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 />

16 th & 17 th July 2012<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 />

16 th & 17 th July 2012


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

16 th & 17 th July 2012<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> 6<br />

Oral presentation – Session 2<br />

<strong>Microbubble</strong> Ultrasound <strong>Characterisation</strong> 12<br />

Oral presentation – Session 3<br />

<strong>Microbubble</strong> Translational Applications 16<br />

Poster presentations 22<br />

Delegate list 49


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

16 th & 17 th July 2012<br />

Monday 16 th July 2012<br />

Program<br />

13:00 – 14.00 Registration<br />

14:00 – 14:10 Stephen Evans Welcome<br />

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

14:10 – 14:50 Nico de Jong <strong>Microbubble</strong>s for Medical Use<br />

14:50 – 15:30 Mohan Edirisinghe Preparation of Bubbles <strong>and</strong> Capsules:<br />

Electrohydrodynamics vs Microfluidics<br />

15:30 – 15:55 Radwa Abou-Saleh &<br />

Sally Peyman<br />

15:55 – 16:30 Tea / Coffee<br />

High On-Chip Production Rates <strong>and</strong> Mechanical<br />

<strong>Characterisation</strong> of Therapeutic <strong>Microbubble</strong>s<br />

for Targeted Drug Delivery.<br />

16:30 – 17:10 Vasileios Koutsos Nanomechanical Properties of <strong>Microbubble</strong>s<br />

Studied by Atomic Force Microscopy<br />

17:10 – 17:35 Jonathan Loughran Investigation into the Effects of Ultrasound on<br />

Attached <strong>Microbubble</strong>s<br />

18:00 – 19:15 Drinks <strong>and</strong> Posters<br />

19:15 – 21:15 <strong>Symposium</strong> Dinner<br />

21:15 - onwards Informal discussions in the Stables pub


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

16 th & 17 th July 2012<br />

Tuesday 17 th July 2012<br />

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

09:00 – 09:40 Paul Dayton <strong>Microbubble</strong> Imaging in Cancer Theranostics<br />

09:40 – 10:20 John Hossack Catheter-Based Monodisperse <strong>Microbubble</strong><br />

Production for Image-guided Vascular Therapy<br />

10:20 – 10:50 Tea / Coffee<br />

10:50 – 11:15 James McLaughlan The Acoustic Evaluation of Liposome-Loaded<br />

<strong>Microbubble</strong>s for Targeted Cancer Therapy<br />

11:15 – 11:55 Ayache Bouakaz Ultrasound Contrast Agents: from Imaging to<br />

Therapy<br />

11:55 – 13:30 Lunch<br />

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

13:30 – 14:10 Carmel Moran <strong>Characterisation</strong> of Ultrasonic Contrast<br />

<strong>Microbubble</strong>s for Preclinical Applications<br />

14:10 – 14:50 Jithin Jose Multi-modal Contrast Agents: Photoacoustic<br />

Imaging <strong>and</strong> Therapy<br />

14:50 – 15:15 Nicola Ingram Microfluidically-Generated <strong>Microbubble</strong>s for<br />

Molecular Imaging <strong>and</strong> Drug Delivery in<br />

Colorectal Cancer – Pre-Clinical Evaluation<br />

15:15 – 15:45 Tea / Coffee<br />

15:45 – 16:25 Robin Clevel<strong>and</strong> Therapeutic Uses of <strong>Microbubble</strong>s: Heating,<br />

Erosion <strong>and</strong> Delivery<br />

16:25 – 17:00 Jane Smith Contrast Enhanced Ultrasound in Clinical<br />

Practice<br />

17:00 – 17:10 Sir Alex Markham Closing remarks


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

16 th & 17 th July 2012<br />

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

Nico de Jong<br />

ErasmusMC, Rotterdam <strong>and</strong> University Twente<br />

Oral Presentation<br />

Title: <strong>Microbubble</strong>s for Medical Use<br />

Abstract: Ultrasound is the most widely used medical imaging modality. It is capable of providing real-time<br />

information about tissue structures <strong>and</strong> blood flow in the heart <strong>and</strong> larger vessels. For determining perfusion<br />

Ultrasound Contrast Agents (UCA) are m<strong>and</strong>atory. Besides using UCA for measuring perfusion there is a<br />

growing interest in the use of coated microbubbles for therapeutic applications. In this case the microbubbles<br />

can act as transport carriers for drug delivery or when targeted, to image the targeted site, referred as<br />

molecular imaging. A third application for therapy is known as sonoporation. Here, the oscillation of<br />

ultrasound-driven microbubbles, in close contact with a cell, lead to an increased permeability of the cell to<br />

macromolecules, hence to an increased uptake of drugs or genes.<br />

In ultrasound contrast imaging one utilizes the non linear vibrations of the bubbles in the ultrasound field.<br />

When the generated ultrasound wave hits a free or encapsulated microbubble, a volume pulsation is initiated.<br />

Depending on the magnitude of the ultrasound wave the pulsation will be: linear to the applied pressure or<br />

non-linear to the applied pressure. Non linear vibration can be split into: stationary (harmonic) <strong>and</strong> transient<br />

(power) scattering. Depending on the agent three acoustic pressure regions can be distinguished: linear<br />

scattering: 0-50, harmonic scattering: 50-200 <strong>and</strong> transient power scattering: 200-2000 KPascal.<br />

Bubble vibration can be seen under the microscope <strong>and</strong> a ultrafast framing camera. A high frame rate is<br />

m<strong>and</strong>atory to record the vibration of a bubble when it is insonified with 2-3 MHz ultrasound, which is the<br />

normal diagnostic frequency. The figure below presents an example of a recording with such a camera<br />

(Br<strong>and</strong>aris 128) showing an isolated bubble with a size of 4.2 µm in diameter that is insonified with an acoustic<br />

pressure of 250 kPa at a frequency of 1.7 MHz. In 64 frames we observe the bubble before, during <strong>and</strong> after<br />

the ultrasound pulse. In the first 13 frames, the microbubble is at rest. Starting at frame 14, the microbubble is<br />

first compressed, <strong>and</strong> then reaches within six cycles a maximum diameter of 5.4 μm <strong>and</strong> a minimum diameter<br />

of 2.6 µm. We can establish in each image frame the bubble diameter. The resulting diameter-time (D-T) curve<br />

is shown in the panel in the middle <strong>and</strong> the corresponding power spectrum in the right panel.<br />

With the camera we have observed numerous phenomenon,<br />

like subharmonics, ultraharmonics, thresholding,<br />

compression only, expansion only, thresholding <strong>and</strong> mode<br />

vibration as shown in the figure. Moreover, the behaviour of<br />

an individual bubble nearby a wall or cell has been<br />

extensively examined to resolve the mechanism behind<br />

sonoporation <strong>and</strong> binding.<br />

The Frequency power<br />

spectrum shows directly that<br />

the bubble not only scatter<br />

the fundamental frequency at<br />

1.7 MHz, but also a second<br />

harmonic component at 3.4<br />

MHz. The level of this second<br />

harmonic is about -17 dB<br />

below the fundamental. This<br />

harmonic vibration amplitude<br />

generates a pressure wave<br />

which is about 50 % of the<br />

fundamental pressure wave.


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

16 th & 17 th July 2012<br />

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

Oral Presentation<br />

Mohan Edirisinghe<br />

University College London<br />

Title: Preparation of Bubbles <strong>and</strong> Capsules: Electrohydrodynamics vs Microfluidics<br />

Abstract: Two major methods gathering tremendous interest for the mass production of<br />

microbubbles <strong>and</strong> micro/nano capsules are co-axial electrohydrodynamic microbubbling<br />

(CEHD-M) <strong>and</strong> T-junction microfluidics (TJM). This lecture will summarise the key recent<br />

developments in these techniques, mainly focussing on efforts to control size <strong>and</strong> size<br />

distribution of the products. In the case of CEHD-M the use of >2 needles has been a major<br />

feature 1 . Regarding TJM, the junction geometry, its dimensions <strong>and</strong> the central zone where<br />

fluids meet <strong>and</strong> interact has been fundamentally changed. These changes have resulted in<br />

several new products 2 <strong>and</strong>, also led to the controlled preparation of nanoparticles from<br />

bubbles directly at the microfluidic junction site, a very useful feature for drug delivery 3 .<br />

References:<br />

1) M-w.Chang, E.Stride <strong>and</strong> M.Edirisinghe, Layered Bodies, Compositions Containing Them <strong>and</strong> Processes<br />

for Producing Them, PCT/GB2012/050276, 8 th Feb. 2012.<br />

2) O.Gunduz, Z.Ahmad, E.Stride <strong>and</strong> M.Edirisinghe, Mater. Sci. Eng. C, 32(2012)1005-1010.<br />

3) O.Gunduz, Z.Ahmad, E.Stride, C.Tamerler <strong>and</strong> M.Edirisinghe, J. Royal Soc. Interface, 9(2012)389-395.


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

16 th & 17 th July 2012<br />

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

Oral Presentation<br />

Radwa Abou-Saleh & Sally Peyman<br />

University of Leeds<br />

Title: High On-Chip Production Rates <strong>and</strong> Mechanical <strong>Characterisation</strong> of Therapeutic<br />

<strong>Microbubble</strong>s for Targeted Drug Delivery.<br />

Abstract: Micron sized, lipid stabilized bubbles of gas are of interest as contrast agents for<br />

ultra-sound (US) imaging <strong>and</strong> increasingly as vehicles for targeted drug delivery.<br />

Microfluidics provides a reproducible means for microbubble production <strong>and</strong> surface<br />

functionalisation using flow-focussing microfluidic devices that combine streams of gas <strong>and</strong><br />

liquid through a nozzle a few microns wide <strong>and</strong> then subjecting the two phases to a<br />

downstream pressure drop. While microfluidics has successfully demonstrated the<br />

generation of monodisperse bubble populations, these approaches inherently produce low<br />

bubble counts. We introduce a new micro-spray flow regime that generates consistently<br />

high bubble concentrations, in the order of 1010 bubbles mL-1. The technique is shown to<br />

be easy to implement <strong>and</strong> highly reproducible. We also demonstrate that it is possible to<br />

attach particles such as liposomes, loaded with quantum dots or fluorescein in a single step<br />

during the MB formation procedure.<br />

The frequency response of these microbubbles to ultrasound excitation mainly depends on<br />

the mechanical properties of the shell which can be determined using atomic force<br />

microscopy (AFM). The addition of different components to the shell architecture can<br />

change the bubbles mechanical properties <strong>and</strong> therefore its ultrasound response. We<br />

present force spectroscopy measurements with tip-less cantilevers, on microbubbles, to<br />

characterize the shell stiffness of a range of different shell coatings, specifically, Streptavidin,<br />

Q-dots, <strong>and</strong> liposomes. Forces ranging from 20-100 nN were applied to single bubbles to<br />

determine the change in stiffness with applied force. It was observed that the shell stiffness<br />

of phospholipid microbubbles increases linearly with increasing applied force. Adding a<br />

Streptavidin coat doubles the stiffness values of the microbubble, with a dramatic three-fold<br />

increase in shell stiffness observed with addition of Q-dots.


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

16 th & 17 th July 2012<br />

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

Oral Presentation<br />

Vasileios Koutsos<br />

University of Edinburgh<br />

Title: Nanomechanical Properties of <strong>Microbubble</strong>s Studied by Atomic Force Microscopy<br />

Abstract: Stable, haemodynamically inert, hollow, micrometer-size spheres composed of an<br />

ultrathin nanoscale biocompatible shell which encapsulates an inert gas are used as ultrasound<br />

contrast agents <strong>and</strong> are normally referred to as microbubbles. They are smaller than the smallest<br />

blood vessel of a human body to allow improved visualization of the vascular bed <strong>and</strong> differentiate<br />

vascular patterns of tumours non-invasively. Such thin-shelled microstructures can be used as<br />

carriers for drug/gene delivery, which is a topic of much current interest in biomedical research.<br />

Furthermore, with appropriate surface modification such microbubbles can acquire targeting<br />

capability for certain cell types (e.g. cancerous cells). Their nano/micromechanical properties are<br />

extremely important since they have to be stable for considerable time until they rupture/degrade<br />

under specific conditions in order to release their load in the right place <strong>and</strong> at the right time.<br />

Moreover, materials at the nanoscale (such as thin-shell structures) may behave differently to those<br />

on the macroscale, so predicting their mechanical properties presents a challenge. We have<br />

conducted a systematic study of both hard-shelled, polymeric-based 1,2 <strong>and</strong> soft-shelled,<br />

phospholipid-based 3 microbubbles employing microcompression by performing atomic force<br />

microscopy force-distance curves. Individual microbubbles of different diameter were compressed<br />

from few nanometers up to approx. 50% of their initial diameter. We have performed multiple<br />

compressions on individual microbbubles to assess their mechanical robustness <strong>and</strong> measured their<br />

stiffness. Furthermore, we used different mechanical theories to estimate the elastic modulus of the<br />

microbubble shell. In the case of phospholipid-based shells, we have also estimated the effective<br />

elastic modulus of the whole microbubble as (if it was) a homogenous object. We compare <strong>and</strong><br />

discuss our results with other phospholipid systems, namely supported lipid membranes, vesicles <strong>and</strong><br />

cells <strong>and</strong> other recent AFM studies of lipid-coated microbubbles. 4,5 Furthermore, as microbubble<br />

science is exp<strong>and</strong>ing to biological targeting <strong>and</strong> drug/gene delivery, we investigated the<br />

characteristics of molecular targeting using modified lipid-shelled microbubbles (biotin–avidin<br />

chemistry <strong>and</strong> the CD31 antibody) to probe individual Sk-Hep1 hepatic endothelial cells. The<br />

modified (targeted) microbubbles provide a single distribution of adhesion forces with a median of<br />

93 pN. This interaction was assigned to the CD31 antibody–antigen unbinding event <strong>and</strong> proves the<br />

capability of single microbubbles to target cell lines. 6<br />

References: (1) E. Glynos, V. Koutsos, W. N. McDicken, C. M. Moran, S. D. Pye, J. A. Ross, V. Sboros.<br />

Nanomechanics of Biocompatible Hollow Thin-Shell Polymer Microspheres. Langmuir 2009, 25 (13), 7514−752;<br />

(2) E. Glynos, V. Sboros, V. Koutsos. Polymeric thin shells: Measurement of elastic properties at the nanometre<br />

scale using atomic force microscopy. Materials Science <strong>and</strong> Engineering: B 2009, 165 (3), 231−234; (3) E.<br />

Buchner Santos, J. K. Morris, E. Glynos, V. Sboros, V. Koutsos. Nanomechanical Properties of Phospholipid<br />

<strong>Microbubble</strong>s. Langmuir 2012, 28 (13), 5753−5760; (4) J. E. McKendry, C. A. Grant, B. R. G. Johnson, P. L.<br />

Coletta, J. A. Evans, S. D. Evans. Force spectroscopy of streptavidin conjugated lipid coated microbubbles.<br />

Bubble Sci., Eng., Technol. 2010, 2 (2), 48−54; (5) C. A. Grant, J. E. McKendry, S. D. Evans. Temperature<br />

dependent stiffness <strong>and</strong> visco-elastic behaviour of lipid coated microbubbles using atomic force microscopy. Soft<br />

Matter 2011, 8, 1321−1326; (6) V. Sboros, E. Glynos, J. A. Ross, C. M. Moran, S. D. Pye, M. Butler, W. N.<br />

McDicken, S. B. Brown, V. Koutsos. Probing microbubble targeting with atomic force microscopy. Colloids <strong>and</strong><br />

Surfaces B: Biointerfaces 2010, 80 (1), 12−17


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

16 th & 17 th July 2012<br />

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

Oral Presentation<br />

Title: Investigation into the Effects of Ultrasound on Attached <strong>Microbubble</strong>s<br />

Jonathan Loughran<br />

Imperial College London<br />

Abstract: <strong>Microbubble</strong> contrast agents have shown great potential in imaging <strong>and</strong><br />

quantifying blood flow <strong>and</strong> tissue perfusion. Recent advance of targeted microbubbles have<br />

made it possible to detect <strong>and</strong> imaging the distribution of specific molecules of interest<br />

expressed on endothelial walls in vivo using ultrasound. Previous studies have been focused<br />

on developing microbubbles with improved targeting efficiency, acoustic techniques to<br />

facilitate the attachment of such bubbles <strong>and</strong> better detection method, little attention has<br />

been paid to study what happens to the microbubbles once they are attached. In this study<br />

the aim is to study the effects of ultrasound exposure to attached microbubbles. Biotin<br />

coated microbubbles were flown through a phantom coated with streptavidin. Optical<br />

microscopy was used to observe the attached microbubbles in the flow phantom under<br />

ultrasound exposure. The bubble size <strong>and</strong> position were recorded during the exposure of<br />

ultrasound. It was found that even at relatively low acoustic pressure a significant amount of<br />

bubbles detach from the wall <strong>and</strong> flow away, some of them shrink at the same time. The<br />

number of bubble detachment increases with acoustic pressure. The different forces acting<br />

on the bubbles during the exposure was analysed to offer some insights into the detachment<br />

mechanisms.<br />

Additional authors:<br />

Jonathan Loughran1, Charles Sennoga1, Robert Eckersley2, Meng-Xing Tang1*<br />

1 Department of Bioengineering, Imperial College London, London SW7 2AZ, UK<br />

2 Biomedical Engineering Department, King's College London, London. SE1 7EH, UK


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

16 th & 17 th July 2012


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

16 th & 17 th July 2012<br />

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

Oral Presentation<br />

Title: <strong>Microbubble</strong> Imaging in Cancer Theranostics<br />

Paul A Dayton<br />

University of North Carolina<br />

Abstract: Theranostics refers to the combination of diagnostic <strong>and</strong> therapeutic approaches,<br />

typically for assessing response to therapy. Although it is possible for an agent to be used<br />

simultaneously for imaging <strong>and</strong> therapy, by definition, theranostics is fundamentally the<br />

combination of a diagnostic test with a therapeutic approach, <strong>and</strong> thus can also involve<br />

either application of a diagnostic technique followed by a therapeutic, or vice versa.<br />

Theranostic approaches are a key aspect in the drive toward personalized medicine, where<br />

the tailoring of decisions <strong>and</strong> treatment practices for individual patients are based on<br />

diagnostic information specifically from that patient. Microbbubles can be used for imaging,<br />

for therapy, or both simultaneously. In this sense, microbubbles combined with acoustics<br />

may be one of the most universal theranostic tools. In this seminar, we will specifically focus<br />

on microbubble contrast agents <strong>and</strong> contrast imaging techniques for assessment of tumor<br />

growth <strong>and</strong> response to therapy. Both perfusion imaging <strong>and</strong> molecular imaging are shown<br />

to differentiate differences between tumors which respond to therapy <strong>and</strong> those which do<br />

not, at time points earlier than determined by traditional tumor size measurements.<br />

Contrast enhanced microvascular mapping enables the delineation of tortuous tumor<br />

microvasculature associated with angiogenesis. When analyzed with segmentation<br />

techniques, this method can also indicate malignancy based on microvascular structure<br />

alone. These results suggest that contrast enhanced ultrasound may play a substantial role<br />

in noninvasive assessment of tumor response to therapies <strong>and</strong> personalized medicine.


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

16 th & 17 th July 2012<br />

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

Oral Presentation<br />

John A. Hossack<br />

University of Virginia<br />

Title: Catheter-Based Monodisperse <strong>Microbubble</strong> Production for Image-guided Vascular<br />

Therapy<br />

Abstract: Current methods for delivering smooth muscle cell antiproliferative drugs to the vessel<br />

wall following percutaneous-transluminal angioplasty <strong>and</strong> stent placement are limited in terms of<br />

ability to tailor drug choice <strong>and</strong> delivery profile to a particular patient’s lesion. We have been<br />

investigating intravascular ultrasound (IVUS)-based methods for imaging <strong>and</strong> drug delivery using<br />

ultrasound in combination with drug associated microbubbles. Following recognition in the field that<br />

precise control of both ultrasound parameters <strong>and</strong> microbubble properties are preferable to achieve<br />

efficacy <strong>and</strong> safety simultaneously, we have been investigating microfluidics-based approaches to<br />

microbubble production that yield mono-disperse microbubbles. In this paper, we present a new<br />

method for supplying the liquid (shell) phase to a flow focusing microfluidic device (FFMD). In place<br />

of individual tubes supplying each inlet, the FFMD is coupled to a pressurized liquid-filled chamber<br />

that drives the fluid from inside the catheter directly into the channels of the microfluidics device.<br />

This method eliminates bulky interconnects, significantly reducing the complexity of the device <strong>and</strong><br />

simplifies device parallelization enabling increased microbubble production rate <strong>and</strong> distributed<br />

production around the circumference of the catheter. Flooded FFMDs were fabricated <strong>and</strong><br />

production properties quantified. The minimum microbubble size produced was 8.1±0.3 µm. The<br />

maximum production rate from a single nozzle FFMD was 660,000 microbubbles/s. Production was<br />

1.5-fold higher when using a parallel device. <strong>Microbubble</strong>s produced in situ in a gelatin flow phantom<br />

were imaged in real time using a co-aligned IVUS <strong>and</strong> an external linear array clinical scanner<br />

demonstrating contrast enhanced imaging of the vessel. In addition, delivery of a co-injected dye was<br />

enhanced in areas subject to both microbubbles <strong>and</strong> ultrasound. Therefore, in situ produced<br />

microbubbles, coupled to an IVUS catheter, have the potential to contribute to the long-term goal of<br />

providing real time image guided diagnosis <strong>and</strong> treatment of vascular disease.<br />

(a)<br />

(b)<br />

(c)<br />

(a) Schematic of an IVUS catheter<br />

functionalized with a flooded FFMD<br />

for producing mono-disperse<br />

microbubbles in situ. (b) IVUS image<br />

of vessel lumen enhanced with realtime<br />

production of microbubbles. (c)<br />

Enhanced delivery of a fluorophore to<br />

the vessel wall subject to both<br />

ultrasound <strong>and</strong> microbubbles


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

16 th & 17 th July 2012<br />

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

Oral Presentation<br />

James McLaughlan<br />

University of Leeds<br />

Title: The Acoustic Evaluation of Liposome-loaded <strong>Microbubble</strong>s for Targeted Cancer<br />

Therapy<br />

Abstract: The use of microbubbles as drug or gene delivery vehicles is a highly active area of research, as they<br />

can be targeted to specific cell populations delivering therapeutic agents to a localised area. Attachment of<br />

drug filled liposomes to the outside of microbubbles is one mechanism. The attachment of liposomes onto the<br />

shell can affect their ultrasound response, which is important for both imaging <strong>and</strong> therapeutic applications as<br />

it could result in expansion only oscillations <strong>and</strong>/or increasing the destruction threshold.<br />

Phospholipid microbubbles (1-10 µm), manufactured in a microfluidic flow cell, were conjugated with<br />

fluorescein filled liposomes (200 nm). A 4 MHz windowed tone burst was used to excite a microbubble<br />

population (0.5x10 6 MB/ml) <strong>and</strong> the 90° scatter was detected using a 1 mm needle hydrophone. The<br />

destruction threshold was determined in a flow cell mounted on top of an inverted microscope. Bright field<br />

images taken before <strong>and</strong> immediately after the ultrasound exposure were used to estimate change in<br />

population for a given mechanical index (MI). The acoustic response <strong>and</strong> destruction threshold was measured<br />

with <strong>and</strong> without attached liposomes.<br />

The figure below, (a) shows bright field <strong>and</strong> fluorescent images of loaded <strong>and</strong> unloaded microbubbles. Only the<br />

loaded microbubbles are visible under fluorescence due to the green fluorophore encapsulated within the<br />

liposomes. Fig. (b) <strong>and</strong> (c) show the scattered frequency spectrum from a population of microbubbles at 50 <strong>and</strong><br />

300 kPa (MI 0.025 <strong>and</strong> 0.15). The subharmonic ratio shown in (d) demonstrates that for low MI the loaded<br />

microbubbles have a significantly larger subharmonic emission than the unloaded microbubbles. Once an MI of<br />

0.1 is exceeded the unloaded microbubbles increase in subharmonic emission, however as (e) <strong>and</strong> (f) show, this<br />

could be due to microbubble destruction <strong>and</strong> release of free gas bubbles. The liposome loading has a negligible<br />

effect on the destruction of microbubbles since (e) <strong>and</strong> (f) show that for both types 90% of the population is<br />

destroyed for a MI of 0.4.<br />

Liposome loading of microbubbles causes an increase in their harmonic response, specifically the subharmonic,<br />

for low ultrasound pressures (MI). Once microbubble destruction is present the acoustic response can no<br />

longer be attributed to the shell of the microbubble, since free gas bubbles could also be present.


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

16 th & 17 th July 2012<br />

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

Oral Presentation<br />

Title: Ultrasound Contrast Agents: from Imaging to Therapy<br />

Ayache Bouakaz<br />

Université François Rabelais, Tours<br />

Abstract: Contrast agents, consisting of tiny gas microbubbles are currently approved for<br />

ultrasound imaging in cardiology <strong>and</strong> in radiology. The microbubbles have a mean size of<br />

about 3 microns <strong>and</strong> are encapsulated by a thin biocompatible layer. Multiple clinical studies<br />

have established the utility of ultrasound contrast agents (UCA) in improving accuracy of<br />

echography for the diagnosis of many diseases <strong>and</strong> in reducing health care costs by<br />

eliminating the need for additional testing.<br />

Future clinical applications of UCA extend beyond imaging <strong>and</strong> diagnostic, offering to<br />

ultrasound technology a new therapeutic dimension. Since a few years, novel therapeutic<br />

strategies are explored using microbubbles <strong>and</strong> ultrasound. Current data demonstrate that<br />

in the presence of microbubbles, ultrasound waves destabilize transiently the cell membrane<br />

allowing the incorporation of drugs, including genes into the cells. Moreover, the<br />

microbubbles might be used as a drug vehicle to achieve a spatially <strong>and</strong> temporally<br />

controlled local release. Besides, microbubbles are able to identify diseased targets through<br />

specific targeting. The talk will focus on the current knowledge of microbubble targeted<br />

therapy fundamentals <strong>and</strong> basic mechanisms. New challenges will be discussed.


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

16 th & 17 th July 2012<br />

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

Oral Presentation<br />

Carmel M Moran<br />

University of Edinburgh<br />

Title: <strong>Characterisation</strong> of Ultrasonic Contrast <strong>Microbubble</strong>s for Preclinical Applications<br />

Abstract: Research using small animals, continues to play a key role in the advancement of<br />

medical, biological <strong>and</strong> veterinary sciences. In particular the mouse model is popular as a<br />

research model as over 85% of the genomic sequences in mice are identical to those found<br />

in humans. In addition, the use of zebra-fish (Danio rerio) has increasingly become an<br />

important tool in medical research with application to the investigation of an extensive<br />

range of human diseases. Its small size <strong>and</strong> corresponding small space requirements,<br />

relatively rapid sexual maturity (approximately 3 months), generation of hundreds of<br />

embryos every week <strong>and</strong> external fertilisation makes it increasingly one of the most<br />

accessible animals for developmental studies. The use of high resolution ultrasound coupled<br />

with light anaesthesia ensures that preclinical ultrasound is a relatively inexpensive , high<br />

throughput technique useful for determining the phenotype of these animals. Ultrasonic<br />

contrast microbubbles, in particular targeted agents are increasingly used to deliver either<br />

genes or drug payload to specific sites within these animals.<br />

In this presentation I will focus on our work on measuring the acoustic properties of<br />

contrast agents at frequencies used for preclinical imaging <strong>and</strong> the implications of our<br />

results on in-vivo animal imaging. We use a Vevo 770 (Visualsonics Inc, Toronto) <strong>and</strong> four<br />

transducers (RMV 710, RMV 707, RMV 704 <strong>and</strong> RMV 711) <strong>and</strong> employ a broadb<strong>and</strong><br />

substation technique to measure the acoustic properties of tissue-mimicking-material<br />

(TMM) <strong>and</strong> three commercially available contrast agents (SonoVue, Definity <strong>and</strong><br />

Micromarker) over the frequency range from 10-47MHz. Specficially, we have measured<br />

the contrast-to-TMM ratio <strong>and</strong> the attenuation of the contrast agents at non-destructive<br />

pressures. Our preclinical studies have focussed on the use of commercially available<br />

clinical contrast agents <strong>and</strong> the settings required to optimise these agents for visualisation<br />

within our preclinical models.


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

16 th & 17 th July 2012<br />

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

Oral Presentation<br />

Title: Multi-modal Contrast Agents: Photoacoustic Imaging <strong>and</strong> Therapy<br />

Jithin Jose<br />

Visualsonics<br />

Abstract: Photoacoustic (PA) imaging is a hybrid imaging modality, for non-invasive<br />

detection of tissue structural <strong>and</strong> functional anomalies. The technique combines the<br />

advantageous properties of optical <strong>and</strong> ultrasound imaging. It provides the excellent<br />

contrast achieved in optical techniques with the high spatial resolution of ultrasound<br />

imaging. Some important applications of this technique include breast cancer detection, skin<br />

cancer visualization <strong>and</strong> small animal imaging. Multifunctional microbubbles (MBs) are<br />

contrast agents integrating multiple disease‐targeting, imaging <strong>and</strong> therapeutic functions.<br />

Multifunctional MBs represent an enabling technology for many potential applications in the<br />

field of photoacoustic imaging. Highly absorbing optical contrast agents are encapsulated in<br />

the bubbles <strong>and</strong> it can be used as multi-modal contrast agents for high resolution ultrasound<br />

<strong>and</strong> photoacoustic imaging. Upon activation the bubbles may introduce significant contrast<br />

enhancement <strong>and</strong> facilitate a variety of potential clinical <strong>and</strong> preclinical applications<br />

including photo thermal therapy.


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

16 th & 17 th July 2012<br />

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

Oral Presentation<br />

Nicola Ingram<br />

University of Leeds<br />

Title: Microfluidically-Generated <strong>Microbubble</strong>s for Molecular Imaging <strong>and</strong> Drug Delivery in<br />

Colorectal Cancer – Pre-Clinical Evaluation<br />

Abstract: Drug delivery to solid tumours <strong>and</strong> metastatic lesions remains a major problem in<br />

the treatment of cancer. Ultrasound (US) contrast agents or microbubbles (MBs) are<br />

currently being developed for their theragnostic capabilities, potentially linking US diagnosis<br />

with drug delivery <strong>and</strong> monitoring of therapeutic response. To this end, the Leeds<br />

<strong>Microbubble</strong> Consortium has developed a novel on-chip flow focusing microfluidic (µF)<br />

device capable of reproducibly generating MBs in sufficient quantity <strong>and</strong> quality for use in<br />

vivo.<br />

We have performed extensive in vitro <strong>and</strong> in vivo evaluation of our µF MBs to analyse their<br />

performance in US imaging <strong>and</strong> targeted drug delivery. Increased µF MB lifetime in vitro<br />

was shown to be dependent on shell lipid concentration. Toxicity was assessed in vitro <strong>and</strong><br />

found to be comparable to Micromarker (Bracco). In vitro binding to endothelial <strong>and</strong><br />

colorectal cancer (CRC) cells using µF MBs conjugated with anti-VEGFR2 antibodies was<br />

examined by cell flow assays. Significantly higher binding was found with VEGFR2 positive<br />

cells compared with negative cells or with µF MBs conjugated to isotype control antibodies.<br />

In vivo imaging in mouse colorectal cancer models with µF MBs showed clear contrast of the<br />

vascular space <strong>and</strong> generated wash-in curves for quantitative evaluation.<br />

We have also studied the spatial <strong>and</strong> temporal expression of VEGFR2 in a CRC model to<br />

determine its suitability for molecular imaging <strong>and</strong> targeted delivery. Using histological<br />

methods, we have shown a peak in CD31+ blood vessels that also expressed VEGFR2 at<br />

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

volume. This was comparable with the signal obtained from VEGFR2-targeted MBs in vivo<br />

where smaller tumours demonstrated a greater subtracted molecular signal than larger<br />

tumours.<br />

To develop on-chip assembled µF MBs for targeted drug delivery, we have optimised<br />

luciferin encapsulation into liposomes to perform proof-of-principle experiments in<br />

luciferase expressing cells in vitro <strong>and</strong> in tumours in vivo. In vitro testing using<br />

bioluminescence demonstrated relatively stable encapsulation of luciferin. Injection of<br />

luciferin liposomes into a luciferase expressing human CRC xenograft showed clear tumour<br />

specific signal. This approach will facilitate pre-clinical evaluation of MB vehicles for UStriggered<br />

targeted drug delivery.<br />

Additional Authors:<br />

Nicola Ingram, Elizabeth Valleley, Tony Evans 1 , Pam Jones, Alex Markham <strong>and</strong> Louise Coletta;<br />

Leeds Institute of Molecular Medicine; 1 Leeds Institute of Genetics, Health <strong>and</strong> Therapeutics


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

16 th & 17 th July 2012<br />

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

Oral Presentation<br />

Title: Therapeutic Uses of <strong>Microbubble</strong>s: Heating, Erosion <strong>and</strong> Delivery.<br />

Robin Clevel<strong>and</strong><br />

University of Oxford<br />

Abstract: The biomedical use of microbubbles was initially driven by the desire to produce<br />

ultrasound contrast agents for diagnostic imaging. However, microbubbles also have utility<br />

as a mechanical catalyst for therapeutic use ultrasound <strong>and</strong> a few applications will be<br />

reviewed here. In focused ultrasound surgery where HIFU is used to thermally ablate tissue<br />

the presence of the appropriate number of cavitation bubbles can result in significant<br />

increases in heating rate—resulting in shorter treatment time. A challenge is that cavitation<br />

nucleation is stochastic <strong>and</strong> hard to control <strong>and</strong> microbubbles can act as cavitation nuclei<br />

with a well characterized threshold that allows for reliable generation of cavitation bubbles.<br />

A second application, is non-thermal tissue fractionation, also known as histotripsy, where<br />

the mechanical action of cavitation bubbles can be used to erode tissue. This is a second<br />

case where having a well defined cavitation nucleation threshold allows for better control of<br />

the tissue damage <strong>and</strong> microbubbles can play that role in this application as well. Finally,<br />

gentle cavitation activity in the vicinity of cells, results in microstreaming that can disrupt cell<br />

membranes allowing for sonoporation. This has application in delivering drugs through the<br />

endothelium, in particular, the blood-brain barrier.


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

16 th & 17 th July 2012<br />

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

Oral Presentation<br />

Title: Contrast Enhanced Ultrasound in Clinical Practice<br />

Jane Smith<br />

St James’s University Hospital<br />

Abstract: Contrast Enhanced Ultrasound (CEUS) has been in clinical use for the last 2<br />

decades. Originally used as cardiac agents <strong>and</strong> then as vascular agents, they established the<br />

patency of blood vessels <strong>and</strong> identified stenoses - for example in the carotid artery. First<br />

generation microbubble agents were relatively unstable, easily burst (even on low power<br />

settings) <strong>and</strong> unable to withst<strong>and</strong> repeated passage through capillary beds.<br />

Improvements in Doppler technology started to render contrast redundant, until it was<br />

discovered that deliberate microbubble destruction, in response to increased pressure, was<br />

able to briefly highlight liver metastases 1 - a technique known as stimulated acoustic<br />

emission.<br />

Although this technique was never a suitable tool for routine practice, it led to the<br />

investigation of microbubbles in detecting <strong>and</strong> characterising liver lesions, giving rise to a<br />

second generation of stable microbubble agents <strong>and</strong> developments in technology which<br />

allow operators to evaluate the contrast patterns for diagnostic purposes.<br />

Currently CEUS is routinely used to characterise incidental liver lesions, <strong>and</strong> also to detect<br />

secondary liver cancers. However its use can be translated into various other applications<br />

involving abdominal organs <strong>and</strong> vascular structures 2 <strong>and</strong> CEUS has become part of the<br />

patient pathway in many European <strong>and</strong> Canadian diagnostic centres.<br />

<strong>Microbubble</strong> agents have the advantage of being safe, non-nephrotoxic <strong>and</strong> easily tolerated<br />

by patients. CEUS has been shown to be at least as good as other imaging (for example MRI<br />

<strong>and</strong> CT) in characterising liver lesions <strong>and</strong> detecting metastases 3 , <strong>and</strong> it is valuable as a quick<br />

<strong>and</strong> accurate problem solver, often obviating the need for further diagnostic imaging.<br />

It’s use is slowly broadening in clinical practice to include other applications, supporting<br />

innovative <strong>and</strong> cost effective patient pathways.<br />

References:<br />

1.: Blomley et al: Improved Imaging of Liver Metastases with Stimulated Acoustic Emission in the Late Phase of<br />

Enhancement with the US Contrast Agent SH U 508A: Early Experience , Radiology, February 1999, 210, 409-416.<br />

2 The EFSUMB Guidelines <strong>and</strong> Recommendations on the Clinical Practice of Contrast Enhanced Ultrasound (CEUS) Update<br />

2011 on non-hepatic applications. 20/12/2011. http://www.efsumb.org/guidelines/guidelines01.asp<br />

3. Trillaud et al. Characterization of focal liver lesions with Sonovue-enhanced sonography: international multicenter-study<br />

in comparison to CT <strong>and</strong> MRI. World J Gastroenterol 2009; 15(30): 3748-56


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

16 th & 17 th July 2012


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

16 th & 17 th July 2012<br />

Number Name Title<br />

1 Mostafa Abdelrahman<br />

2 Radwa Abou-Saleh<br />

Poster Presentations<br />

Frequency dependence of flow sensitivity in Power<br />

Doppler <strong>and</strong> CEUS Imaging<br />

Developing <strong>and</strong> investigating therapeutic<br />

microbubbles<br />

3 Jennifer Bain Biomimetic synthesis of magnetic nanoparticles.<br />

4 Jonathan Blockley<br />

5 Jonathan Casey<br />

6 Michael Conneely<br />

Ultrasound measurements of known microbubble<br />

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

Single Bubble Sub-harmonic <strong>Characterisation</strong> of Lipid<br />

Shelled <strong>Microbubble</strong>s: Effect of Functionalization <strong>and</strong><br />

Adherence<br />

Computationally assisted design <strong>and</strong> experimental<br />

validation of a novel ‘flow-focused’ microfluidics chip<br />

for generating monodisperse microbubbles.<br />

7 Kevin Critchley Cadmium Free Quantum Dots for Biomedical Imaging<br />

8 Pratik Desai Ultrasound mediated microbubble generation<br />

9 Joe Fiabane<br />

10 Sevan Harput<br />

A high-Reynolds number microfluidic device for<br />

microbubble generation<br />

Separating the Second Harmonic Response of Tissue<br />

<strong>and</strong> <strong>Microbubble</strong>s using Bispectral Analysis<br />

11 George Heath Actin Coated <strong>Microbubble</strong>s<br />

12 Nicola Ingram<br />

13 Fairuzeta Ja'afar<br />

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

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

delivery using contrast-enhanced high-frequency<br />

ultrasound.<br />

Binding Differentials of <strong>Microbubble</strong>s to Activated<br />

versus Non-Activated HUVECs


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

16 th & 17 th July 2012<br />

14 Tom Kokhuis Mutually interacting targeted microbubbles<br />

15 Klazina Kooiman Binding area of targeted microbubbles<br />

16 Gael Leaute<br />

17 Jonathan Loughran<br />

18 Gemma Marston<br />

Finite element modelling of non-spherical<br />

microbubble dynamics in the vicinity of a soft<br />

membrane<br />

Investigation into the Effects of Ultrasound on<br />

Attached <strong>Microbubble</strong>s<br />

Imaging the development of functional tumour<br />

vasculature, <strong>and</strong> the effect of combretastatin A-4 on<br />

tumour blood flow rate in vivo<br />

19 Jono McKendry Cerato Ulmin a new UCA shell material?<br />

20 Sally Peyman<br />

21 Linsey Phillips<br />

22 Ben Raiton<br />

23 Sara Shakil Qureshi<br />

24 Peter Smith<br />

25 Peter Smith<br />

High production rates of therapeutic microbubbles for<br />

targeted drug delivery.<br />

New Nanodroplets Increases Tissue Ablation in<br />

Response to High Intensity Focused Ultrasound<br />

Single-well acoustic tweezers using ultrasonic<br />

travelling waves.<br />

Hardware accelerated beamforming for microbubble<br />

detection<br />

Composite High Frequency, Low Frequency System for<br />

Pre-Clinical <strong>Microbubble</strong> Sonoporation<br />

Development of an Ultrasound Array Research<br />

Platform (UARP)


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

16 th & 17 th July 2012<br />

Poster Number: 1<br />

Mostafa Abdelrahman<br />

University of Leeds<br />

Title: Frequency dependence of flow sensitivity in Power Doppler <strong>and</strong> CEUS Imaging<br />

Abstract:<br />

Introduction:<br />

The task of detecting <strong>and</strong> measuring tumour blood flow is critical for both pre-clinical<br />

research <strong>and</strong> clinical diagnosis. It is well known that this is enhanced if contrast bubbles is<br />

used, but the optimisation of the scanning conditions continues to be somewhat<br />

controversial, especially for high frequency pre-clinical scanning. In addition, the role of<br />

Power Doppler is also unclear.<br />

In this study, we have used repeat scanning under a variety of conditions to compare <strong>and</strong><br />

optimise sensitivity to blood flow.<br />

Materials <strong>and</strong> methods:<br />

DLD1 colorectal cancer cell xenografts were grown subcutaneously in CD-1 nude mice. The<br />

animals were scanned using Power Doppler ultrasound (PDUS), contrast-enhanced PDUS<br />

(CE-PDUS), <strong>and</strong> contrast-enhanced ultrasound (CEUS). Images were acquired in 2D <strong>and</strong> 3D<br />

<strong>and</strong> at 25- <strong>and</strong> 40-MHz frequencies. After imaging, mice were sacrificed, imaged with PDUS<br />

(control) <strong>and</strong> the xenografts were then fixed <strong>and</strong> processed for histology <strong>and</strong><br />

immunohistochemistry. Images from live <strong>and</strong> dead animals were scored for colour pixel<br />

density <strong>and</strong> functional parameters, such as peak enhancement (PE) <strong>and</strong> perfusion rate<br />

(slope), were derived from CEUS time-intensity-curves (TIC).<br />

Results: PDUS was found to have a very poor signal to noise ratio (live animals scoring to<br />

dead animals scoring) due to tissue clutter <strong>and</strong> motion artifacts. However, it showed<br />

significant enhancement in flow detectability after the injection of bubbles. The<br />

enhancement was comparable for the 25- <strong>and</strong> 40-MHz transducers <strong>and</strong> was only significant<br />

in 2D imaging. CEUS was found to have higher detectability than CE-PDUS in 2D <strong>and</strong> 3D, <strong>and</strong><br />

that was more evident at 25-MHz. At 40-MHz there was a significant correlation between 3D<br />

imaging <strong>and</strong> PE (R2 = 0.74 (p < 0.001) <strong>and</strong> 0.96 (p < 0.0001) for 3D PDUS <strong>and</strong> 3D CEUS<br />

respectively), <strong>and</strong> at 25-MHz there was significant correlation between 2D CEUS <strong>and</strong> PE (R2 =<br />

0.74, p < 0.0001) <strong>and</strong> between 3D CEUS <strong>and</strong> TIC slope (R2 = 0.74 (p < 0.005))<br />

Conclusion: PDUS without bubbles is not reliable for the comparison of tumour vascularity<br />

between animals. Although 40-MHz transducer provided superior spatial depiction of the<br />

vascular network, the 25-MHz transducer had superior sensitivity to blood flow, especially in<br />

the CEUS mode.


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

16 th & 17 th July 2012<br />

Poster Number: 2<br />

Title: Developing <strong>and</strong> investigating therapeutic microbubbles<br />

Radwa Abou-Saleh<br />

University of Leeds<br />

Abstract: <strong>Microbubble</strong>s have been used previously as ultrasound-assisted drug delivery<br />

systems, as they can be targeted towards <strong>and</strong> imaged at the desired regions to enhance the<br />

drug therapy action. Conjugates of liposomes <strong>and</strong> gas filled microbubbles have evolved as a<br />

better route for drug delivery using ultrasound to rupture the microbubble <strong>and</strong>/or<br />

liposomes, <strong>and</strong> therefore releasing the drug at the required location. The frequency<br />

response of microbubbles to ultrasound excitation mainly depends on the mechanical<br />

properties of the shell which can be determined using atomic force microscopy (AFM). The<br />

addition of different components to the shell architecture can change the bubbles<br />

mechanical properties <strong>and</strong> therefore ultrasound response.<br />

Here we are preparing microbubbles with a phospholipid shell in a microfluidic device with<br />

different coatings. For drug delivery purposes, liposomes encapsulating Quantum Dots (Qdots),<br />

Fluorescein or Luciferin is attached to the bubbles <strong>and</strong> their delivery dose estimated.<br />

Force spectroscopy measurements with tip-less cantilevers, on microbubbles, have been<br />

performed to characterize the shell stiffness of a range of different shell coatings,<br />

specifically, Streptavidin <strong>and</strong> Q-dots. Forces ranging from 20-100 nN were applied to single<br />

bubbles to determine the change in stiffness with applied force. It was observed that the<br />

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

Additional authors:<br />

Radwa H. Abou-Saleh 1 , Sally A. Peyman 1 , Neil H.Thomson 1,2 <strong>and</strong> Stephen D.Evans 1<br />

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

2. Dept. of Oral Biology, Leeds Dental Institute, LS2 9LU, UK


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

16 th & 17 th July 2012<br />

Poster Number: 3<br />

Title: Biomimetic synthesis of magnetic nanoparticles.<br />

Jennifer Bain<br />

University of Leeds<br />

Abstract: Magnetotactic bacteria use an internal vesicle synthesis process for the formation<br />

of magnetite nanoparticles (MNPs) within the vesicle (magnetosomes). This biological<br />

process displays high levels of control <strong>and</strong> monodispersity with respect to MNPs. Formation<br />

within an internal vesicle with the aid of biomineralisation proteins allows such control to be<br />

realised.<br />

Exploitation of this process via biomimetic precipitation of MNPs within an asymmetric<br />

vesicle to form an artificial magnetosome is being developed, which could have applications<br />

in a range of biomedical <strong>and</strong> data storage nanotechnologies. The simplest route to<br />

magnetite MNP formation is co-precipitation. If it is possible to engineer co-precipitation<br />

inside an asymmetrical vesicle, forming a single magnetite crystal, it will essentially mimic<br />

the formation <strong>and</strong> ideally the behavior of a magnetosome. Magnetospirillum magneticum<br />

protein; Mms6, has been expressed <strong>and</strong> purified in previous work1. Incorporation of Mms6<br />

into this system should ensure MNP uniformity with regards to both the particle<br />

composition <strong>and</strong> consequently magnetic properties. To date three routes to co-precipitation<br />

have been established <strong>and</strong> optimized for the system, early results have shown encapsulation<br />

within an amphiphilic AB-block-copolymer vesicle to be promising.<br />

Additional authors:<br />

Jennifer Bain, Dr. Jonathan Bramble, Dr. Andrea Rawlings, Dr. Sarah Stanil<strong>and</strong>


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

16 th & 17 th July 2012<br />

Poster Number: 4<br />

Jonathan Blockley<br />

Institution: University of Leeds<br />

Title: Ultrasound measurements of known microbubble populations on a st<strong>and</strong>ard<br />

microscope stage<br />

Abstract: Ultrasound measurements were made on lipid microbubbles in the frequency<br />

range 1-15 MHz. <strong>Microbubble</strong>s were produced using a microfluidic technique that gives a<br />

narrow dispersion of microbubble size, <strong>and</strong> some control over mean radius.<br />

The apparatus allows ultrasound attenuation measurements to be made on a small<br />

population of microbubbles while simultaneously imaging with a st<strong>and</strong>ard upright optical<br />

microscope. The microscope can be focused at a depth of choice to view the microbubbles<br />

as they rise, or survey the microbubbles after they have risen to the top of the chamber.<br />

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

Attempts to confine the microbubbles in the observation area for prolonged testing have<br />

been partially successful, but the technique is highly size dependent.


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

16 th & 17 th July 2012<br />

Poster Number: 5<br />

Jonathan Casey<br />

Imperial College London<br />

Title: Single Bubble Sub-harmonic <strong>Characterisation</strong> of Lipid Shelled <strong>Microbubble</strong>s: Effect of<br />

Functionalization <strong>and</strong> Adherence<br />

Abstract:<br />

Introduction: The detection <strong>and</strong> utilisation of targeted microbubble (MB) ultrasound contrast agents<br />

for molecular imaging <strong>and</strong> targeted drug/gene delivery can be improved by making use of acoustic<br />

phenomena arising as a result of targeting or proximity to the target area[1]. Due to a number of<br />

factors; for example low MB concentration at the target site <strong>and</strong> signal masking by bubble subpopulations<br />

of varying size, these differences are small <strong>and</strong> difficult to deduce from st<strong>and</strong>ard bulk<br />

acoustic methods used for the characterisation of MBs. To better underst<strong>and</strong> how the dynamics of<br />

whole populations of MBs are affected by factors such as, sub-population size, shell chemistry <strong>and</strong><br />

targeting it is essential to first underst<strong>and</strong> the acoustic response of single MBs. Previous work by the<br />

author has examined the fundamental <strong>and</strong> second harmonic response of MBs under adhesion [2].<br />

The objective of this study was to examine how the sub harmonic (SH) response of MBs changes as a<br />

function of adhesion. SH scattering shows particular promise for MB imaging because the<br />

surrounding tissue does not produce any SH non-linearity.<br />

Methods: Single bubble acoustics was performed on in-house produced phospholipid MBs ranging in<br />

diameter from 1-10μm. The testing rig consisted of a pair of focussed transducers (transmit<br />

transducer centre frequency 3.5 MHz, receive transducer centre frequency 1 MHz) <strong>and</strong> a 40x<br />

microscope objective lens co-focally aligned on a capillary fibre. MBs were insonated with 2 MHz,<br />

10cycle, 275 KPa peak negative pressure pulses to establish the acoustic characteristics.<br />

To determine the effect of binding MB were examined either bound to the capillary fibre via a Biotin-<br />

Streptavidin bond or left unattached by the removal of one or both or the binding elements.<br />

Results/Discussion<br />

SH scattering was detected in all testing regimes. In agreement with theory the generation of SHs is<br />

seen to be a size dependant phenomenon, occurring predominantly when the MBs are activated<br />

near their resonant frequency. There was no measureable difference detected in this SH scattering<br />

between the adherent <strong>and</strong> free MBs at this size. SH scattering was also detected when a MB<br />

was driven at twice the resonant frequency. In this size region adherent MBs exhibited an increase in<br />

SH scattering in comparison with their unattached counterparts. Increasing ability to control the size<br />

distribution statistics of MBs the differences identified between adherent <strong>and</strong> free MBs could play an<br />

important role in their identification in targeted ultrasound diagnosis <strong>and</strong> therapy.<br />

MBs could play an important role in their identification in targeted ultrasound diagnosis <strong>and</strong> therapy.<br />

References:<br />

1. Dayton, P.A., Improving Technology for Molecular Imaging with Ultrasound. IEEE, 2009.<br />

2. Casey, J et al., Single bubble Acoustic <strong>Characterisation</strong> <strong>and</strong> Stability Measurement of Adherent<br />

<strong>Microbubble</strong>s, IEEE IUS-2011<br />

Additional authors:<br />

Jonathan Casey1, Charles Sennoga2, Meng-Xing Tang2, Robert Eckersley3<br />

1Imaging Sciences Dept, Imperial College London, United Kingdom, 2Dept of Bioengineering, Imperial<br />

College London, United Kingdom, 3Biomedical Engineering Dept, Kings College London, United<br />

Kingdom


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

16 th & 17 th July 2012<br />

Poster Number: 6<br />

Michael Conneely<br />

University of Dundee<br />

Title: Computationally assisted design <strong>and</strong> experimental validation of a novel ‘flow-focused’<br />

microfluidics chip for generating monodisperse microbubbles.<br />

Abstract: Whilst initially developed as a diagnostic aid to improve echogenicity in ultrasound<br />

imaging, gas-filled lipid microbubbles are now emerging as a next generation 'theranostic'<br />

tool in the medical arena. Here, their therapeutic potential has now been realized through<br />

their unique capability to deliver molecular species such as drugs <strong>and</strong> genes by means of<br />

disrupting the cell membrane in response to ultrasound wave stimulus.<br />

The main aim for the present study was to computationally validate an in-house design for a<br />

novel glass microfluidic chip. The chip features a distinctive junction geometry, wherein the<br />

liquid <strong>and</strong> gas phases meet, <strong>and</strong> more importantly, produce stable monodisperse<br />

microbubbles (which may exhibit a more easily controlled, <strong>and</strong> thus clinically reliable<br />

response to pulsed ultrasound). The shell material in our microbubbles was composed of<br />

PEGylated neutral lipid mixed with cholesterol <strong>and</strong> including a gas core of either nitrogen or<br />

perfluorobutane. <strong>Microbubble</strong> diameters were formed within the range: 2-10μm depending<br />

upon the liquid <strong>and</strong> gas flow parameters. COMSOL Multiphysics was employed to create a 2-<br />

D simulation of the chip, <strong>and</strong> for validation, this was compared directly against our<br />

observational results. Figure 1 shows the bubble formation process as observed with a high<br />

speed camera (above) <strong>and</strong> as modeled in COMSOL (below). Future parameterized studies<br />

implemented via COMSOL, will allow us to ascertain the factors that will inform next<br />

generation chip-design, as well as the optimum operational parameters.<br />

Additional authors:<br />

Michael Conneely* 1 , Vikas Hegde 2 , Hans Rolfsnes 1 , Adrian Mason 2 , Charlie Main 1 , Frances JD Smith 2 ,<br />

WH Irwin McLean 2 & Paul A Campbell 1,2<br />

1 Carnegie Physics Laboratory, University of Dundee, Dundee DD1 4HN. Scotl<strong>and</strong> UK<br />

2 Division of Molecular Medicine, University of Dundee, Dundee DD1 4HN. Scotl<strong>and</strong> UK<br />

Figure 1. Comparison between experimental (top) <strong>and</strong> numerical model (below) results.


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

16 th & 17 th July 2012<br />

Poster Number: 7<br />

Title: Cadmium Free Quantum Dots for Biomedical Imaging<br />

Kevin Critchley<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)<br />

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 />

16 th & 17 th July 2012<br />

Poster Number: 8<br />

Title: Ultrasound mediated microbubble generation<br />

Pratik Desai<br />

University of Sheffield<br />

Abstract: Ultrasound mediated drug delivery via microbubbles is a technique that could<br />

potentially revolutionise the treatment of tumours <strong>and</strong> is currently under study in many<br />

research laboratories. Before being used as drug delivery vectors, however, microbubbles<br />

need to be manufactured: a monodisperse distribution centred at a diameter smaller than 6-<br />

8 µm is the desired target (to account for the capillary system). In this paper, the authors<br />

investigate the use of an acoustic modulation to pilot the size of bubbles produced by a<br />

pressure-controlled microfluidic actuator. In this study, the authors added a bespoke diffuser<br />

to the pre-conditioning actuator, so as to induce the production of smaller bubbles & further<br />

increase the throughput of the system. A resonating cavity was used to induce vibration of<br />

the diffuser at a designed frequency. As shown in Figure 1, the performance of the system<br />

changed as the acoustic oscillator approached resonance: the throughput of the microfluidic<br />

actuator was increased manifold <strong>and</strong> generated a narrower bubble size distribution, coupled<br />

with a decrease in bubble size (75 % between 3-4 μm) The design at NPL of an in house<br />

detection system (Figure 2), capable of detecting bubbles up to a size of 3 μm, was an<br />

important aspect of the project. Inverse problems posed by the Fredholm integral, necessary<br />

to garner a size distribution from the raw data, were solved numerically. Use of protein /lipid<br />

coating & PFC core will further stabilise the bubble & reduce size. If successful, the concept<br />

could lead to a substantial reduction in the cost of the currently expensive conventional<br />

encapsulated microbubbles for medical imaging, even in other microfluidic-based<br />

manufacturing techniques.<br />

Figure 1 Bubble Size Distribution at different frequencies Figure 2 Rig<br />

Additional authors:<br />

P.D. Desai* 1 , G. Memoli 2 , D. Kuvshinov 1 <strong>and</strong> W. Zimmerman 1<br />

Affiliation: 1 Department of Chemical & Biological Engineering, University of Sheffield; 2 Acoustics Group,<br />

National Physical Laboratory


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

16 th & 17 th July 2012<br />

Poster Number: 9<br />

Joe Fiabane<br />

Robert Gordon University, Aberdeen<br />

Title: A high-Reynolds number microfluidic device for microbubble generation<br />

Abstract: A key requirement of the generation of microbubbles for use in drug delivery is<br />

narrow size distribution <strong>and</strong> close control over size, as this is a dominant factor governing<br />

ultrasound response. Microfluidic devices offer superior performance in this regard,<br />

compared with conventional production methods such as sonication <strong>and</strong> mechanical<br />

agitation. However previously reported devices require very small internal geometry,<br />

necessitating high operating pressures <strong>and</strong> resulting in frequent blockages. Hence it is<br />

desirable to develop a device which produces microbubbles much smaller than its internal<br />

channel widths. The best device thus far reported produces microbubbles one tenth of the<br />

minimum channel width. We present a device capable of producing microbubbles as small as<br />

2.5 μm via a minimum channel diameter of 100 μm. Pressurised air surrounds an annular<br />

gas-in-liquid jet passing through a 100 μm orifice. The jet diameter, <strong>and</strong> hence the resulting<br />

microbubble diameter, can be controlled simply by varying the surrounding air pressure.<br />

Furthermore, the device operates in a novel high-Reynolds number regime, which would<br />

typically be expected to yield highly polydisperse results. However this device is able to<br />

maintain a relatively narrow size distribution with polydispersity index 16% at optimal<br />

performance. Operating regimes are described <strong>and</strong> size distribution data are presented.<br />

Studies have also been carried out to investigate the effect of varying core gas <strong>and</strong> shell<br />

composition on the effect of microbubble stability.<br />

Additional authors:<br />

Joe Fiabane*, Ritu Malik, Prem Nagappanpillai, Paul Prentice, Ketan Pancholi


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

16 th & 17 th July 2012<br />

Poster Number: 10<br />

Sevan Harput<br />

University of Leeds<br />

Title: Separating the Second Harmonic Response of Tissue <strong>and</strong> <strong>Microbubble</strong>s using Bispectral<br />

Analysis<br />

Abstract: The second harmonic generation in medical ultrasound is either caused by tissue or<br />

ultrasound contrast agents (UCAs). The conventional signal processing techniques cannot separate<br />

the harmonic response from microbubbles <strong>and</strong> tissue. The second order spectral analysis, commonly<br />

known as the frequency analysis, is the most common way of evaluating the microbubble response.<br />

Although frequency analysis can estimate the power spectrum effectively, it suppresses the phase<br />

relation between the frequency components. In this study, third order (bispectral) analysis is used to<br />

evaluate the microbubble response <strong>and</strong> separate the second harmonic generated by tissue <strong>and</strong><br />

microbubbles via the phase coupling between fundamental <strong>and</strong> harmonic components.<br />

To verify the validity of the proposed technique, an ultrasound phantom was created with tissue<br />

mimicking material (TMM) <strong>and</strong> a chamber for UCAs. A st<strong>and</strong>ard medical probe was used to scan the<br />

phantom using a 96-channel Ultrasound Array Research Platform (UARP). An LFM chirp sweeping<br />

between 3-4 MHz was used for excitation <strong>and</strong> the second harmonic was received by the same probe<br />

at 6-8 MHz. It was observed in the measurements that CTR can drop below 0 dB for low UCA<br />

concentrations as shown in the figure, where it is impossible to distinguish between TMM <strong>and</strong><br />

microbubbles. However, bispectral analysis showed phase coherence between fundamental <strong>and</strong><br />

second harmonic for TMM as high as 83%. No significant correlation between the second harmonic<br />

<strong>and</strong> fundamental components was observed for microbubbles, where the phase coherence value<br />

was less than 10%.<br />

Results show that higher-order spectral analysis allows the separation of the harmonics generated by<br />

microbubbles <strong>and</strong> tissue. Unlike other tissue harmonic suppression techniques based on multipleexcitations,<br />

bicoherence is not susceptible to motion artefacts since movement does not affect the<br />

phase relation between fundamental <strong>and</strong> harmonic components.<br />

Additional authors:<br />

Sevan Harput, James McLaughlan, Peter R. Smith,<br />

David M. J. Cowell, Stephen D. Evans a<br />

<strong>and</strong> Steven Freear<br />

Ultrasound Group, School of Electronic <strong>and</strong><br />

Electrical Engineering University of Leeds, Leeds, UK.;<br />

a School of Physics & Astronomy, University of Leeds,<br />

Leeds, UK.


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

16 th & 17 th July 2012<br />

Poster Number: 11<br />

Title: Actin Coated <strong>Microbubble</strong>s<br />

George Heath<br />

University of Leeds<br />

Abstract: The protein actin forms long semi-flexible polymers known as microfilaments<br />

found as a supporting cortex in all eukaryotic cells with the function of supporting the cell<br />

membrane <strong>and</strong> shape. We currently utilize this to create novel biomimetic microbubble<br />

coatings which will potentially allow subharmonic ultrasound imaging by creating nonlinear<br />

radial oscillations. We use cationic lipids to polymerise actin at the lipid-liquid interface<br />

creating a lipid microbubble with a monolayer coating of actin filaments. Actin-microbubbles<br />

were characterised with florescence microscopy, AFM force spectroscopy <strong>and</strong> bubble<br />

dissolution. These techniques show the actin shell increases bubble stiffness, resistance to<br />

gas permeation <strong>and</strong> elasticity when compared to lipid alone. We plan to further this coating<br />

by adding actin binding proteins with crosslinks, branches <strong>and</strong> capping proteins, in an aim to<br />

create a network with tension.


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

16 th & 17 th July 2012<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 using contrast-enhanced high-frequency ultrasound.<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 />

We used the VisualSonics Vevo 770 micro-US system to evaluate bolus injection<br />

characteristics to show functional vessels within the tumour <strong>and</strong> binding of MBs to tumour<br />

vasculature in 2D. In addition, we validated VEGFR2-targeted binding of Micromarker<br />

microbubbles <strong>and</strong> microfluidics-generated microbubbles in vitro under flowing conditions.<br />

Immunohistochemistry on SW480 xenografts showed that VEGFR2 was indeed present in<br />

tumour vessels, in line with human studies. There was a peak in CD31+ vessels that also<br />

expressed VEGFR2 at approximately 50mm3 in size. Expression then decreased as the<br />

tumours increased in volume. Both Micromarker <strong>and</strong> microfluidics microbubbles specifically<br />

bound VEGFR2+ve cells under flowing conditions. VEGFR2-targeted MB injection <strong>and</strong><br />

subsequent destruction of bound MBs showed a greater binding of microbubbles in smaller<br />

tumours.<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 microbubbles in pre-clinical models of CRC.


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

16 th & 17 th July 2012<br />

Poster Number: 13<br />

Fairuzeta Ja'afar<br />

Imperial College London<br />

Title: Binding Differentials of <strong>Microbubble</strong>s to Activated versus Non-Activated HUVECs<br />

Abstract: There is growing evidence to show that late-phase contrast-enhanced ultrasound<br />

imaging (LP-CEUSI) of the carotid using non-targeted commercial microbubbles (MBs)<br />

reflects biological features of inflammation.1 Whereas a number of potential initiators <strong>and</strong><br />

mechanism(s) have been proposed as suitable explanations for this non-targeted MB<br />

retention in the vasculature, much uncertainty remains about how these vascular cues<br />

influence MB adhesion. In this study, we evaluated the binding differentials of various<br />

commercial MBs (SonoVue, Optison <strong>and</strong> Definity/Luminity) <strong>and</strong> the experimental<br />

agent BR38, with models of inflamed (activated) <strong>and</strong> normal (non-activated) vasculature in<br />

vitro. We employed human umbilical vein endothelial cells (HUVECs) <strong>and</strong> observed MB<br />

binding using bright-field microscopy under physiological flow conditions. We then examined<br />

the role of MB surface charge on HUVECs/MBs binding affinities using a combination of laser<br />

Doppler velocimetry (LDV) <strong>and</strong> phase analysis light scattering (PALS) on a Zetasizer Nano Z<br />

(Malvern Instruments). We found a ~3-fold <strong>and</strong> ~1-fold increase in the number densities of<br />

Optison <strong>and</strong> SonoVue MBs adherent respectively, on activated versus non-activated<br />

HUVECs (p


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

16 th & 17 th July 2012<br />

Poster Number: 14<br />

Title: Mutually interacting targeted microbubbles<br />

Tom Kokhuis<br />

Erasmus MC<br />

Abstract: Ultrasound application can rupture the adhesion of targeted microbubbles (MBs) due to a mutual<br />

interaction between the bubbles called secondary Bjerknes force (SBF) 1,2 . At lower pressures, targeted MBs<br />

were observed to move up to several hundreds of nanometers towards each other during ultrasound<br />

application, but recoiled back again to their initial position within 100 �s after the ultrasound was turned off 2 .<br />

Interestingly, the MBs remained spherical throughout the experiments. In this study we investigated the origin<br />

of this elastic restoring force in more detail using simultaneous top <strong>and</strong> side view high-speed imaging.<br />

Homemade biotinylated MBs were prepared by sonication as described by Klibanov et al 3 <strong>and</strong> targeted to the<br />

topside of an avidin-coated polystyrene capillary. Isolated pairs of MBs were insonified with 20 cycles at 2.25<br />

MHz (P_ = 0 – 200 kPa). Oscillatory <strong>and</strong> translationary dynamics were imaged with two orthogonally placed<br />

lenses. Images were superimposed <strong>and</strong> relayed to the Br<strong>and</strong>aris128 camera 4 . A typical example of the effect of<br />

SBF on the position of targeted MBs (in top view) is shown in panel A. The attractive SBF caused each MB to<br />

move by about 600 nm towards the other MB. In top view the MBs did not exhibit significant deformation,<br />

confirming earlier observations. However, side view imaging revealed the attractive SBF causes a MB to<br />

elongate in the direction of the other MB, thereby tending towards a prolate shape. This effect is illustrated in<br />

panel B for bubble 2. The aspect ratio of the MB as seen from the top (red dots) is approximately constant <strong>and</strong><br />

near unity throughout the experiment. However, the aspect ratio from the side (blue dots) increases by about<br />

20% from 1.1 to 1.3. Panel C (top view) <strong>and</strong> panel D (side view) show the corresponding images of the MBs at t<br />

= 0 �s <strong>and</strong> t = 11.26 �s (i.e. last frame).<br />

These results show that the attractive force between pulsating targeted MBs is opposed by a restoring force<br />

due to bubble deformation, which is driving the recoil after the US is turned off. The recoil could be modeled<br />

with a simple mass-spring system, yielding a value for the spring stiffness k of the order 10 -3 N/m. Moreover,<br />

the translational dynamics of interacting MBs was predicted by a hydrodynamic point particle model, including<br />

a value of the spring stiffness k of the very same order as derived experimentally from the recoiling curves.<br />

Underst<strong>and</strong>ing the effect of ultrasound application on targeted MBs is crucial for further advances in the realm<br />

of molecular imaging.<br />

References:<br />

[1] Schmidt et al., J Control. Release, 131 (1), 2008<br />

[2] Kokhuis et al., Proc. IEEE Ultrason. Symp., Orl<strong>and</strong>o, 2011<br />

[3] Klibanov et al., Invest. Radiol., 39 (3), 2004<br />

[4] Chin et al., Rev Sci Instr, 74 (12), 2003<br />

Additional authors:<br />

T.J.A. Kokhuis 1,7 * , B.A. Naaijkens 2,7 , L.J.M. Juffermans 3,7 , O. Kamp 4,7 , M. Versluis 5,6 <strong>and</strong> N. de Jong 1,7<br />

1 Biomedical Engineering, Thorax Center, Erasmus MC, Rotterdam, the Netherl<strong>and</strong>s<br />

2 Department of Pathology, VU Medical Center, Amsterdam, the Netherl<strong>and</strong>s<br />

3 Department of Physiology, VU Medical Center, Amsterdam, the Netherl<strong>and</strong>s<br />

4 Department of Cardiology, VU University Medical Center, Amsterdam, the Netherl<strong>and</strong>s<br />

5 Physics of Fluids Group, University of Twente, Enschede, the Netherl<strong>and</strong>s <strong>and</strong> MIRA Institute of<br />

Biomedical Technology <strong>and</strong> Technical Medicine, University of Twente, Enschede, the Netherl<strong>and</strong>s<br />

7 Interuniversity Cardiology Institute of the Netherl<strong>and</strong>s, Utrecht, the Netherl<strong>and</strong>s


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

16 th & 17 th July 2012<br />

Poster Number: 15<br />

Title: Binding area of targeted microbubbles<br />

Klazina Kooiman<br />

Erasmus MC<br />

Abstract: For molecular imaging using ultrasound contrast agents, targeted microbubbles (tMB) are designed<br />

with specific lig<strong>and</strong>s linked to the coated shell 1 . Research is ongoing to determine the binding force of tMB <strong>and</strong><br />

to distinguish bound from unbound tMB using ultrasound 2,3 . For this, the actual surface of the tMB that binds<br />

to a pathology is important. This study focuses on determining the binding area of bound tMB by fluorescence<br />

microscopy.<br />

Lipid-coated MBs (3-7 µm in diameter; composition in mol%: DPPC or DSPC 59.1; PEG-40 stearate 35.7; DSPE-<br />

PEG(2000) 4.1; DSPE-PEG(2000)-biotin 0.8) with a C4F10 gas core were made by sonication 4 . Binding area of<br />

tMB was studied by adding the lipid dye DiD to the MB before sonication <strong>and</strong> coating an Opticell membrane<br />

with fluorescent streptavidin Oregon Green 488 (5 µg/ml). High-resolution images were recorded using 4Pi<br />

microscopy 5 . The overlap of the red <strong>and</strong> green fluorescence was calculated using Fiji software on basis of<br />

intensity threshold.<br />

The binding area of bound tMB was found to be 11 ± 4% of the total MB surface for tMB with DPPC as the main<br />

lipid (n=24) <strong>and</strong> 6 ± 4% for tMB with DSPC as the main lipid (n=22). Examples are given in the Figure. The<br />

average binding area was smaller for the DSPC tMB <strong>and</strong> can be explained by the heterogeneous distribution of<br />

the lig<strong>and</strong> throughout the MB coating 6 . These findings can be used to improve the binding of tMB <strong>and</strong> for the<br />

ongoing research to distinguish bound from unbound tMB.<br />

References: 1 Lindner et al, Nat Rev Cardiol 6:475, 2009; 2 Dayton et al, Mol Imaging 3:125, 2004; 3<br />

Zhao et al J Acoust Soc Am 120:EL63, 2006; 4 Klibanov et al, Invest Radiol 39:187, 2004; 5 Hell et al, J<br />

Microsc-Oxf 187:1, 1997; 6 Kooiman et al, IEEE Proceedings 2010.<br />

Klazina Kooiman *a , Tom J.A. Kokhuis a,b , Ilya Skachkov a , Johan G. Bosch a , Antonius F.W. van der Steen<br />

a,b , Wiggert A. van Cappellen c , Nico de Jong a,b<br />

Additional authors:<br />

a Dept. of Biomedical Engineering, Thoraxcenter, Erasmus MC, Rotterdam, the Netherl<strong>and</strong>s<br />

b Interuniversity Cardiology Institute of the Netherl<strong>and</strong>s, Utrecht, the Netherl<strong>and</strong>s<br />

c Dept. of Reproduction <strong>and</strong> Development, Erasmus MC, Rotterdam, the Netherl<strong>and</strong>s<br />

Figure. Targeted microbubbles (red fluorescent) bound to a streptavidin-coated surface<br />

(green fluorescent) recorded by 4Pi microscopy. A) microbubble with DPPC as main<br />

lipid (4.7 µm in diameter); B) microbubble with DSPC as main lipid (4.5 µm in<br />

diameter).<br />

This research is supported by the<br />

Center for Translational Molecular<br />

Medicine <strong>and</strong> the Dutch Heart<br />

Foundation (PARISk).


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

16 th & 17 th July 2012<br />

Poster Number: 16<br />

Gael Leaute<br />

University of Leeds<br />

Title: Finite element modelling of non-spherical microbubble dynamics in the vicinity of a<br />

soft membrane<br />

Abstract: A combination of microbubbles <strong>and</strong> ultrasonic insonation can increase the<br />

permeability of a cell membrane to increase uptake of a therapeutic drug. Previous studies<br />

have shown that non-spherical deformation can occur near a boundary, often forming jets<br />

that may create localised cell membrane damage.1 In this study, a visco-elastic finite<br />

element model for simulating the response of a phospholipid encapsulated microbubble to<br />

ultrasound in the vicinity of a soft membrane was implemented. The model was used to<br />

study the non-spherical deformations at high insonation pressures <strong>and</strong> investigate the<br />

factors which affect the likelihood of jetting.<br />

Transient response of a microbubble was simulated with Comsol Multiphysics(TM) Finite<br />

Element Analysis software. <strong>Microbubble</strong>s with an initial radii of R0 = 1-5 µm <strong>and</strong> a 2 nm thick<br />

lipid coating, situated from 100nm to R0 a soft membrane were considered. The ratio ξ=d/R0<br />

<strong>and</strong> the amplitude of the ultrasound field were considered to study the likelihood of jetting,<br />

where d is the shortest distance between the cell membrane <strong>and</strong> the microbubble wall.<br />

The distance d at which jetting occurs was found dependent on the microbubble radius <strong>and</strong><br />

the peak negative pressure. For a pressure of 500 kPa <strong>and</strong> ξ ≤ 0.4 jetting was present <strong>and</strong><br />

increased in magnitude as d was reduced. However, for ξ > 0.4 non-spherical shell<br />

deformations were present, but no jetting was observed. For ξ < 0.08 a minimum Pa of 200<br />

kPa was sufficient to establish jetting. The likelihood of jetting was found dependent on ξ<br />

<strong>and</strong> as d decreased the pressure required for jetting also decreased. This could suggest that<br />

targeted microbubbles bound to a cell are likely to result in jetting at lower pressures than<br />

unbound microbubbles.<br />

References:<br />

P. Prentice et al. Nature Physics, 1(2):107–110, 2005.<br />

Additional authors:<br />

Gaël Léauté*, David M. J. Cowell, James McLaughlan, Sevan Harput <strong>and</strong> Steven Freear


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

16 th & 17 th July 2012<br />

Poster Number: 17<br />

Title: Investigation into the Effects of Ultrasound on Attached <strong>Microbubble</strong>s<br />

Jonathan Loughran<br />

Imperial College London<br />

Abstract: <strong>Microbubble</strong> contrast agents have shown great potential in imaging <strong>and</strong><br />

quantifying blood flow <strong>and</strong> tissue perfusion. Recent advance of targeted microbubbles have<br />

made it possible to detect <strong>and</strong> imaging the distribution of specific molecules of interest<br />

expressed on endothelial walls in vivo using ultrasound. Previous studies have been focused<br />

on developing microbubbles with improved targeting efficiency, acoustic techniques to<br />

facilitate the attachment of such bubbles <strong>and</strong> better detection method, little attention has<br />

been paid to study what happens to the microbubbles once they are attached. In this study<br />

the aim is to study the effects of ultrasound exposure to attached microbubbles. Biotin<br />

coated microbubbles were flown through a phantom coated with streptavidin. Optical<br />

microscopy was used to observe the attached microbubbles in the flow phantom under<br />

ultrasound exposure. The bubble size <strong>and</strong> position were recorded during the exposure of<br />

ultrasound. It was found that even at relatively low acoustic pressure a significant amount of<br />

bubbles detach from the wall <strong>and</strong> flow away, some of them shrink at the same time. The<br />

number of bubble detachment increases with acoustic pressure. The different forces acting<br />

on the bubbles during the exposure was analysed to offer some insights into the detachment<br />

mechanisms.<br />

Additional authors:<br />

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

1 Department of Bioengineering, Imperial College London, London SW7 2AZ, UK<br />

2 Biomedical Engineering Department, King's College London, London. SE1 7EH, UK


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

16 th & 17 th July 2012<br />

Poster Number: 18<br />

Title: Imaging the development of functional tumour vasculature, <strong>and</strong> the effect of<br />

combretastatin A-4 on tumour blood flow rate in vivo<br />

Gemma Marston<br />

University of Leeds<br />

Abstract:<br />

Contrast enhanced high-frequency ultrasound (CE HFUS) enables longitudinal studies in<br />

mouse models <strong>and</strong> can be used to investigate both qualitative <strong>and</strong> quantitative changes in<br />

tumour vasculature, including tumour blood flow <strong>and</strong> perfusion. We have developed CE<br />

HFUS protocols with the VisualSonics Vevo770 in order to characterise the development of<br />

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

35 days of tumour growth, <strong>and</strong> have undertaken a pilot study to image <strong>and</strong> quantify the<br />

effect of combretastatin A-4 (CA4) on tumour blood flow kinetics.<br />

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

kinetics <strong>and</strong> vascular data. Tumour vascular space <strong>and</strong> tumour volume was calculated from<br />

3D CE HFUS images. This showed that the percentage tumour vascular space decreased<br />

significantly as the tumour volume increased. Furthermore, smaller tumours had faster rates<br />

of blood flow than larger tumours <strong>and</strong> smaller tumours had significantly higher maximum<br />

levels of vascular perfusion than larger tumours. Post treatment of CA-4 showed that the<br />

blood flow rate within tumours significantly decreased, with maximum intensity persistence<br />

showing a significant decrease in total blood flow through the tumour.<br />

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

development in vivo <strong>and</strong> provides a relatively non-invasive method to assess development of<br />

anti-angiogenic <strong>and</strong> vascular disruptive therapies, <strong>and</strong> response to treatment in pre-clinical<br />

trials.


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

16 th & 17 th July 2012<br />

Poster Number: 19<br />

Title: Cerato Ulmin a new UCA shell material?<br />

Jono McKendry<br />

University of Leeds<br />

Abstract: The hydrophobin cerato ulmin (CU) has been used as a stabilising coating for<br />

bubbles on the micrometer length scale. CU coated bubbles are shown to display greater<br />

stability than commonly used contrast agent coating lipids, with sample lifetimes greater<br />

than 10 days. Using AFM force spectroscopy the mechanical properties of the cerato ulmin<br />

coated bubbles have been investigated. Using tipless AFM cantilevers to compress the<br />

bubbles their stiffness has been quantified (254 mN/m for a bubble of radius ~ 9μm) <strong>and</strong><br />

dynamic visco-elastic measurements have been performed. Furthermore, methods of<br />

production of spherical <strong>and</strong> linear bubbles are described with discussion of the potential of<br />

non-spherical microbubbles as ultrasound contrast agents.<br />

Additional authors:<br />

Dr J. E. McKendry 1 , Dr C. Grant 2 , Prof P. S. Russo 3 , Prof S. D. Evans 1<br />

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

2. Polymer IRC, University of Bradford, Bradford, BD7 1AL<br />

3. Department of Chemistry & Macromolecular Studies Group, Louisiana State University,<br />

Baton Rouge, Louisiana 70803-1804


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

16 th & 17 th July 2012<br />

Poster Number: 20<br />

Title: High production rates of therapeutic microbubbles for targeted drug delivery.<br />

Sally Peyman<br />

University of Leeds<br />

Abstract: Micron sized, lipid stabilized bubbles of gas are of interest as contrast agents for ultrasound<br />

(US) imaging <strong>and</strong> increasingly as vehicles for targeted drug delivery. Microfluidics provides a<br />

reproducible means for microbubble production <strong>and</strong> surface functionalisation using flow-focussing<br />

microfluidic devices that combine streams of gas <strong>and</strong> liquid through a nozzle a few microns wide <strong>and</strong><br />

then subjecting the two phases to a downstream pressure drop. While microfluidics has successfully<br />

demonstrated the generation of monodisperse bubble populations, these approaches inherently<br />

produce low bubble counts. We introduce a new micro-spray flow regime that generates consistently<br />

high bubble concentrations, in the order of 10 10 bubbles mL -1 , that are more clinically relevant<br />

compared to traditional monodisperse bubble populations. The technique is shown to be easy to<br />

implement <strong>and</strong> highly reproducible. By using multiplexed chip arrays containing up to 4 bubble<br />

devices, the time taken to produce one millilitre of sample was ~10 minutes. Further, we also<br />

demonstrate that it is possible to attach liposomes, loaded with quantum dots or fluorescein <strong>and</strong><br />

form microbubbles containing an internal layer of oil for the delivery of hydrophobic drugs, in a<br />

single step during MBs formation.<br />

Additional authors:<br />

Sally A. Peyman, Radwa Abou-Saleh, Stephen D. Evans.<br />

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


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

16 th & 17 th July 2012<br />

Poster Number: 21<br />

Linsey Phillips<br />

University of North Carolina<br />

Title: New Nanodroplets Increases Tissue Ablation in Response to High Intensity Focused<br />

Ultrasound<br />

Abstract: Introduction: We are investigating a new low-boiling point perfluorocarbon emulsion<br />

composed of decafluorobutane (b.p.=2°C) <strong>and</strong> dodecafluoropentane (b.p= 30°C) for enhanced<br />

thermal delivery during high intensity focused ultrasound (HIFU) ablation. <strong>Microbubble</strong>s (MBs) are<br />

known to enhance thermal delivery, however they can shield energy deposition to deeper tissues,<br />

<strong>and</strong> are too large to extravasate. Our nanodroplets (NDs) are 240 +/- 65 nm in diameter, small<br />

enough to extravasate into tumor tissue, <strong>and</strong> are inherently more sensitive to ultrasound due to their<br />

low boiling point. We hypothesized that these NDs would increase HIFU ablation lesions in tissuemimicking<br />

phantoms compared to agent-free controls. Materials <strong>and</strong> Methods: A range of<br />

concentrations of MBs or NDs were added to individual albumin-acrylamide tissue-mimicking<br />

phantoms. A focused 1 MHz array applied continuous wave HIFU at 4 MPa for 20 seconds to<br />

phantoms maintained at 37°C. Temperature was monitored at the acoustic focus within the<br />

phantoms using a needle-type thermocouple probe. Results <strong>and</strong> Discussion: The addition of NDs or<br />

MBs significantly increased the size of lesions in the phantoms in response to HIFU. Lesion size was<br />

highly dose dependent (see Fig. 1A). The temperature rose faster in phantoms containing NDs<br />

compared to those containing MBs or no agents. Lesion formation occurred at the surface of<br />

phantoms containing MBs, wherein these MBs shielded deep lesion formation (Fig. 1B). ND lesions<br />

formed around the acoustic focus (Fig. 1B). Conclusion: This study demonstrates that our new<br />

nanodroplets containing a low boiling point PFC enhance HIFU ablation. Furthermore these agents<br />

induce targeted lesion formation at the site of interest while avoiding non-targeted heating.<br />

Additional authors: Linsey C. Phillips*, Connor Puett, Paul S. Sheeran, Paul A. Dayton<br />

Department of Biomedical Engineering, University of North Carolina at Chapel Hill, USA<br />

Figure 1 A) HIFU induced lesion volumes (mean ± S.D., n ≥ 3) in albumin-acrylamide phantoms containing<br />

microbubbles (MB) or nanodroplets (ND) as a function of concentration <strong>and</strong> B) corresponding images of each type of<br />

lesion.


<strong>Microbubble</strong> <strong>Symposium</strong>: <strong>Fabrication</strong>, <strong>Characterisation</strong> <strong>and</strong> Translational Applications 45<br />

16 th & 17 th July 2012<br />

Poster Number: 22<br />

Title: Single-well acoustic tweezers using ultrasonic travelling waves.<br />

Ben Raiton<br />

University of Leeds<br />

Abstract:<br />

SAW devices can be used in bioscience applications to manipulate micrometre-scale particles.<br />

“Wells” are expected to form every half wavelength within a 1D pattern <strong>and</strong> √2/2 times the<br />

wavelength within a 2D pattern. A well of 10s of μm can be obtained by creating a st<strong>and</strong>ing wave<br />

with a frequency of a few MHz. However, this also means that a high number of wells will form<br />

within a working region of a few millimetres. Two apertures emitting acoustic travelling waves with<br />

opposite phase results in the generation of a pressure null at the focal depth. This way, a single well<br />

can be obtained over a few millimetres. As is the case for the st<strong>and</strong>ing waves, increasing the pressure<br />

level will apply a stronger radiation force on the trapped particles <strong>and</strong> form a narrower well. Then, by<br />

translating the array or even steering the beams the trapped particles can be manipulated.<br />

Lipid shelled microbubbles with a size distribution of 1-10 μm are flown through a 200 μm cellulose<br />

tube at a rate of 7ml/hr. A linear array is set to emit 5 MHz 50 cycle tone bursts at a repetition<br />

frequency of 30 kHz. The whole array aperture is focused, at 30 mm, onto the tube. Although each<br />

element emits the same tone burst, both halves of the aperture are out of phase by 180°. Once<br />

sufficient microbubbles have accumulated the flow is stopped. The array is then translated along the<br />

tube axis to displace the trapped cluster across the microscope viewing window which has a width of<br />

1.2 mm.<br />

A concentration of 100x10 6 microbubbles /ml were flown until enough microbubbles were<br />

accumulated to form a cluster of 50 μm in diameter. Whilst the tube remained insonified, the flow<br />

was stopped. The trapped cluster was then translated within the viewing window at up to 1mm/s.<br />

After over 60 sec of manipulation there was no visible change in the cluster diameter. A single<br />

microbubble aggregation was successfully formed over the entire viewing window. The cluster could<br />

then be translated to any desired location along the tube. The impact on the microbubble lifetime<br />

was limited as they were constantly held at the pressure null. Using a 1D setup, a cluster could be<br />

translated over 10s of mm as long as it is restricted to a narrow channel. The same technique could<br />

be extended to a 2D setup so that a cluster could be freely translated across a 2D plane such as a cell<br />

layer.


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

16 th & 17 th July 2012<br />

Poster Number: 23<br />

Title: Hardware accelerated beamforming for microbubble detection<br />

Sara Shakil Qureshi<br />

University of Sciences <strong>and</strong> Technology, Pakistan<br />

Abstract: A flexible, real-time digital beamformer implementation is described for<br />

microbubble detection. The architecture enables acquisition of both beamformed <strong>and</strong> prebeamformed<br />

data of up to 96 channels from a single transmit event, <strong>and</strong> is suitable for a<br />

range of imaging methods such as linear array imaging, phased array imaging <strong>and</strong> synthetic<br />

aperture imaging.<br />

The Ultrasound Array Research Platform (UARP) receives ultrasound data from 96 parallel<br />

channels to an Altera Stratix III FPGA after 12 bit Analogue to Digital conversion at 50 MHz.<br />

Samples are adjusted to remove any DC Offset error caused by the Analogue Front End.<br />

Samples are then delayed <strong>and</strong> weighted according to delay profiles <strong>and</strong> apodization<br />

coefficients respectively. Each adjusted, delayed <strong>and</strong> weighted output stream in the<br />

aperture is then added in a pipelined adder. A clipping circuit ensures that beamformed data<br />

outside of a 16 bit output range is captured <strong>and</strong> does not appear as false data. All<br />

parameters including apodization coefficients, aperture size <strong>and</strong> output range select are<br />

controllable at run-time. The beamformed output is saved into on-chip memory of 4096<br />

samples depth, alongside the corresponding pre-beamformed data. The user has the option<br />

of transferring either or both forms of data to the host PC, thus offering greatest access to<br />

the received ultrasound data. The architecture also permits fast acquisition of microbubble<br />

sequences by storing multiple beamformed scanlines in memory.<br />

Additional authors:<br />

Sara S. Qureshi 12 *, Peter R. Smith 1 , Dr David M. J. Cowell 1 , Dr Kashif M. Rajpoot 2 , <strong>and</strong> Dr Steven<br />

Freear 1<br />

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

Kingdom. 2 School of Electrical Engineering <strong>and</strong> Computer Science, National University of Sciences <strong>and</strong><br />

Technology, Pakistan.<br />

Received<br />

Echo<br />

Data<br />

Parameter<br />

Loading<br />

Block<br />

Analog<br />

Front<br />

End<br />

DC<br />

Offset<br />

Adjust<br />

Block<br />

Delay Loading<br />

Apodization<br />

Loading<br />

Fine<br />

Delay<br />

Block<br />

Coarse<br />

Delay<br />

Block<br />

Software Interface (MATLAB)<br />

Beamformed Data Path<br />

Pre-Beamformed Data Path<br />

USB Link<br />

NIOS PROCESSOR<br />

Apodization<br />

Block<br />

Summing<br />

Block<br />

Field Programmable Gate Array (FPGA) – Stratix III<br />

Ultrasound Array Research Platform (UARP)<br />

Range<br />

Selection<br />

Block<br />

Data<br />

Read<br />

Block<br />

Pre-Beamformed<br />

Data<br />

AND<br />

Beamformed Data<br />

Memory


<strong>Microbubble</strong> <strong>Symposium</strong>: <strong>Fabrication</strong>, <strong>Characterisation</strong> <strong>and</strong> Translational Applications 47<br />

16 th & 17 th July 2012<br />

Poster Number: 24<br />

Peter Smith<br />

University of Leeds<br />

Title: Composite High Frequency, Low Frequency System for Pre-Clinical <strong>Microbubble</strong><br />

Sonoporation<br />

Abstract: Ultrasound contrast agents or microbubbles are primarily used clinically to<br />

increase vasculature delineation or perform cardiac perfusion measurements. Research has<br />

shown a potential secondary function of microbubbles as a drug or gene delivery method for<br />

therapeutic applications. This secondary function relies on the microbubble oscillating under<br />

an applied ultrasound field. Oscillations of microbubbles in close proximity to cell<br />

membranes cause them to rupture temporarily - a technique known as ‘Sonoporation’. This<br />

technique has shown potential in enabling more targeted therapy by delivering genes or<br />

drug loaded microbubbles to tumour sites.<br />

Pre-clinical in vivo studies are the first step in evaluating the efficacy of microbubble<br />

drug/gene delivery. The use of small animal models for these studies has a number of<br />

advantages, but requires high frequency ultrasound systems to achieve satisfactory image<br />

quality. High frequency ultrasound may provide high resolution images, but is inefficient for<br />

driving microbubble oscillations. Current trends therefore seek to combine high frequency<br />

imaging with low frequency excitation of microbubbles. This provides best trade-off<br />

between image quality <strong>and</strong> Sonoporation efficiency.<br />

This work reports on progress of a two part study, investigating: 1) the optimum low<br />

frequency ultrasound parameters for microbubble destruction, <strong>and</strong> 2) using these optimised<br />

parameters in In vivo mouse trials with liposome-loaded microbubbles. Results are obtained<br />

using a high frequency (40 MHz) pre-clinical imaging system (VisualSonics Vevo 770) in<br />

conjunction with a custom single channel, low frequency (0.1 – 10 MHz) system.<br />

Additional authors:<br />

Peter R. Smith* 1 , Dr James R. McLaughlan 1 , Dr David M. J. Cowell 1 , Dr Nicola Ingram 2 <strong>and</strong> Dr. Louise<br />

Coletta 2<br />

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

Kingdom. 2 Leeds Institute of Molecular Medicine, Wellcome Trust Brenner Building, St James’s<br />

University Hospital, Leeds, United Kingdom.


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

16 th & 17 th July 2012<br />

Poster Number: 25<br />

Title: Development of an Ultrasound Array Research Platform (UARP)<br />

Peter Smith<br />

University of Leeds<br />

Abstract: Commercial ultrasound systems enable a medical professional to perform quick<br />

<strong>and</strong> accurate diagnosis of medical conditions, assist in surgical procedures, or apply<br />

treatment. These systems provide a user with the optimal apparatus to perform specific,<br />

well-defined tasks in a clinical setting, but are often highly constrained or inflexible.<br />

In a research environment, non-conventional operation of components in the ultrasound<br />

system is often required. Techniques such as pulse inversion, power modulation <strong>and</strong><br />

harmonic imaging have previously demonstrated non-conventional transmitter sequencing,<br />

signal acquisition <strong>and</strong> receive processing. A lack of system control may therefore be a<br />

significant barrier to research when using a clinical system. Moreover, implementation of a<br />

new research method may require software development <strong>and</strong>/or hardware modifications,<br />

where information or support from the manufacturer is required. Furthermore, the<br />

information needed to evaluate the new research method may not be available from the<br />

commercial system, or available in the correct format. For example, the output of<br />

commercial systems is often a set of high-definition processed images showing relative pixel<br />

intensity which is often an unsuitable output for researchers. Access to pre-beamformed RF<br />

data lets you analyze the received signal both qualitatively <strong>and</strong> quantitatively, which<br />

improves the diagnostic ability of the system <strong>and</strong> permits further study of results.<br />

In the research environment a custom flexible platform provides the user with full system<br />

control <strong>and</strong> offers distinct advantages over a commercial clinical system. This work reports<br />

on the continual development of a custom Ultrasound Array Research Platform (UARP),<br />

developed to offer the flexibility required for a research context. An overview of the<br />

platform is provided, with examples of non-conventional operation given, particularly with<br />

respect to ultrasound-microbubble interactions. Future developments <strong>and</strong> applications are<br />

also presented.<br />

Additional authors:<br />

Peter R. Smith*, Dr David M. J. Cowell, Dr James R. McLaughlan, Sevan Harput, Benjamin Raiton,<br />

Olaoluwa Olagunju, Sara Shakil Qureshi, Charlie Winckler <strong>and</strong> Dr Steven Freear<br />

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

Kingdom


<strong>Microbubble</strong> <strong>Symposium</strong>: <strong>Fabrication</strong>, <strong>Characterisation</strong> <strong>and</strong> Translational Applications 49<br />

16 th & 17 th July 2012<br />

Delegate List<br />

Name Email Affiliation<br />

Mostafa Abdelrahman mtp5ma@leeds.ac.uk University of Leeds<br />

Radwa Abou-Saleh R.H.Saleh@leeds.ac.uk University of Leeds<br />

Mohamed Ashawesh ashawish77@yahoo.co.uk University of Sheffield<br />

Jennifer Bain pmjb@leeds.ac.uk University of Leeds<br />

Jonathan Blockley pyjb@leeds.ac.uk University of Leeds<br />

Ayache Bouakaz ayache.bouakaz@univ-tours.fr Université François Rabelais,<br />

Tours<br />

Richard Bushby R.J.Bushby@leeds.ac.uk University of Leeds<br />

Jonathan Casey jonathan.casey08@imperial.ac.uk Imperial College London<br />

Robin Clevel<strong>and</strong> robin.clevel<strong>and</strong>@eng.ox.ac.uk University of Oxford<br />

Louise Coletta P.L.Coletta@leeds.ac.uk University of Leeds<br />

Michael Conneely m.j.conneely@dundee.ac.uk University of Dundee<br />

Kevin Critchley K.Critchley@leeds.ac.uk University of Leeds<br />

Paul Dayton padayton@bme.unc.edu University of North Carolina<br />

Nico de Jong n.dejong@erasmusmc.nl Erasmus MC<br />

Pratik Desai pratikdr@gmail.com University of Sheffield<br />

Mohan Edirisinghe m.edirisinghe@ucl.ac.uk University College London<br />

Stephen Evans S.D.Evans@leeds.ac.uk University of Leeds<br />

Tony Evans J.A.Evans@leeds.ac.uk University of Leeds<br />

Joe Fiabane f.j.fiabane@rgu.ac.uk Robert Gordon University,<br />

Aberdeen<br />

Steven Freear S.Freear@leeds.ac.uk University of Leeds<br />

Chris Fury chris.fury@npl.co.uk National Physical Laboratory<br />

Sevan Harput eensha@leeds.ac.uk University of Leeds<br />

George Heath py06gh@leeds.ac.uk University of Leeds<br />

John Hossack hossack@virginia.edu University of Virginia<br />

Nicola Ingram n.ingram@leeds.ac.uk University of Leeds<br />

Fairuzeta Ja'afar fairuzeta.jaafar06@imperial.ac.uk Imperial College London<br />

Benjamin Johnson b.r.g.johnson@leeds.ac.uk University of Leeds<br />

Pam Jones P.Jones@leeds.ac.uk University of Leeds<br />

Jithin Jose jjose@visualsonics.com Visualsonics<br />

Tom Kokhuis t.kokhuis@erasmusmc.nl Erasmus MC<br />

Klazina Kooiman k.kooiman@erasmusmc.nl Erasmus MC


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

16 th & 17 th July 2012<br />

Vasileios Koutsos vasileios.koutsos@ed.ac.uk University of Edinburgh<br />

Dmitriy Kuvshinov d.kuvshinov@sheffield.ac.uk University of Sheffield<br />

Gael Leaute elgyvl@leeds.ac.uk University of Leeds<br />

Marjorie Longo mllongo@ucdavis.edu UC Davis<br />

Jonathan Loughran j.loughran09@imperial.ac.uk Imperial College London<br />

Alex Markham A.F.Markham@leeds.ac.uk University of Leeds<br />

Gemma Marston G.Marston@leeds.ac.uk University of Leeds<br />

Jono McKendry jonomckendry@gmail.com University of Leeds<br />

James McLaughlan J.R.McLaughlan@leeds.ac.uk University of Leeds<br />

Carmel Moran Carmel.Moran@ed.ac.uk University of Edinburgh<br />

Chris Newman c.newman@sheffield.ac.uk University of Sheffield<br />

Olaoluwa Olagunju eloo@leeds.ac.uk University of Leeds<br />

Sally Peyman S.Peyman@leeds.ac.uk University of Leeds<br />

Linsey Phillips linsey@live.unc.edu University of North Carolina<br />

Ben Raiton elbdr@leeds.ac.uk University of Leeds<br />

Tim Ryan Tim.Ryan@epigem.co.uk Epigem Limited<br />

Sara Shakil Qureshi sara.shakil@seecs.edu.pk University of Sciences <strong>and</strong><br />

Technology, Pakistan<br />

Jane Smith Janes.Bates@leedsth.nhs.uk St James's University Hospital,<br />

Leeds<br />

Peter Smith efy3prs@leeds.ac.uk University of Leeds<br />

Mengxing Tang mengxing.tang@imperial.ac.uk Imperial College London<br />

Neil Thomson N.H.Thomson@leeds.ac.uk University of Leeds<br />

Michel Versluis m.versluis@utwente.nl University of Twente<br />

Charles Winckler University of Leeds

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

Saved successfully!

Ooh no, something went wrong!