong> <strong>and</strong> Translational Applications 23<br />

4 th & 5 th July 2011<br />

Session 3: <strong>Microbubble</strong> Translational Applications<br />

Oral Presentation<br />

Title: Bubbles in the clinical setting<br />

Jane Smith (Bates)<br />

St James’s University Hospital<br />

Abstract: US contrast agents are now an important part of the diagnostic process, <strong>and</strong> a<br />

recognised problem-solving tool.<br />

The main (non-cardiac) use hinges around lesion characterisation in the liver, with patterns<br />

of contrast take-up allowing the operator to establish a definitive diagnosis. Detection of<br />

malignant lesions in the liver is also a useful application, particularly in patients at high risk<br />

of liver metastases. Other uses include trauma, monitoring of treatments (such as ablation)<br />

<strong>and</strong> guiding targeted biopsies in the liver.<br />

The advantages of bubbles in the clinical field are established; it is a safe, cost effective <strong>and</strong> a<br />

reliable addition to our available diagnostic tools. It has high patient acceptability <strong>and</strong>,<br />

importantly, allows the operator to make a diagnosis which is immediately available -<br />

shortening the diagnostic pathway <strong>and</strong> reducing or eliminating patient anxiety.<br />

This talk will outline some of the common clinical applications <strong>and</strong> current capabilities of<br />

commercially available bubbles.


24 <strong>Microbubble</strong> <strong>Symposium</strong>: <strong>Fabrication</strong>, <strong>Characterisation</strong> <strong>and</strong> Translational Applications<br />

4 th & 5 th July 2011<br />

Number Name Title<br />

1 Mostafa Abdelrahman<br />

2 Radwa Abou-Saleh<br />

3 Muhammad Arif<br />

4 Jonathan Blockley<br />

5 Matthew Booth<br />

6 Christian Buchcic<br />

7 Joanne Burke<br />

Poster Presentations<br />

Optimizing contrast-enhanced ultrasound technique for<br />

mice tumour imaging: choosing the right transducer<br />

<strong>and</strong> bubble concentration<br />

Probing the effect of poly(ethylene glycol) on the<br />

mechanical behavior of streptavidin <strong>and</strong> quantum dot<br />

coated microbubbles<br />

Chirp coded excitation with fractional Fourier transform<br />

for ultrasound harmonic imaging<br />

Ultrasound measurements of trapped microbubbles on<br />

a st<strong>and</strong>ard microscope stage<br />

The optical properties <strong>and</strong> stability of cadmium free<br />

quantum dots<br />

<strong>Microbubble</strong>s as functional ingredients in complex food<br />

systems<br />

Interfacial study of class II hydrophobin <strong>and</strong> β-casein<br />

mixtures: relationship to bubble stability<br />

8 Caroline Harfield Opto-acoustic trapping of microbubbles<br />

9 Sevan Harput<br />

10 Sevan Harput<br />

11 George Heath Controlling actin filaments<br />

12 Nicola Ingram<br />

13 Gaël Léauté<br />

Periodic clustering of microbubbles by secondary<br />

radiation force<br />

Effect of nonlinear frequency modulated signals on<br />

subharmonic emission from microbubbles<br />

Pre-clinical evaluation of VEGFR2 as an imaging<br />

biomarker <strong>and</strong> targeting molecule for therapeutic<br />

delivery.<br />

Effect of excitation pulses on the sonoporation<br />

efficiency of mono- <strong>and</strong> polydisperse ultrasound<br />

contrast agents


<strong>Microbubble</strong> <strong>Symposium</strong>: <strong>Fabrication</strong>, <strong>Characterisation</strong> <strong>and</strong> Translational Applications 25<br />

4 th & 5 th July 2011<br />

14 Jonathan Loughran<br />

15 Paul Mackin<br />

16 Richard O'Rorke<br />

17 Sally Peyman<br />

18 Benjamin Raiton<br />

19 Charles Sennoga<br />

20 Peter Smith<br />

Selective imaging of adherent microbubbles with<br />

correction of tissue motion<br />

Non-Invasive ultrasound imaging in a mouse model of<br />

pancreatic ductal adenocarcinoma<br />

Trapping microbubbles in a moveable acoustic array<br />

using surface acoustic waves<br />

3D exp<strong>and</strong>ing geometry for improved on-chip<br />

microbubble production rates<br />

Counter flow microbubble channelling using acoustic<br />

radiation force funnel<br />

Comparision of methods for sizing <strong>and</strong> counting of<br />

microbubbles<br />

Pre-distorted chirp excitations for polydisperse<br />

microbubble populations


26 <strong>Microbubble</strong> <strong>Symposium</strong>: <strong>Fabrication</strong>, <strong>Characterisation</strong> <strong>and</strong> Translational Applications<br />

4 th & 5 th July 2011<br />

Poster Number: 1<br />

Mostafa Abdelrahman<br />

University of Leeds<br />

Title: Optimizing contrast-enhanced ultrasound technique for mice tumour imaging:<br />

choosing the right transducer <strong>and</strong> bubble concentration<br />

Abstract: High-frequency contrast-enhanced ultrasound (HF-CEUS) has been shown to be a<br />

valuable tool in the assessment of tumour angiogenesis in mice. However, this technique is<br />

still to be optimized <strong>and</strong> calibrated. Forty MHz is the most commonly used frequency in mice<br />

CEUS imaging due to the high spatial resolution it provides. However, 25 MHz frequency<br />

provides more penetration power <strong>and</strong> is closer to the resonance frequency of bubbles of the<br />

sizes used in mice imaging (~3MHz). In this study the response of 40 MHz <strong>and</strong> 25 MHz<br />

transducers to contrast was investigated.<br />

Both transducers were used to image contrast added to blood-mimicking fluid (BMF) at<br />

different concentrations (5 x 10 5 - 5 x 10 7 bubble/ml). The contrast-BMF mixture was driven<br />

through a flow phantom using an infusion pump at flow velocities of 1, 2, <strong>and</strong> 3 mm/s.<br />

Greyscale intensity was used as a measure for signal enhancement from bubbles.<br />

The results showed that the 25 MHz was 1.5 times more sensitive to contrast than the 40<br />

MHz. It was also found that the effect of increasing contrast concentration is less important<br />

at concentrations more than 1.25 x 10 7 bubbles/ml for both transducers. Finally, it was<br />

shown that flow velocity has no significant effect on contrast detectability.<br />

In conclusion, based on the results of this study it is recommended to use ultrasound at 25<br />

MHz frequency instead of 40 MHz in CEUS studies, <strong>and</strong> it is possible to use lower bubble<br />

concentrations without significantly reducing the signal.


<strong>Microbubble</strong> <strong>Symposium</strong>: <strong>Fabrication</strong>, <strong>Characterisation</strong> <strong>and</strong> Translational Applications 27<br />

4 th & 5 th July 2011<br />

Poster Number: 2<br />

Radwa Abou-Saleh<br />

University of Leeds<br />

Title: Probing the effect of poly(ethylene glycol) on the mechanical behavior of streptavidin<br />

<strong>and</strong> quantum dot coated microbubbles<br />

Abstract: The use of microbubbles as targeted delivery vehicles has been receiving wide<br />

spread interest for a range of medical applications. Potentially ultrasound can be used for<br />

localized delivery that is by rupturing bounded microbubbles in a specific area <strong>and</strong> introduce<br />

payloads into cells via sonoporation. The frequency response of microbubbles to ultrasound<br />

excitation mainly depends on the mechanical properties of the shell[1] which can be<br />

determined using AFM[2]. Here, we perform force spectroscopy measurements-with tipless<br />

cantilever on encapsulated microbubbles to characterize the shell stiffness of a range of<br />

microbubble coatings, specifically, Streptavidin <strong>and</strong> Quantum dot coated microbubbles.<br />

Forces ranging from 20-100nN were applied to single bubbles to determine the change in<br />

stiffness with applied force. In addition, the same coatings were tested in the presence <strong>and</strong><br />

absence of a Poly(ethylene glycol) (PEG) layer with the same protocol. It was observed that<br />

the shell stiffness of phospholipid microbubbles increases linearly with increasing force.<br />

Adding a streptavidin coat doubles the stiffness values of the microbubble, with a dramatic<br />

increase observed with addition of Quantum dots leading to a 3x increase in shell stiffness.<br />

Moreover, PEG-ylating the bubble shell led to distinct compression behaviour in the forcedistance<br />

curve <strong>and</strong> presents a characteristic non-linear increase of stiffness in the presence<br />

of streptavidin <strong>and</strong> Quantum dot coatings.<br />

References:<br />

1. Ferrara, K., R. Pollard, <strong>and</strong> M. Borden, Ultrasound microbubble contrast agents: fundamentals <strong>and</strong><br />

application to gene <strong>and</strong> drug delivery. Annu Rev Biomed Eng, 2007. 9: p. 415-47.<br />

2. Sboros, V., et al., Nanomechanical probing of microbubbles using the atomic force microscope.<br />

Ultrasonics, 2007. 46(4): p. 349-54.<br />

Additional authors:<br />

Radwa H. Abou-Saleh, Jono E. McKendry, Sally A. Peyman, Neil H. Thomson <strong>and</strong> Stephen D.<br />

Evans<br />

School of Physics <strong>and</strong> Astronomy, University of Leeds, LS2 9JT, UK<br />

Phopholipid molecule<br />

PEG chain<br />

Biotin molecule


28 <strong>Microbubble</strong> <strong>Symposium</strong>: <strong>Fabrication</strong>, <strong>Characterisation</strong> <strong>and</strong> Translational Applications<br />

4 th & 5 th July 2011<br />

Poster Number: 3<br />

Muhammad Arif<br />

University of Leeds<br />

Title: Chirp coded excitation with fractional Fourier transform for ultrasound harmonic<br />

imaging<br />

Abstract: Medical ultrasound imaging techniques such as tissue harmonic imaging <strong>and</strong><br />

contrast-enhanced harmonic imaging provides better spatial <strong>and</strong> contrast resolution by<br />

producing the image with the second harmonic component (SHC) of the nonlinear received<br />

signal. The extraction of the SHC from the received signal can be done by linear b<strong>and</strong>-pass<br />

filtering technique; however it works well only for a narrow-b<strong>and</strong>width signal in which the<br />

SHC is not overlapped with the fundamental component. A multi-pulse detection scheme<br />

such as pulse inversion (PI) can be used to extract the overlapped SHC; however it suffers<br />

under motion artifacts <strong>and</strong> reducing the system frame-rate. In this paper, fractional Fourier<br />

transform (FrFT) is proposed as a filtering tool, with wide-b<strong>and</strong>width chirp coded excitation,<br />

for the extraction of the overlapped SHC.<br />

Simulation <strong>and</strong> experiments were performed in order to validate the proposed method. A<br />

Hann windowed linear chirp with duration of 10 us, <strong>and</strong> b<strong>and</strong>width ranging from 1-3 MHz<br />

was used as an excitation signal. The nonlinear received signals were processed using the<br />

FrFT in order to extract the SHC. The extracted SHC is then processed using the harmonic<br />

matched filter in order to perform second harmonic pulse compression (SHPC) <strong>and</strong> to<br />

restore the axial resolution.<br />

Both simulation <strong>and</strong> experimental results indicate at least a 13 dB improvement in the range<br />

sidelobes level of the FrFT filtered compressed SHC when compared with the unfiltered<br />

compressed SHC as shown in the enclosed figure.<br />

Additional authors:<br />

Muhammad Arif, Sevan Harput, Peter Smith, David Cowell, <strong>and</strong> Steven Freear<br />

Ultrasound Group School of Electronic <strong>and</strong> Electrical Engineering, University of Leeds, Leeds LS2 9JT


<strong>Microbubble</strong> <strong>Symposium</strong>: <strong>Fabrication</strong>, <strong>Characterisation</strong> <strong>and</strong> Translational Applications 29<br />

4 th & 5 th July 2011<br />

Poster Number: 4<br />

Jonathan Blockley<br />

Institution: University of Leeds<br />

Title: Ultrasound measurements of trapped microbubbles on a st<strong>and</strong>ard microscope stage<br />

Abstract: It can be difficult to be certain what the size <strong>and</strong> position distributions of<br />

microbubble populations are when making ultrasound measurements. Depending on the<br />

method of production, a broad spread of sizes can result in a small portion of the population<br />

dominating an ultrasound signal, so unpicking the effects of the spread of sizes is a<br />

challenge. Existing methods to negate these population effects, or at least obtain repeatable<br />

population distributions include mixing <strong>and</strong> pumping through tubes to simulate transport in<br />

the vascular system, both of which involve fluid motion. Alternatively, clear results can be<br />

obtained by testing single bubbles; this technique requires very high sensitivity <strong>and</strong><br />

extrapolation of data to the population response. Secondary issues in both cases include the<br />

methods of bubble isolation, which include confinement in tubes, optical trapping <strong>and</strong> use of<br />

ultrasound radiation force. Some of these methods introduce complex boundary effects.<br />

Here, an apparatus has been designed to allow ultrasound measurements to be made on a<br />

small population of microbubbles that will be simultaneously imaged with a st<strong>and</strong>ard upright<br />

optical microscope. The microbubbles will be contained within a thin slice of fluid <strong>and</strong> enter<br />

the observation area from below, making use of their buoyancy force. Ultrasound<br />

measurements will be made as the microbubbles float through the observation area. The<br />

optical microscope can be focused at a depth of choice to image the microbubbles as they<br />

rise, or survey the microbubbles that were insonated after they have risen to the top of the<br />

chamber. Thus the size distribution <strong>and</strong> concentration of the microbubbles can be<br />

ascertained in situ. Bubbles of various sizes could be trapped within the observation area<br />

<strong>and</strong> their acoustic responses matched to the observed size distribution of the population.<br />

This system also has the advantage that the bubbles can be tested at a distance 100 to 1000<br />

times their radius from any surface, with the absence of boundaries minimising reflections<br />

<strong>and</strong> allowing maximum sensitivity.


30 <strong>Microbubble</strong> <strong>Symposium</strong>: <strong>Fabrication</strong>, <strong>Characterisation</strong> <strong>and</strong> Translational Applications<br />

4 th & 5 th July 2011<br />

Poster Number: 5<br />

Title: The optical properties <strong>and</strong> stability of cadmium free quantum dots<br />

Matthew Booth<br />

University of Leeds<br />

Abstract: Quantum dots (QDs) have been predicted to make a big impact in the<br />

development of biomedical imaging [1] <strong>and</strong> significant progress has been made with QDs<br />

being used for targeted imaging in small animals [2]. However, the majority of biomedical<br />

QD studies to date have utilised cadmium based QDs <strong>and</strong> this raises important questions<br />

regarding toxicity [3]. In order to develop a QD system that could be potentially introduced<br />

into a clinical environment the semiconductor must be cadmium free. In this study, the<br />

photoluminescence (PL) quantum efficiency of heavy metal free CuInS2/ZnS quantum dots in<br />

aqueous solution is investigated. CuInS2 possesses a narrow b<strong>and</strong> gap enabling biologically<br />

important near infra red emission to be easily achieved, <strong>and</strong> complete surface passivation<br />

with ZnS results in high quantum efficiency. We consider the PL response to changes in pH,<br />

temperature <strong>and</strong> buffer in order to quantify their behaviour in the biological environment.<br />

Importantly we also examine the biocompatibility <strong>and</strong> long term photostability, providing<br />

useful information for the application of these promising quantum dots in biological assays.<br />

The role that surface chemistry has in providing both the stability in solution <strong>and</strong> the<br />

potential for bioconjugation required for biological applications proves to be significant [4].<br />

References:<br />

[1] I. Medintz, H. Uyeda, E. Goldman et al. Nature Materials, 4, 6 (2009), 435-446<br />

[2] A. Hoshino, K. Hanaki, K. Suzuki et al. Biochem. <strong>and</strong> Biophys. Res. Comm. 314, 1 (2004) 46-53<br />

[3] B. Rzigalinski, J. Strobl, Toxicology <strong>and</strong> Applied Pharmacology, 238 (2009), 280-288<br />

[4] J. Weng, J. Ren, Current Medicinal Chemistry, 13, 8 (2006), 897-909<br />

Additional authors:<br />

Matt Booth <strong>and</strong> Kevin Critchley<br />

School of Physics <strong>and</strong> Astronomy, University of Leeds, UK


<strong>Microbubble</strong> <strong>Symposium</strong>: <strong>Fabrication</strong>, <strong>Characterisation</strong> <strong>and</strong> Translational Applications 31<br />

4 th & 5 th July 2011<br />

Poster Number: 6<br />

Title: <strong>Microbubble</strong>s as functional ingredients in complex food systems<br />

Christian Buchcic<br />

Wageningen UR<br />

Abstract: Besides the utilization of microbubble technology in areas like medical<br />

engineering, a vast amount of other possible applications can be envisaged. <strong>Microbubble</strong>s<br />

have also found attention of the Food Science Community. Our vision is to apply<br />

microbubbles as functional ingredients in complex food systems in order to modify their<br />

textural <strong>and</strong> sensorial properties. The challenge is to find relations between characteristics<br />

of the shell material, the microbubbles, <strong>and</strong> their functionality in food systems.<br />

Additional authors:<br />

Christian Buchcic 1, 2 , Tijs Rovers<br />

2, 3<br />

1<br />

Wageningen UR, Physical Chemistry <strong>and</strong> Colloid Science, P.O. Box 8038, 6700 EK Wageningen, The<br />

Netherl<strong>and</strong>s<br />

2<br />

TI Food <strong>and</strong> Nutrition, P.O. Box 557, 6700 AN Wageningen, The Netherl<strong>and</strong>s<br />

3<br />

Wageningen UR, Physics <strong>and</strong> Physical Chemistry of Foods, P.O. Box 8129, 6700 EV Wageningen, The<br />

Netherl<strong>and</strong>s


32 <strong>Microbubble</strong> <strong>Symposium</strong>: <strong>Fabrication</strong>, <strong>Characterisation</strong> <strong>and</strong> Translational Applications<br />

4 th & 5 th July 2011<br />

Poster Number: 7<br />

Joanne Burke<br />

University of Leeds<br />

Title: Interfacial study of class II hydrophobin <strong>and</strong> β-casein mixtures: relationship to bubble<br />

stability<br />

Abstract: Class II Hydrophobin (HFBII) is a highly surface active molecule <strong>and</strong> in the context<br />

of aeration can be considered to be an air structuring protein conferring exceptional stability<br />

to foams. Recently, Cox et al [1,2] demonstrated that HFBII can produce liquid foams which<br />

do not coarsen <strong>and</strong> are stable for periods of several months, which is far in excess than that<br />

obtained with any other commonly used proteins. This is of interest to the food industry,<br />

since producing shelf stable foams in food formulations is very difficult. Although HFBII,<br />

when used alone, has proven to be very promising in terms of foam stability, a greater<br />

underst<strong>and</strong>ing is required in order to optimise formulations <strong>and</strong> stability in real food<br />

systems. Factors which have been explored for this poster include the concentration of HFBII<br />

(low amounts 10 -4 wt %) as well as the effect of pH on the surface shear viscosity which both<br />

seem to be very significant. Furthermore, from a practical point of view, mixtures of HFBII<br />

with other commonly used foaming agents should be tested in order to obtain information<br />

of more direct relevence to complex food systems since it is unknow as to wether HFBII will<br />

maintain its functionality in such systems or if synergystic effects will occur. Therefore this<br />

poster will also describe the surface rheological properties of protein films formed from<br />

mixtures of HFBII with β-casein.<br />

References:<br />

[1] Cox, A.; Cagnol. F.; Russell, A.B.; Izzard, M.J. Langmuir 2007, 23, 7995-8002.<br />

[2] Cox, A.: Aldred, D.; Russell, A.B. Food Hydrocolloids 2009, 23, 366-376.<br />

Additional authors:<br />

Burke, J 1* , Murray, B 1 , Cox, A 2 <strong>and</strong> Petkov, J 3<br />

1 Food Colloids Group, School of Food Science <strong>and</strong> Nutrition, University of Leeds, Leeds LS2 9JT, UK<br />

2 Unilever R&D, Colworth Science Park, Sharnbrook MK44 1LQ, UK<br />

3 Unilever R&D, Port Sunlight, Bebingtom CH63 3JW, UK


<strong>Microbubble</strong> <strong>Symposium</strong>: <strong>Fabrication</strong>, <strong>Characterisation</strong> <strong>and</strong> Translational Applications 33<br />

4 th & 5 th July 2011<br />

Poster Number: 8<br />

Title: Opto-acoustic trapping of microbubbles<br />

Caroline Harfield<br />

University College London<br />

Abstract: Microbubbbles are used as ultrasound contrast agents for both diagnostic <strong>and</strong><br />

therapeutic applications <strong>and</strong> are also being investigated for use in gene <strong>and</strong> drug therapy.<br />

For the latter, an improved underst<strong>and</strong>ing of the response of both individual microbubbles<br />

<strong>and</strong> their populations to ultrasound excitation is essential. The challenges associated with<br />

conventional acoustic experiments on microbubbles have produced irreproducible <strong>and</strong> often<br />

contrasting results. In particular, creating a controlled environment in which a single bubble<br />

can be isolated so that it is unaffected by surrounding bubbles is extremely difficult.<br />

Therefore a non-contact method for trapping <strong>and</strong> isolating a single microbubble during<br />

ultrasound excitation would represent an exceptionally valuable tool. The potential of<br />

optical tweezers for trapping <strong>and</strong> manipulating microbubbles have been suggested as a<br />

suitable confinement method. As a feasibility study, a theoretical model of a single bubble<br />

exposed to a combination of optical <strong>and</strong> acoustic fields was developed in order to determine<br />

the magnitude of the forces acting on upon the bubble. Both the radial <strong>and</strong> translational<br />

motion of the bubble were modelled using a Rayleigh Plesset type equation coupled to a<br />

model for the acoustic radiation force. The results from the simulations indicate the Primary<br />

acoustic radiation (Bjerknes) force on a typical contrast agent bubble was 3 or more orders<br />

of magnitude greater that the trapping force which could be provided by the laser, which is a<br />

trend supported by the existing experimental <strong>and</strong> theoretical literature. In conclusion,<br />

classical optical tweezers are not a suitable means of confining acoustically driven bubbles. A<br />

more sophisticated design, combining the spatial precision of optical trapping with the<br />

strength of acoustic localisation will be described.<br />

Additional authors:<br />

Caroline Harfield 1,3,* , A Pawlikowska 2,3 , E. Stride 1 , P. H. Jones 2 , G Memoli 3 ,<br />

1- Department of Mechanical Engineering, UCL<br />

2- Department of Physics <strong>and</strong> Astronomy, UCL<br />

3- National Physical Laboratory


34 <strong>Microbubble</strong> <strong>Symposium</strong>: <strong>Fabrication</strong>, <strong>Characterisation</strong> <strong>and</strong> Translational Applications<br />

4 th & 5 th July 2011<br />

Poster Number: 9<br />

Title: Periodic clustering of microbubbles by secondary radiation force<br />

Sevan Harput<br />

University of Leeds<br />

Abstract: Two bubbles excited above or below their resonance frequencies will result in an<br />

attractive force, while if one is below <strong>and</strong> the other is above resonance, the net force will be<br />

repulsive. However, the attractive <strong>and</strong> repulsive bubble behavior is not always predictable in<br />

a microbubble cloud. The sign reversal of secondary Bjerknes force acting on microbubble<br />

clusters is observed due to the multiple scattering effects. Regardless of their sizes,<br />

microbubbles attract each other <strong>and</strong> form stable bubble clusters.<br />

Objective: The mutual interaction between the pulsating bubbles causes the formation of<br />

the stable bubble clusters due to secondary radiation force. This aggregation pattern of the<br />

bubbles is also referred as “bubble grapes”. This behaviour of microbubbles can affect the<br />

results of ultrasound measurements in-vivo <strong>and</strong> in-vitro.<br />

Methods: The aggregation behaviour of Sonovue microbubbles are observed at 2 MHz<br />

between 50 kPa <strong>and</strong> 500 kPa peak-negative pressures with the flow rates of 1 mL/h to<br />

10mL/h. A custom design tank is used for the experiments with a 150 micrometer thick<br />

mylar window for optical observations <strong>and</strong> a special mount for the transducers. A<br />

transparent cellulose tube with a diameter of 250 micrometers is placed at the focal point of<br />

the transducer where the ultrasound contrast agents are flown. The onset of microbubble<br />

aggregation pattern is captured by Nikon Eclipse Ti-S inverted microscope.<br />

Results: Under certain conditions, the microbubble aggregation pattern is periodic. The<br />

attached figure shows the microbubble aggregation pattern for 3 different flow rates <strong>and</strong><br />

pressure levels.<br />

Additional authors:<br />

Sevan Harput, Benjamin Raiton, Dr. James Mclaughlan, Prof. Stephen Evans a <strong>and</strong> Dr. Steven<br />

Freear<br />

Ultrasound Group, School of Electronic <strong>and</strong> Electrical Engineering, University of Leeds, Leeds, UK.<br />

a School of Physics & Astronomy, University of Leeds, Leeds, UK.


<strong>Microbubble</strong> <strong>Symposium</strong>: <strong>Fabrication</strong>, <strong>Characterisation</strong> <strong>and</strong> Translational Applications 35<br />

4 th & 5 th July 2011<br />

Poster Number: 10<br />

Sevan Harput<br />

University of Leeds<br />

Title: Effect of nonlinear frequency modulated signals on subharmonic emission from<br />

microbubbles<br />

Abstract: Performance of linear frequency modulated (LFM) <strong>and</strong> nonlinear frequency<br />

modulated (NLFM) excitation is compared according to their subharmonic emission for nondestructive<br />

contrast imaging. The subharmonic emission of the microbubbles is measured as<br />

a function of pressure <strong>and</strong> b<strong>and</strong>width for a sinusoidal tone-burst, LFM <strong>and</strong> NLFM signals.<br />

Objective: The aim of this work is to improve the ultrasound image quality by using coded<br />

excitation <strong>and</strong> subharmonic emission from microbubbles. The subharmonic component has<br />

the potential to improve the contrast-to-tissue ratio (CTR) as it is only generated by the<br />

microbubble contrast agents. The coded excitation methods increase the signal to noise<br />

ratio (SNR) <strong>and</strong> penetration depth by using longer pulse durations.<br />

Methods: The scattering properties of SonoVue contrast agent were measured in a<br />

cylindrical chamber containing the 1:1000 diluted SonoVue suspension <strong>and</strong> mixed with a<br />

magnetic stirrer during the experiments. An excitation frequency of 5 MHz is chosen to<br />

excite the microbubbles at twice of their resonance frequency. A 5 MHz transducer mounted<br />

perpendicular to a 1 mm needle hydrophone was placed 10 mm from the chamber.<br />

<strong>Microbubble</strong>s were acoustically excited by a sinusoidal tone-burst, LFM <strong>and</strong> NLFM signals<br />

between peak negative pressures of 25-200 kPa.<br />

Results: It is found that the NLFM signal gives similar or higher subharmonic levels than the<br />

sinusoidal tone-burst <strong>and</strong> LFM signal. In average, NLFM excitation with 20% <strong>and</strong> 40%<br />

b<strong>and</strong>width achieves 3 dB higher subharmonic levels than LFM excitation.<br />

Additional authors:<br />

Sevan Harput, Dr. Muhammad Arif <strong>and</strong> Dr. Steven Freear<br />

Ultrasound Group, University of Leeds, Leeds, UK.


36 <strong>Microbubble</strong> <strong>Symposium</strong>: <strong>Fabrication</strong>, <strong>Characterisation</strong> <strong>and</strong> Translational Applications<br />

4 th & 5 th July 2011<br />

Poster Number: 11<br />

Title: Controlling actin filaments<br />

George Heath<br />

University of Leeds<br />

Abstract: Actin is the most abundant protein found in eukaryotic cells. The 42kDa globular<br />

protein is a major component of the cytoskeleton as it assembles into filaments producing a<br />

supporting scaffold beneath cell membranes. As well as providing mechanical support the<br />

use of actin as a scaffold in biology allows diverse processes such as cell division, cell<br />

locomotion, muscle contraction <strong>and</strong> vesicle transportation. This diverse functionality is<br />

achieved through a huge protein toolbox of actin binding proteins which can be used to: cap,<br />

cut, crosslink, buddle, branch <strong>and</strong> move actin filaments. Offering the potential to create<br />

extremely varied range of structures (either mimicking biology or not) which can be tuned to<br />

the desired application. We use Atomic Force Microscopy (AFM) in fluid to obtain high<br />

resolution images of phalloidin stabilized actin filaments electrostaticly bound to Nickel<br />

treated mica <strong>and</strong> confirm that controlling relative concentrations of Actin <strong>and</strong> a<br />

capping/severing protein Gelsolin tailors filament length.


<strong>Microbubble</strong> <strong>Symposium</strong>: <strong>Fabrication</strong>, <strong>Characterisation</strong> <strong>and</strong> Translational Applications 37<br />

4 th & 5 th July 2011<br />

Poster Number: 12<br />

Nicola Ingram<br />

University of Leeds<br />

Title: Pre-clinical evaluation of VEGFR2 as an imaging biomarker <strong>and</strong> targeting molecule for<br />

therapeutic delivery.<br />

Abstract: We are developing novel ways to image functional vasculature in vivo for assessing<br />

tumour development <strong>and</strong> response to therapeutics as well as delivering tumour targeted<br />

therapeutics for treatment of colorectal cancer (CRC). VEGFR2 has been described as a<br />

marker of angiogenesis but its expression <strong>and</strong> distribution in colorectal xenograft tumour<br />

development has not been determined. We have therefore used contrast enhanced high<br />

frequency ultrasound (CE HFUS) for dynamic imaging of VEGFR2 in a longitudinal study of<br />

SW480 CRC xenografts.<br />

Immunofluorescence using multiple antibodies to VEGFR2 was carried out on endothelial<br />

cell lines <strong>and</strong> primary cells. Immunohistochemistry for VEGFR2 expression in vessels was<br />

carried out on SW480 xenograft tissue. For CE HFUS, microbubbles (MB) were coated with<br />

VEGFR2 antibody <strong>and</strong> injected intravenously into SW480-tumour bearing mice. We used the<br />

VisualSonics Vevo 770 micro-US system to evaluate bolus injection characteristics to show<br />

functional vessels within the tumour <strong>and</strong> binding of MBs to tumour vasculature both in 2D<br />

<strong>and</strong> in 3D.<br />

Immunofluorescence studies demonstrated the lack of a suitable murine endothelial cell line<br />

to use for tumour vascular targeting in pre-clinical studies. Immunohistochemistry on SW480<br />

xenografts showed that VEGFR2 was indeed present in tumour vessels, in line with human<br />

studies. There was a peak in CD31+ vessels that also expressed VEGFR2 at approximately<br />

50mm 3 in size. Expression then decreased as the tumours increased in volume. Therefore,<br />

smaller size xenografts were used for in vivo targeting experiments. VEGFR2-targeted MB<br />

injection <strong>and</strong> 3D destruction of bound MBs showed approximately 20% of the tumour<br />

vasculature in SW480 xenografts had bound microbubbles.<br />

VEGFR2 expression was upregulated in small, developing CRC xenografts <strong>and</strong> can be<br />

therefore used to image vasculature <strong>and</strong> target MBs in pre-clinical models of CRC.<br />

Additional authors:<br />

Nicola Ingram* 1 , Elizabeth Vallely 1 , Pam Jones 1 , Alex Markham 2 , P Louise Coletta 1 .<br />

1 Molecular Gastroenterology, 2 Translational Medicine, Leeds Institute of Molecular Medicine, St James’s<br />

University Hospital, Leeds, U.K.


38 <strong>Microbubble</strong> <strong>Symposium</strong>: <strong>Fabrication</strong>, <strong>Characterisation</strong> <strong>and</strong> Translational Applications<br />

4 th & 5 th July 2011<br />

Poster Number: 13<br />

Gaël Léauté<br />

University of Leeds<br />

Title: Effect of excitation pulses on the sonoporation efficiency of mono- <strong>and</strong> polydisperse<br />

ultrasound contrast agents<br />

Abstract: Nyborg 1 theory allows estimations of the shear stress caused by an oscillating<br />

hemispherical gas bubble on a boundary. Doinikov <strong>and</strong> Bouakaz 2 further developed this<br />

model for ultrasound contrast agents (UCA) in the vicinity of a boundary <strong>and</strong> developed a<br />

method to estimate the sonoporation efficiency of UCA-cell solution to an ultrasound field.<br />

This was done with a modified Rayleigh-Plesset equation that incorporated a rigid boundary<br />

near the oscillating microbubbles. The amount of shear stress exerted by the microbubbles,<br />

on this boundary, is calculated <strong>and</strong> by assuming a shear stress threshold of 12 Pa for<br />

sonoporation to occur, the number of cells affected can be estimated. The sonoporation<br />

efficiency is then defined as the ratio of the number of sonoporated cells to the total<br />

number of cells, by assuming cells <strong>and</strong> microbubbles have the same uniform distribution <strong>and</strong><br />

concentration. It was shown that sonoporation efficiency becomes minimal when the driving<br />

frequency is equal to the microbubble populations resonance frequency at which<br />

attenuation of the acoustic field becomes maximal.<br />

Although the theoretical background of the sonoporation mechanism is still in its infancy,<br />

our study will use this model as the basis to investigate the effect of ultrasound pulse shape<br />

on sonoporation efficiency, specifically whether tone bursts or linear swept frequency<br />

excitation pulses would provide the greatest sonoporation efficiency for mono- <strong>and</strong>/or<br />

polydisperse microbubble populations. An experiment could shed light on whether the<br />

acoustic field attenuation has the strong influence on sonoporation efficiency as described<br />

by the theoretical results found by Doinikov et al. The ratio of cells which have undergone<br />

sonoporation could be measured by causing the uptake of a fluorophore into the cell<br />

through sonoporation induced by different microbubble populations.<br />

References:<br />

1 W.L. Nyborg, Acoustic streaming near a boundary, J. Acoust. Soc. Am. 30 (1958) 329-339.<br />

2 A.A. Doinikov <strong>and</strong> A. Bouakaz, Theoretical investigation of shear stress generated by a contrast microbubble on<br />

the cell membrane as a mechanism for sonoporation, J. Acoust. Soc. Am. 128 (2010) 11-19.<br />

Additional authors:<br />

Gaël Y.V. Léauté * , Dr. James Mclaughlan <strong>and</strong> Dr. Steven Freear<br />

Ultrasound Group, School of Electronic <strong>and</strong> Electrical Engineering, University of Leeds, Leeds, UK.


<strong>Microbubble</strong> <strong>Symposium</strong>: <strong>Fabrication</strong>, <strong>Characterisation</strong> <strong>and</strong> Translational Applications 39<br />

4 th & 5 th July 2011<br />

Poster Number: 14<br />

Jonathan Loughran<br />

Imperial College London<br />

Title: Selective imaging of adherent microbubbles with correction of tissue motion<br />

Abstract: In order to improve the efficiency of molecular imaging using targeted<br />

microbubbles, a method is needed to distinguish the free flowing microbubbles from<br />

adherent microbubbles which have found its predefined targeting site. While simple<br />

temporal low pass filtering has been used to this end assuming adherent bubbles are<br />

stationary, this technique would not work in clinical situations when tissue motion is present<br />

causing movement in the adherent bubbles too. This study aims to address this problem by<br />

incorporating motion correction into the imaging process.<br />

In house biotin coated lipid shell microbubbles were inserted into a streptavidin coated<br />

cellulose tube of 200μm in diameter under steady flow conditions created using a syringe<br />

pump. The tube located in a water bath was observed optically using a microscopy to verify<br />

that bubbles were indeed adhering to the surface of the tube, while pulse inversion image<br />

sequences were simultaneously taken using an ultrasound scanner. Motion was created to<br />

the tube via a 3D stage holding the tube in place. The image sequence recorded were<br />

corrected for motion using cross-correlation <strong>and</strong> the results show that it was possible to<br />

selectively detect the adherent microbubbles using the low pass filtering techniques after<br />

the motion correction.<br />

Additional authors:<br />

Jonathan Loughran 1 , Charles Sennoga 12 , Robert Eckersley 2 <strong>and</strong> Meng-Xing Tang 1<br />

1 Department of Bioengineering, Imperial College London, South Kensington Campus, London SW7 2AZ, United<br />

Kingdom;<br />

2 Imaging Sciences Department, Imperial College London, Hammersmith Campus, London W12 0HS United<br />

Kingdom;


40 <strong>Microbubble</strong> <strong>Symposium</strong>: <strong>Fabrication</strong>, <strong>Characterisation</strong> <strong>and</strong> Translational Applications<br />

4 th & 5 th July 2011<br />

Poster Number: 15<br />

Paul Mackin<br />

University of Cambridge<br />

Title: Non-Invasive ultrasound imaging in a mouse model of pancreatic ductal<br />

adenocarcinoma<br />

Abstract: Pancreatic ductal adenocarcinoma (PDA) is the fifth most common cause of cancer<br />

death in the UK causing 7,781 deaths in 2008. Prognosis is extremely poor; survival of<br />

patients with advanced disease is about 6 months. Despite major efforts in basic <strong>and</strong> clinical<br />

research, most therapies still fail to improve survival <strong>and</strong> novel therapies are urgently<br />

required for this fatal disease.<br />

Our lab has produced a genetically engineered mouse model (KPC) carrying Kras <strong>and</strong> p53<br />

mutations that recapitulate the clinical <strong>and</strong> pathological features of human disease including<br />

site of occurrence, tumours, metastasis, tumour microenvironment, haemorrhagic ascites<br />

etc. This model is therefore likely to be more predictive of response to novel therapies that<br />

can then be translated to patients.<br />

Our ‘Mouse Hospital’ team streamline the production of experimental mice suitable for<br />

enrolment onto therapeutic studies. Weekly palpation of the mutant mice confirms tumour<br />

presence. Tumours are measured by non-invasive ultrasound imaging. Mice with a defined<br />

tumour burden (6-9-mm-diameter) are enrolled onto study.<br />

We are refining the assessment of pre enrolable tumours by precise palpations <strong>and</strong> reducing<br />

the stress of mice being repeatedly ultrasound scanned to confirm <strong>and</strong> follow the growth<br />

pattern of the tumours as well as the associated implications of repeated exposure to<br />

anaesthetic.<br />

In the past allograft <strong>and</strong> xenograft models have used countless animals to show therapeutic<br />

efficacy in the model only, rarely translating efficacy to the clinic. In contrast, we believe that<br />

the KPC model is a better predictor of response to therapy. Indeed, we have shown that this<br />

model responds to Gemcitabine, the st<strong>and</strong>ard of care for pancreatic cancer, in a similar<br />

manner as seen in the clinical setting.<br />

To date, over ten drugs have been tested in the Mouse Hospital. Studies highlighting the<br />

barrier to drug delivery by the excessive stroma are of particular interest. We have shown<br />

that the efficacy of chemotherapy can be improved by co-administration of a drug which<br />

depletes tumour-associated stroma, resulting in an extended lifespan.<br />

These promising results are currently translated into clinical practice; two clinical trials have<br />

recently opened to investigate experimental drugs from the Mouse Hospital either in<br />

advanced or pre-operative PDA patients.


<strong>Microbubble</strong> <strong>Symposium</strong>: <strong>Fabrication</strong>, <strong>Characterisation</strong> <strong>and</strong> Translational Applications 41<br />

4 th & 5 th July 2011<br />

Poster Number: 16<br />

Richard O’Rorke<br />

University of Leeds<br />

Title: Trapping microbubbles in a moveable acoustic array using surface acoustic waves<br />

Abstract: St<strong>and</strong>ing pressure waves at MHz frequencies are used to position <strong>and</strong> transport<br />

microbubbles within a microfluidic device. We show that surface acoustic waves (SAWs) –<br />

nanometer scale vibrations on a piezoelectric crystal – can be excited to form a st<strong>and</strong>ing<br />

wave on the surface, which couples into the liquid channel of an adjacent microfluidic<br />

device. This results in a st<strong>and</strong>ing pressure wave in the liquid, which we use to manipulate<br />

suspended particles. We discuss the formation of one-dimensional arrays of micron-sized<br />

latex beads in proving work, in the context of the primary acoustic radiation force. Our<br />

acoustic alignment technique is applied to a sample of microbubbles <strong>and</strong> we demonstrate<br />

microbubble array transport, through small increments in st<strong>and</strong>ing SAW frequency. Finally,<br />

we present two-dimensional particle arrays which could be applied to microbubbles to allow<br />

individual microbubbles to be spatially controlled for subsequent characterization.<br />

Additional authors:<br />

Richard O’Rorke, C. Wood, C. Walti, A. G. Davies, S. D. Evans, J. E. Cunningham.


42 <strong>Microbubble</strong> <strong>Symposium</strong>: <strong>Fabrication</strong>, <strong>Characterisation</strong> <strong>and</strong> Translational Applications<br />

4 th & 5 th July 2011<br />

Poster Number: 17<br />

Title: 3D exp<strong>and</strong>ing geometry for improved on-chip microbubble production rates<br />

Sally Peyman<br />

University of Leeds<br />

Abstract: Micron sized, lipid stabilized bubbles of gas have been utilized in the medical world<br />

as contrast agents for ultra-sound (US) imaging. The water-gas interface created by the<br />

bubbles enhances existing images by reflecting sound waves more efficiently than tissue<br />

interfaces alone. Much interest has been gained recently to combine these advantages with<br />

other medical procedures, such as drug delivery or gene therapy. Conventionally,<br />

microbubble solutions are made by sonication or shaking of lipid solutions in a single step<br />

batch method. Whilst this provides adequate means to prepare simple microbubble<br />

solutions, if true multi-functional bubbles for imaging <strong>and</strong> therapy are to be realized, a more<br />

sophisticated generation method would be required.<br />

The area of microfluidics provides an exciting platform in which complex microbubbles can<br />

be generated in a controlled <strong>and</strong> reproducible manner by offering advancements in<br />

automation, fluid control, versatility <strong>and</strong> microbubble manipulation. <strong>Microbubble</strong>s are<br />

generated in flow-focussing microfluidic devices by nipping streams of gas <strong>and</strong> liquid<br />

through a tiny nozzle <strong>and</strong> then subjecting the two phases to a downstream pressure drop. To<br />

date microbubble generation has been performed solely on planar devices, in which<br />

pressure drops are created on a single axis. We propose a 3D expansion to further improve<br />

microbubble production rates by creating a large pressure drop in the outlet channel (figure<br />

1). In addition, we present a new bubble production regime. The microchip outlet exp<strong>and</strong>ed<br />

in a vertical direction from 25 µm at the nozzle to 50 µm in the outlet. Bubble concentrations<br />

produced using this device were up to 1 x 10 9 bubbles / mL, improving bubble<br />

concentrations from designs without the 3D expansion by a factor of 10.<br />

Figure 1. Photograph of the flow-focussing microfluidic device showing lipid<br />

<strong>and</strong> gas streams, the 3D expansion area <strong>and</strong> the bubble formation area.


<strong>Microbubble</strong> <strong>Symposium</strong>: <strong>Fabrication</strong>, <strong>Characterisation</strong> <strong>and</strong> Translational Applications 43<br />

4 th & 5 th July 2011<br />

Poster Number: 18<br />

Title: Counter flow microbubble channelling using acoustic radiation force funnel<br />

Benjamin Raiton<br />

University of Leeds<br />

Abstract:<br />

Background, Motivation <strong>and</strong> Objective<br />

The key advantage when employing acoustic travelling waves to manipulate microspheres is<br />

the ability to penetrate biological tissues. Other techniques such as optical tweezers or<br />

st<strong>and</strong>ing-wave traps are limited by their working distance. Previous publications reported on<br />

the use of Gaussian focused ultrasound to translate 125 μm lipid droplets <strong>and</strong> an opposite<br />

phase induced pressure black spot to align flowing particles. When microbubbles (MB) are<br />

used as drug carriers, a single element transducer is commonly used to emit a plane<br />

pressure wave. The effect, referred to as primary radiation force, is a translation of the MB<br />

away from the vessel lumen <strong>and</strong> towards the vessel wall to facilitate bonding to the<br />

endothelium. The axial primary radiation force (APRF) is calculated here according to Dayton<br />

et al. [1] along with the lateral primary radiation force (LPRF) following the simplified<br />

equation by Gor’kov [2].<br />

Statement of Contribution/Methods<br />

Lipid MB used in the experiment have a size distribution of 1-10 μm. The MB are flowed<br />

through a 1 mm wide channel with a height of 100 μm, at a rate similar to that of venules. A<br />

low peak-negative-pressure (PNP) pulse of 200 cycles is emitted every 6.25 ms. The 96<br />

elements of a st<strong>and</strong>ard medical imaging probe are focused at the channel centre at 24 mm.<br />

Each half of the array transmits the same tone burst, but out of phase by 180°.<br />

Results<br />

In the figure, captured from beneath the vessel, the fluid is flowing from left to right. A clear<br />

formation of 3 parallel channels, flowing in the opposite direction, is observed as predicted<br />

by the simulation. The thin grey trails are made up of unaggregated MB whilst the larger <strong>and</strong><br />

darker dots indicate MB clustering. The clusters break up as soon as the ultrasound source is<br />

switched off.<br />

Discussion <strong>and</strong> Conclusions<br />

Thanks to the use of a 1D linear array the spacing between the channels can be dynamically<br />

varied by altering the focal depth or number of elements used. The center of the three<br />

channels can also be shifted up <strong>and</strong> down by steering the focal point of the array.<br />

References:<br />

[1] P.A. Dayton et al., UFFC IEEE Trans, vol.44, no.6, pp,1264-1277, 1997<br />

*2+ L.P. Gor’kov, Sov. Phys. 6 (9), 773-775, 1962<br />

Additional authors:<br />

Benjamin Raiton, Sevan Harput, Dr James Mclaughlan, Dr Steven Freear<br />

Ultrasound Group, School of Electronic <strong>and</strong> Electrical Engineering, University of Leeds, Leeds, West Yorkshire,<br />

United Kingdom


44 <strong>Microbubble</strong> <strong>Symposium</strong>: <strong>Fabrication</strong>, <strong>Characterisation</strong> <strong>and</strong> Translational Applications<br />

4 th & 5 th July 2011<br />

Poster Number: 19<br />

Title: Comparision of methods for sizing <strong>and</strong> counting of microbubbles<br />

Charles Sennoga<br />

Imperial College London<br />

Abstract: The size <strong>and</strong> number count of microbubble populations are essential information<br />

for interpreting their acoustic behaviour <strong>and</strong> have a direct impact on clinical contrast<br />

enhanced ultrasound imaging <strong>and</strong> microbubble mediated therapy. There are three common<br />

ways of sizing <strong>and</strong> counting microbubbles, namely the electro-impedance volumetric zone<br />

sensing (ES) also called a Coulter Counter/Multisizer, optical microscopy (OM) <strong>and</strong> laser<br />

diffraction (LD). However the efficacy of these methods have not been thoroughly evaluated.<br />

In this study these methods was assessed. Microspheres with certified mean size diameter<br />

(MS) <strong>and</strong> number concentration (C) were used to assess sizing <strong>and</strong> counting reproducibility<br />

<strong>and</strong> reliability of ES, OM <strong>and</strong> LD methods. SonoVue was used to validate ES, OM <strong>and</strong> LD<br />

sizing <strong>and</strong> counting of microbubbles. Statistical analyses of intra-method variability for the<br />

SonoVue MS showed that the microbubble sizing was best obtained using OM compared<br />

to ES <strong>and</strong> LD. The microbubble counting reproducibility was best obtained using ES as<br />

compared to OM <strong>and</strong> LD.<br />

Additional authors:<br />

Charles A. Sennoga 12 , James S. M. Yeh 2 , Julia Alter 2 , Eleanor Stride 3 , Petros<br />

Nihoyannopoulos 4, John M. Seddon 5 , Dorian O. Haskard 4 , Joseph V. Hajnal 2 , Robert J.<br />

Eckersley 2 , Meng-Xing Tang 1<br />

1 Department of Bioengineering, Imperial College London, South Kensington Campus, London SW7<br />

2AZ, United Kingdom;<br />

2 Imaging Sciences Department, Imperial College London, Hammersmith Campus, London W12 0HS United<br />

Kingdom;<br />

3 Department of Mechanical Engineering, University College London, Torrington Place, London WC1E 7JE<br />

United Kingdom;<br />

4 National Heart <strong>and</strong> Lung Institute, Imperial College London, Hammersmith Campus, London W12 0HS United<br />

Kingdom;<br />

5 Department of Chemistry, Imperial College London, South Kensington Campus, London SW7 2AY, United<br />

Kingdom;


<strong>Microbubble</strong> <strong>Symposium</strong>: <strong>Fabrication</strong>, <strong>Characterisation</strong> <strong>and</strong> Translational Applications 45<br />

4 th & 5 th July 2011<br />

Poster Number: 20<br />

Title: Pre-distorted chirp excitations for polydisperse microbubble populations<br />

Peter Smith<br />

University of Leeds<br />

Abstract: Ultrasound contrast agents are ideally monodisperse with single resonance frequency.<br />

Commercial microbubble populations however are often polydisperse e.g. SonoVue (approximately<br />

a 1-10 µm diameter range <strong>and</strong> mean value of 2.5 µm). As a consequence, a microbubble population<br />

will not be resonant at a single frequency but across a frequency range. The use of a frequency<br />

modulated or chirp excitation can increase the number of microbubbles which exhibit resonant<br />

behaviour within a population. However, a conventional chirp excitation will suffer from tapering<br />

effects according to the transducer’s b<strong>and</strong>width <strong>and</strong> frequency characteristics (as shown in the top<br />

plots of the figure).<br />

Whilst chirp excitation allows a large population of polydisperse microbubbles to be excited at their<br />

resonant frequencies; due to the nature of the transducer-tapered chirp, only a particular sub-set of<br />

the microbubble population are exposed to maximum pressure under resonant conditions. This work<br />

seeks to equalise the transmitted pressure across frequency so that all microbubbles within the<br />

population are subject to comparable pressure at their point of resonance.<br />

Chirp excitation sequences are designed which have an additional amplitude modulation function.<br />

This amplitude modulation pre-distorts the chirp signal in order to compensate for the filtering effect<br />

of the transducer. By pre-distorting the excitation sequence <strong>and</strong> shaping the pulse to reflect the<br />

attenuating response of the transducer, it is possible to achieve approximately constant pressure<br />

over a desired frequency b<strong>and</strong> (as shown in the bottom plots of the figure).<br />

Excitation sequences are proposed that incorporate both the characteristics of the microbubble<br />

population <strong>and</strong> the frequency response of the transducer. Their aim is to achieve constant amplitude<br />

across a frequency range by effectively increasing the 3dB excitation b<strong>and</strong>width of the transducer.<br />

Additional authors: Peter R. Smith*, Dr David M. J. Cowell, Dr James McLaughlan <strong>and</strong> Dr Steven<br />

Freear Ultrasound Group, School of Electronic <strong>and</strong> Electrical Engineering, University of Leeds, United Kingdom


46 <strong>Microbubble</strong> <strong>Symposium</strong>: <strong>Fabrication</strong>, <strong>Characterisation</strong> <strong>and</strong> Translational Applications<br />

4 th & 5 th July 2011<br />

Delegate List<br />

Name Affiliation Email<br />

Mostafa Abdelrahman University of Leeds mtp5ma@leeds.ac.uk<br />

Radwa Abou-Saleh University of Leeds R.H.Saleh@leeds.ac.uk<br />

Muhammed Arif University of Leeds elma@leeds.ac.uk<br />

Jonathan Blockley University of Leeds pyjb@leeds.ac.uk<br />

Matthew Booth University of Leeds js06mab@leeds.ac.uk<br />

Christian Buchcic Wageningen University christian.buchcic@wur.nl<br />

Joanne Burke University of Leeds fs08jhb@leeds.ac.uk<br />

Richard Bushby University of Leeds R.J.Bushby@leeds.ac.uk<br />

Paul Campbell University of Dundee p.a.campbell@dundee.ac.uk<br />

Louise Coletta University of Leeds P.L.Coletta@leeds.ac.uk<br />

David Cowell University of Leeds D.M.J.Cowell@leeds.ac.uk<br />

Kevin Critchley University of Leeds K.Critchley@leeds.ac.uk<br />

Peter Culmer University of Leeds P.R.Culmer@leeds.ac.uk<br />

Verya Daeichin Erasmus MC v.daeichin@erasmusmc.nl<br />

Kari Dempsey Leeds General Infirmary kjd@medphysics.leeds.ac.uk<br />

Robert Eckersley Imperial College London. r.eckersley@csc.mrc.ac.uk<br />

Lydia Eidemueller VisualSonics leidemueller@visualsonics.com<br />

Stephen Evans University of Leeds S.D.Evans@leeds.ac.uk<br />

Tony Evans University of Leeds J.A.Evans@leeds.ac.uk<br />

Sarah Fawcett University of Cambridge Sarah.Fawcett@cancer.org.uk<br />

Damien Fouan Bf Systemes company damien.fouan@bf-systemes.fr<br />

Steve Freear University of Leeds S.Freear@leeds.ac.uk<br />

Caroline Harfield University College London c.harfield@ucl.ac.uk<br />

Sevan Harput University of Leeds eensha@leeds.ac.uk<br />

George Heath University of Leeds py06gh@leeds.ac.uk<br />

Carola Heneweer University Hospital Schleswig-<br />

Holstein, Kiel Germany<br />

c.heneweer@rad.uni-kiel.de<br />

Robert Hewson University of Leeds R.W.Hewson@leeds.ac.uk<br />

Nicola Ingram University of Leeds n.ingram@leeds.ac.uk<br />

Benjamin Johnson University of Leeds B.R.G.Johnson@leeds.ac.uk<br />

Pam Jones University of Leeds P.Jones@leeds.ac.uk<br />

Fabian Kiessling University of Aachen fkiessling@ukaachen.de<br />

Klazina Kooiman Erasmus MC k.kooiman@erasmusmc.nl


<strong>Microbubble</strong> <strong>Symposium</strong>: <strong>Fabrication</strong>, <strong>Characterisation</strong> <strong>and</strong> Translational Applications 47<br />

4 th & 5 th July 2011<br />

Dmitriy Kuvshinov University of Sheffield d.kuvshinov@sheffield.ac.uk<br />

Sean Lawler University of Leeds S.Lawler@leeds.ac.uk<br />

Gael Leaute University of Leeds elgyvl@leeds.ac.uk<br />

Ine Lentacker Ghent University Ine.Lentacker@UGent.be<br />

Marjorie Longo University of California mllongo@ucdavis.edu<br />

Mihaela Lorger University of Leeds medmlo@leeds.ac.uk<br />

Jonathan Loughran Imperial College London<br />

Paul Mackin Cambridge Research Institute Paul.Mackin@cancer.org.uk<br />

Sir Alex Markham University of Leeds A.F.Markham@leeds.ac.uk<br />

Gemma Marston University of Leeds G.Marston@leeds.ac.uk<br />

Jonathan McKendry University of Leeds jonomckendry@gmail.com<br />

James McLaughlan University of Leeds J.R.McLaughlan@leeds.ac.uk<br />

Gianluca Memoli National Physical Laboratory gianluca.memoli@npl.co.uk<br />

Serge Mensah CNRS mensah@lma.cnrs-mrs.fr<br />

Anne Neville University of Leeds A.Neville@leeds.ac.uk<br />

Richard O'Rorke University of Leeds R.D.O'Rorke@leeds.ac.uk<br />

Sally Peyman University of Leeds S.Peyman@leeds.ac.uk<br />

Malcolm Povey University of Leeds m.j.w.povey@food.leeds.ac.uk<br />

Ben Raiton University of Leeds elbdr@leeds.ac.uk<br />

Tijs Rovers Wageningen University tijs.rovers@wur.nl<br />

Tim Ryan Epigem Limited Tim.Ryan@epigem.co.uk<br />

Vassilis Sboros University of Edinburgh Vassilis.Sboros@ed.ac.uk<br />

Charles Sennoga Imperial College London c.sennoga@imperial.ac.uk<br />

Peter Smith University of Leeds efy3prs@leeds.ac.uk<br />

Jane Smith (was Bates) St James's University Hospital, Leeds Janes.Bates@leedsth.nhs.uk<br />

Eleanor Stride University College London eleanor.stride@ucl.ac.uk<br />

Chao Sun University of Edinburgh C.Sun-2@sms.ed.ac.uk<br />

Mengxing Tang Imperial College London mengxing.tang@imperial.ac.uk<br />

Neil Thomson University of Leeds N.H.Thomson@leeds.ac.uk<br />

Philippe Trochet VisualSonics ptrochet@visualsonics.com<br />

Richard Wakefield University of Leeds medrjw@leeds.ac.uk<br />

Nicholas Watson University of Leeds n.j.watson@leeds.ac.uk<br />

Stephen Wolstenhulme University of Leeds hcsswo@leeds.ac.uk

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

Saved successfully!

Ooh no, something went wrong!