Вы находитесь на странице: 1из 12

European Journal of Radiology 70 (2009) 242–253

Contents lists available at ScienceDirect

European Journal of Radiology


journal homepage: www.elsevier.com/locate/ejrad

Ultrasound triggered image-guided drug delivery


Marcel R. Böhmer a,c,∗ , Alexander L. Klibanov b , Klaus Tiemann c , Christopher S. Hall d ,
Holger Gruell a , Oliver C. Steinbach a
a
Philips Research Europe, Biomolecular Engineering, HTC11, 5656 AE Eindhoven, The Netherlands
b
Cardiovascular Division, Department of Medicine, Cobb Hall, University of Virginia School of Medicine, Hospital Drive, Cobb Hall RM 1026, Charlottesville, VA 22908-158, USA
c
Department of Cardiology and Angiology, University Hospital Münster, Albert Schweitzerstrasse 33, 48149 Münster, Germany
d
Philips Research North America, Ultrasound Imaging and Therapy, 345 Scarborough Road, Briarcliff Manor, NY 10510, USA

a r t i c l e i n f o a b s t r a c t

Article history: The integration of therapeutic interventions with diagnostic imaging has been recognized as one of
Received 13 January 2009 the next technological developments that will have a major impact on medical treatments. Important
Accepted 14 January 2009 advances in this field are based on a combination of progress in guiding and monitoring ultrasound
energy, novel drug classes becoming available, the development of smart delivery vehicles, and more in
Keywords: depth understanding of the mechanisms of the cellular and molecular basis of diseases. Recent research
Ultrasound
demonstrates that both pressure sensitive and temperature sensitive delivery systems hold promise for
Image guidance
local treatment. The use of ultrasound for the delivery of drugs has been demonstrated in particular
MRI
Microbubbles
the field of cardiology and oncology for a variety of therapeutics ranging from small drug molecules to
Temperature sensitive liposome biologics and nucleic acids.
© 2008 Elsevier Ireland Ltd. All rights reserved.

1. Introduction can also be used non-destructively for increasing the efficacy for
delivery of drugs and genetic material. Especially for chemothera-
The role of medical imaging technologies in medical care is shift- peutic regimens to be successful in cancer treatment, the particular
ing from a tool for diagnosis of a disease to being an integral part drug must be effective in the tumor environment and administered
of therapeutic interventions such as in image-guided treatments. in doses that cause tumor eradication while keeping severe side
Stereotactic systems use images obtained before surgery, e.g., MR effects within acceptable limits, commonly called the therapeutic
and CT, for accurate guidance of a surgical tool to the target anatomy. window.
Instead of tissue removal, one can use high intensity focused ultra- Performing minimally invasive therapy, such as ultrasound
sound (HIFU) as a surgery tool. Using HIFU, energy can be focused mediated drug delivery (USDD), under image guidance requires
precisely to a small volume of interest. HIFU allows ablation of adequate definition of the region of interest and accurate compen-
tissue by local administration of thermal dosages. Image-guided sation for motion. Especially in the heart the feedback provided is
therapy offers the potential to direct therapeutic action precisely to necessary to target the therapy accurately. The region of interest can
the point in the tissue where it is needed and not to other tissues. be identified by detection of an abnormal morphology. Molecular
When this is possible, a high and local thermal dose can be admin- imaging holds promise to apply minimally invasive therapy in an
istered. Image-guided delivery using HIFU requires the integration early stage of a disease as malignancies can be detected in an early
of imaging for diagnosis and treatment planning and a therapy that stage. Molecular imaging uses targeted contrast agents, which are
can be accurately directed and controlled by simultaneous image agents decorated with, for instance, antibodies or fragments thereof
guidance, resulting in less side effects. that specifically interact with specific markers such as endothelial
The use of ultrasound for local hyperthermia was recognized markers of inflammation or angiogenesis.
early as reviewed by Moyer and Clement [1,2]. Direct exposure New methods in ultrasound and magnetic resonance (MR)
to therapeutic ultrasound produces irreversible cell death through provide higher resolution information in two and three spatial
coagulative necrosis, and is currently being clinically evaluated in dimensions, with acquisition and display occurring nearly in real
breast, kidney, and liver tumors [3]. There is an increasing level of time. Computer image processing methods offer ways of clarifying,
literature evidence [3–9] that demonstrates how ultrasound energy highlighting, or detecting specific regions in tissue. Developments
in MR thermometry provide a technical solution to follow the deliv-
ery of a thermal dose to a lesion. For treatment, a volume of interest
∗ Corresponding author. inside a patient is delineated based on MR imaging, and subse-
E-mail address: marcel.bohmer@philips.com (M.R. Böhmer). quently heated by focused ultrasound. The tissue temperature is

0720-048X/$ – see front matter © 2008 Elsevier Ireland Ltd. All rights reserved.
doi:10.1016/j.ejrad.2009.01.051
M.R. Böhmer et al. / European Journal of Radiology 70 (2009) 242–253 243

mapped by MR thermometry and fed back into the control of the For near surface heating, one can use higher frequencies (between
ultrasound transducer to provide full temporal and spatial control 3 and 10 MHz, typically) whereas for deeper seated structures one
of the heating [10–12]. Therefore, the technique gives more than a may have to use frequencies below 3 MHz as higher frequencies are
feedback on the location of the region of interest, it also provides attenuated more than lower frequencies.
information on a physical parameter, which can be used to guide
and control the therapy [13,14]. MRI can also be used to monitor 2.2. Triggering
changes in the permeability of the vasculature, as shown by Treat
et al. [15] The choice of when to fire the acoustic activation pulse for tar-
Contrast ultrasound imaging, using microbubbles, also pro- geted drug release is not always a trivial decision. In the case of
vides useful information for image-guided drug delivery. For these thermal release, the triggering choice may be governed by the abil-
agents, optimized detection algorithms are available on ultrasound ity to keep the target volume within the field of view. In particular,
diagnostic imaging systems. With respect to therapy transducer breathing, heartbeat, or inadvertent patient motion may occur over
design, developments in electronic steering of the beam improve the relatively long exposure times needed to reach a release temper-
the size of focal region and reduce grating lobes while maintaining ature. In order to combat these effects, the acoustic excitation can
a small number of elements and a compact size. be gated for respiration or the portion of the heart cycle. In heat-
In this review we focus on the use of ultrasound for therapy activated applications, the thermal energy needs to be deposited
and provide examples in cardiology and oncology. We will review quickly enough and over a longer period of time to fight the effects
drug delivery vehicles based on temperature and pressure sensitive of thermal diffusion and convection associated with blood flow. As
systems. Such systems are either modified slow release systems a result, the triggering can be used in exhalation and end-diastole
with a temperature sensitive component or contrast agents that where needed.
have been modified to include or enhance drug delivery. Develop- In cases of pressure-activated particles, the requirements are
ments in ultrasound and MRI imaging, and new agents to follow not as stringent for motion during the exposure but rather
and quantify drug release, will be described. between repeated exposures. The exposure typically lasts for tens
of microseconds and as a result, there is not much motion of the
insonified volume. However, for multiple exposures, care needs to
2. Ultrasound mediated drug delivery systems—equipment
be taken to expose the same volume of tissue. This is accomplished
design considerations
by triggering on respiration and heartbeat. Another important fac-
tor for pressure-release particles is that the triggering rate be set so
The equipment for ultrasound mediated delivery varies widely
that within the destroyed/released volume, ample time is allowed
dependent on the application and often on the clinical availabil-
for reperfusion. The time and therefore the trigger rate can be set
ity of ultrasound imaging or therapy devices. The following section
according to perfusion rate monitored in real time with imaging
will describe the components parts of ultrasound therapy devices
or with a priori knowledge of the flow rate. In myocardial delivery
for drug delivery with a discussion on the relevant importance and
applications, triggered ultrasound has been shown to be superior
design limitations. We will then follow with a discussion of spe-
to continuous ultrasound [16,17], presumably because the latter
cific application requirements that depend on the target volume
destroys most of the microbubbles within the cardiac chambers.
within the subject. As diagnostic imaging systems are not designed
Continuous ultrasound destroys microbubbles within the LV cav-
for therapy, which is in particular reflected in the focusing of the
ity and myocardial arterioles before they ever reach the capillary
ultrasound beam, we will not consider these studies here.
bed. In addition, because myocardial capillary blood flow velocity is
slow (1 mm/s) [18], the capillary bed can never completely fill with
2.1. Signal excitation microbubbles during continuous ultrasound at destructive acoustic
pressure.
The two primary physical mechanisms for activating particle
based therapies can be separated into heat- and pressure-activated 2.3. Transducer
particles. In the case of heat-activated particles, typically the
goal is to deposit enough acoustic energy into the targeted vol- The choice of transducer is driven primarily by the applica-
ume to raise the temperature in order to release an encapsulated tion. All transducers are made of piezo-electric material including
drug. In these cases, the duty cycle of the acoustic signal is of lead zirconate titanate (PZT) and composites with piezo-electrics
paramount importance and so the electrical excitation and ampli- embedded in a backing material. The size of the transducer is gov-
fication will consist of pure tone (long pulse lengths) signal sources erned in part by the maximal acoustic pressure and power needed.
and high power amplifiers (class D op-amps are popular). In the The choice of ceramic or composite is often made according to ease
case of pressure-mediated release, the acoustic signal is often of of handling but also by the efficiency of the electrical to acoustical
shorter duration and lower overall energy deposition than that energy conversion.
used in heat-release applications. Pressure based release uses par- In addition to the electrical and acoustic requirements for the
ticles, mostly microbubbles, which deposit or release a drug when transducer, the size and shape are controlled by the application. The
encountering a peak negative pressure beyond a particular thresh- ability to couple acoustic energy into the body to the target organ
old (usually) between 0.5 and 5 MPa peak negative pressure. These is controlled by the acoustic window into the body. The acoustic
signals are quite short in duration but high in acoustic pressure. A window refers to the acoustic opening in tissue overlying the region
typical approach is to transmit a short (<10 cycles) windowed pulse of interest that allows for acoustic waves to propagate to the treated
through a voltage amplifier. volume. The choice of shape and size is therefore governed by the
The choice of frequency for the excitation pulse is driven by two near field (entry into the body) to avoid such structures as bone and
factors: the first is the optimal interaction of drug-bearing particle air-filled cavities.
and the second is the propagation path to the volume of inter- Another major influence in the design and research phase of an
est. For example, for pressure-mediated drug delivery, a choice of ultrasound therapy system is the requirement of treating small vs.
frequency close to the resonance of the microbubble might be desir- more human-sized subjects. Besides the considerations mentioned
able in many cases. In the case of heat-mediated approaches, the later in this paper about circulation time and clearance organs, the
absorption of tissue of acoustic energy increases with frequency. depth of the target volume below the surface of the animal, the size
244 M.R. Böhmer et al. / European Journal of Radiology 70 (2009) 242–253

of the organs, and amount of peripheral blood flow for heat con-
vection among other factors are not simple linear scale factors. For
example, the depth of the treatment zone will determine the choice
of ultrasonic frequency which can lead to inadvertent heating in the
overlying tissue in the case of heat-activated particles, which may
also cause release in areas not intended. Also, the choice of a lower
frequency to penetrate to deeper tissue can negatively impact the
heat deposition as the lower frequencies are not absorbed at the
same rate as higher frequencies.

2.4. Steering

In many applications for ultrasound mediated drug delivery, it is


advantageous to be able to treat a large volume of tissue with great
spatial resolution. Many approaches have been employed with two
major divisions: spatial movement of the transducer and electronic
steering of the focus of the transducer. The first approach is simply
performed by placing a single element transducer (or low element
count) on a translatable stage to allow large volumes of tissue to
Fig. 1. TEM picture of 2 ␮m (polylactide)-shelled microbubbles prepared by emul-
be addressed. In many applications this is appropriate and cost-
sification and freeze-drying technology.
effective, but it is not fast. Electronic steering is accomplished by
dividing a therapy transducer into multiple, individually address-
able parts. The electronic signal applied to each element is retarded myocardium but also to other tissues. As noted by Cosgrove [19] and
by a phase shift in such a way to control the location in the tissue Schneider [20], microbubbles can be used for dynamic detection of
where the acoustic waves coherently interfere. Such steering allows macro and microvascular flow in many organs. Microbubbles are
for rapid (on order of milliseconds) changes in treatment location. also used to study the blood supply to the liver. Primary hepatocel-
These arrays have been used in diagnostic imaging and are known as lular carcinomas (HCC) are supplied by the hepatic artery. After a
the class of phased arrays. In therapy applications, the use of phased bolus injection of ultrasound contrast agent, these lesions are high-
arrays has been more limited because of technical challenges as lighted by the perfusion of contrast agent before the rest of the liver
mentioned in the following sections. is fully perfused. Contrast liver imaging has been the subject of a
Several issues must be addressed when using arrays of elements multi-center study and described by Lencioni et al. [21]
in therapeutic drug delivery. In particular, the size of the array can
often be large because of the needed pressures or acoustic energy. 3.2. Thin-shelled and hard-shelled microbubbles
As a result, when dividing into multiple elements, the element
count can be quite large—often into the thousands in order to avoid Microbubble agents can be classified as soft- or thin-shelled
effects such as grating lobes. Grating lobes occur because of the and hard-shelled agents. Ultrasound contrast agents do not only
inadvertent phase coherence occurring in the acoustic field in unin- scatter ultrasound efficiently, they also react to low energy ultra-
tended areas. This complication has implications for drug delivery sound by emitting specific frequencies. Thin-shelled agents are
especially in cases where exposure or release of a drug in a sensitive microbubbles having a lipid monolayer with a thickness of about
organ may lead to undesired side effects. Clever approaches have 2–3 nm. They undergo volume expansions and contractions that
been suggested to avoid these grating lobes without requiring a generate an acoustic signal [22], of which non-linear components
large number of elements, including the use of sparse arrays, irregu- give the most specific information for imaging [23,24]. As the shell
lar shaped and spaced elements, and limiting the number of phases of these microbubbles is so thin, fluorinated gases are needed to
to be applied to the elements to simplify the driving electronics. keep the microbubble stable for a sufficient time in the circula-
tion. Hard-shelled microbubbles have typical shell thicknesses in
3. Pressure-mediated delivery the range of 20–100 nm. An example of polymer-shelled microbub-
bles is given in Fig. 1. They hardly show volume expansions at low
3.1. Ultrasound contrast agents acoustic pressure [25,26]. Nevertheless some of these agents do
generate acoustic signals as well without losing gas. Studies using
Microbubbles used as ultrasound contrast agents are tiny gas an extremely fast camera [27] have given first indications that they
bubbles, small enough to pass the lung capillary bed. To prevent often indent like a badly inflated ball, which is a way to conserve
dissolution of the gas they have a shell made from a lipid, a pro- their surface area and allow for a change in the volume [28,29]. At
tein or a biodegradable polymer. Lipid-shelled microbubbles are higher pressures the microbubbles are destroyed showing dramatic
used in clinical practice and have a monolayer of phospholipid. changes in the gas volume as shown in Fig. 2, where the activa-
An albumin-shelled agent, Optison, is also clinically available and tion of a polymer-shelled microbubble is given. Polymer-shelled
polymer-shelled agents have reached the end of phase III clini- microbubbles do not need very hydrophobic gases to be stable in
cal trials. Microbubbles are used for left ventricle opacification; circulation.
the ultrasound contrast between the blood in the left ventricle
and the myocardium is low and can be increased significantly by 3.3. Targeted microbubbles
intravenous injection of a small number of microbubbles. Typically
108 –109 microbubbles are injected for a diagnostic ultrasound scan. The use of microbubbles is currently being extended to targeted
Microbubbles can be used to improve endocardial border delin- imaging and drug delivery applications. For molecular imaging
eation and, thereby wall motion abnormalities. In addition it allows applications, the shell is coated with specific ligands. A typical
for analysis of myocardial perfusion, which further helps to iden- example is the targeting of endothelial markers of angiogenesis and
tify the myocardium at risk. Perfusion cannot only be applied to the inflammation [30,31].
M.R. Böhmer et al. / European Journal of Radiology 70 (2009) 242–253 245

Fig. 2. Series of events upon insonation of polymer-shelled microcapsule, frames 14–16, 20, 40, 60 of a movie recorded at 15 and 2.25 MHz; MI = 1. Ultrasound switched on
at frame 15. Movie taken on the Brandaris128 camera at Erasmus Medical Centre and Twente University {Chin, 2003 #156).

In the early proof-of-concept phase of research, targeting ligands antibody-carrying microbubbles have been successfully used for
are conveniently bound to the shell via biotin–streptavidin–biotin targeted ultrasound contrast imaging in the areas of TNF-induced
bridges, see Fig. 3. Biotinylated lipids and biotinylated biodegrad- inflammation or ischemia-reperfusion injury [37]. Detection of
able polymers [32] used for microbubble shell preparation are ICAM-1 upregulation in transplant rejection model was achieved by
available or can be synthesized. Microbubbles targeted to vascu- targeting microbubbles with biotinylated anti-ICAM-1 antibodies
lar endothelial growth factors and selectins have shown strong [38]. A large set of studies was conducted at imaging angio-
enhanced ultrasound images in the areas of upregulation of these genic endothelium, via biotinylated antibodies against ␣v ␤3 in the
markers in the vasculature [33]. tumor vasculature setting [39] as well as therapeutic angiogene-
Direct coupling of targeting ligands to the microbubble shell sis [40]. Tumor vasculature status can be evaluated by targeting
using peptide bond formation chemistry [34], see Fig. 3, is nec- streptavidin-carrying lipid microbubbles decorated with biotiny-
essary at the next step towards clinical trials, when the presence lated antibodies against VEGF receptor 2 [41]. The ease of use of
of non-endogenous proteins, such as avidin or streptavidin, is not ultrasound imaging allows comparative targeted imaging of two
desired. markers in the tumor vasculature of the same animals, for instance
Initial model system studies [35] showed that biotinylated endoglin versus VEGF receptor [33].
microbubbles can be targeted to avidin-coated surface in vitro, At this time, covalent coupling methods, lacking avidin–biotin
and ultrasound imaging of these targeted bubbles was success- scheme, are gaining wider acceptance, showing the successful cou-
ful. Most of the experimental targeted ultrasound imaging efforts pling of small ligands [42] or antibodies [43] with good yield. This
have been focused on the various in vivo animal models, from covalent approach will be more applicable in the clinic.
thrombus targeting [36] to the ultrasound imaging of a variety of Binding of antibodies to the vascular endothelium targets is very
molecules upregulated on vascular endothelium. Anti-P-selectin- strong and selective, but the formation of the bond between the
antibody and antigen is, typically, relatively slow. As the microbub-
bles at the target surface experience shear, especially in the fast
(arterial) flow, the relatively long time required to obtain firm
binding might not be sufficient for the antibody; for instance, in
the flow having wall shear stress over 2 dyn/cm2 , anti-P-selectin
antibody-targeted microbubbles are not accumulating at the tar-
get efficiently [44]. To achieve leukocyte adhesion to the inflamed
endothelium, nature has a set of fast-binding ligands on the leuko-
cyte membrane, such as PSGL-1 glycoprotein, that binds to P-
and E-selectin. Glycosulfopeptide-carrying microbubbles were tar-
geting P-selectin-coated surfaces in fast flow conditions quite
successfully [45] A simple variant, essentially a portion of the same
PSGL-1molecule, sialyl Lewis X, can be immobilized on microbub-
bles. Microbubble targeting via this ligand can be assisted by
co-immobilizing the antibody on the bubble, so the rapid attach-
ment of microbubbles to the target is aided by firm antibody binding
[46]. An alternative is to increase the ligand concentration on the
microbubble surface, e.g., by using polymeric version of sialyl Lewis
Fig. 3. Inclusion of drugs in lipid-shelled microbubbles. (A) Lipid-shelled microbub- X, which is available commercially, polymeric sialyl Lewis X, is
ble consisting of gas encapsulated by a lipid monolayer, a fraction of the lipid can be capable of firm but rapid cooperative multipoint binding with the P-
pegylated (not shown). (B) Lipid-shelled microbubble with an additional oil-phase
selectin target surface, and provides efficient microbubble targeting
to increase the reservoir size to incorporate hydrophilic drugs {Unger, 1998 #122}.
(C) Lipid-shelled decorated with liposomes via biotin streptavidin bridges. [47].
246 M.R. Böhmer et al. / European Journal of Radiology 70 (2009) 242–253

Fig. 4. Polymer-shelled microbubbles (A) have a thicker shell into which drugs can be incorporated directly, hydrophilic drugs can be incorporated with a double emulsion
method [60] (B). Half-oil filled polymer-shelled microbubbles (C) give an additional liquid reservoir into which hydrophobic drugs can be incorporated [28], the drug can
either be in solution or precipitated. Multilayer constructs (D) or the attachment of liposomes (E) is also possible with polymer-shelled microbubbles.

3.4. Therapeutic use of microbubbles: sonoporation Miller and Dou [57] investigated the enhancement of lung metas-
tasis from an implanted mouse melanoma tumor after application
For ultrasound induced drug delivery based on microbubbles of ultrasound in the presence of microbubbles. At high pressure
two approaches are distinguished, see for instance the review by (5 MPa) and a 1 Hz rate to avoid heating more lung metastases were
Hernot and Klibanov [5] where the distinction is made between indeed found in the presence of microbubble than in their absence.
co-administration of drugs and microbubbles and drug-loading of However at lower pressure (2 MPa) no enhancement was found. In
the microbubbles themselves. In the case of co-administration the the absence of microbubbles an elevated level of metastasis was
function of the microbubbles is to enhance the permeability of already found in at a peak negative pressure of 5 MPa.
the endothelial wall. This can either be affected by rupture of the
endothelial wall, leading to extravasation of relatively large enti- 3.5. Incorporation of drugs and genetic material in microbubbles
ties such as red blood cells and polymer particles [48] or in a more
subtle way, at lower pressures, by microbubbles causing temporary Instead of co-injecting drugs and microbubbles, microbubbles
opening of cell membranes.Many of the properties of cell mem- can also be modified to contain drugs [58,59] or DNA [60–62]. The
branes are shear dependent. Marmottant and Hilgenfeldt [49,50] advantage, as explicitly shown by Lentacker et al. [61] in an in
demonstrated that the oscillations of microbubbles induce local vitro setting is that the therapeutic molecule is close to where the
shear stress by altering the flow of liquid near the cell surface. Van acoustic action is, and therefore opens the opportunity for a dose
Wamel et al. [51,52] have shown the deformation of cell membranes reduction while maintaining its therapeutic efficacy.
in the presence of an oscillating microbubble directly using an ultra- If drugs have sufficient affinity for the lipid monolayer they can,
fast camera. Sonoporation is the term that is used to describe the in principle, be incorporated directly in lipid-shelled microbub-
formation of pores by ultrasound. If the pores are too large they bles (Fig. 4A). However, as the monolayer is very thin, the amount
cannot reseal leading to cell death, however, if the pores can seal that can be incorporated is extremely low. Unger [59] has added
again they will stay open for a time and, in principle allow passage an additional oil, triacetin, to make a thicker hydrophobic layer
of therapeutics, such as radionuclides [53] or plasmid DNA (see ref. to increase the incorporation of paclitaxel, as shown schemati-
[8] for an overview). Mehier-Humbert et al. [54] investigated the cally in Fig. 4B. This approach is limited to hydrophobic drugs; a
percentage of GFP positive Matt-B III cells following plasmid DNA more general applicable route is to attach liposomes or lipoplexes
delivery using lipid- and polymer-shelled microbubbles. to the microbubble (Fig. 4C). Another option is to bind the drug
A great challenge in sonoporation is to open the blood brain to the outside, for instance in a form associated with lipids, such
barrier in a reversible way. Treat et al. [15] and Hynynen et al. [55] a drug carrying liposomes or lipoplexes: positively charged lipid
have shown that pores made in a rabbit brain close again in about complexed to negatively charged nucleic acid [61]. They have
6 h. They also demonstrated that contrast enhanced MRI is a very demonstrated high transfection efficiency by showing luciferase
suitable method to follow sonoporation. The method has been used activity in vitro. Attachment of liposomes can allow the incorpo-
to deliver doxorubicin across the blood brain barrier in rats, as well ration of both hydrophilic molecules into the aqueous core of the
as to deliver genes under MRI guidance. liposome or hydrophobic drugs into the lipid bilayers. Finally mul-
As stressed in this paragraph the combination of microbubbles tilayer technology [63] can be used to deposit large therapeutic
and ultrasound have an effect on the properties of adjacent cells. A molecules layer by layer on a microbubble surface.
number of authors have found that this effect extends to tumor In principle layer-by-layer deposition or attachment of lipo-
growth. For instance Miller and Song [56] reported that tumor somes is also possible for polymer-shelled agents, see Fig. 5.
growth of renal carcinoma in mice in the presence of Optison and However, different routes are also available to prepare drug loaded
ultrasound is reduced. Damaging tumors by ultrasound could have polymer-shelled agents. Emulsification of a polymer solution con-
an effect on metastasis because of the disintegration of the tumor. taining a carrier solvent and an alkane in an aqueous phase is a
M.R. Böhmer et al. / European Journal of Radiology 70 (2009) 242–253 247

Fig. 5. Schematic representation of targeting by bioti-streptavidin bridges (left) and by NHS coupling (right).

starting point for making polymer capsules. The carrier solvent 3.6. Targeted imaging and drug delivery with microbubbles
is removed from the emulsion droplets and the polymer phase
separates from the shell closing in the alkane in the center. The Microbubbles typically do not exhibit long circulation times.
alkane is subsequently removed by freeze-drying. This prepara- The Reticulo–Endothelial System takes them out of the circula-
tion route is suitable for hydrophobic drugs. Fabrication techniques tion and contrast is observed over time periods of about 20 min
starting from monodispersed emulsions, such as a technique based in humans and a few minutes in mice and rats. The limited cir-
on submerged inkjet-printing [64] allow for a precise incorpora- culation has consequences for targeted imaging and therapy as
tion of a known amount of drug per particle and therefore improve compared to nanomedicine formulations. To achieve a long circu-
the control over the doseage. Direct emulsification techniques can lation time, small particles, around 100–200 nm, have to be chosen,
also yield a narrow size distribution with a maximum between 1 and their surface charges have to be screened, for which espe-
and 2 ␮m and >95% of the microbubbles smaller than 3 ␮m [28]. cially poly-ethylene glycol is used. Although microbubbles can be
Hydrophilic drugs cannot be incorporated directly in an emulsi- surface-modified with PEG, their size is optimized for acoustic
fication procedure. One possibility is to employ double emulsion activity and therefore in the micron range. Fortunately the imag-
technology. In a double emulsion, an aqueous phase, containing ing of microbubbles is very sensitive and as the non-adhering
the drug, is first emulsified in an organic phase (containing the microbubbles disappear rapidly from the blood stream only the
shell-forming polymer), and this first emulsion is subsequently few remaining adhering microbubbles can be imaged. Secondly
emulsified in a second aqueous phase. Poly-lactide-co glycolide the acoustic signature of a microbubble differs, it shifts to lower
microbubbles containing plasmid DNA have been prepared this way frequency, if the bubble adheres [71,72]. To exploit this for imag-
[60]. Gene delivery from these microbubbles was shown in rats and ing, however, monodisperse microbubbles are needed [73]. Finally
also an effect on tumor growth was demonstrated. the use of radiation forces has been explored. These are long, low
An alternative way to prepare polymer-shelled microbubbles is amplitude acoustic pulses that drive the bubbles to the vessel wall
spray-drying [65] Hydrophobic drugs can in principle be included [74–76]. Instead of long circulating small agents that pass by the
in the spray. In the preparation method of Palmowski [66,67], the region of interest many times and may adhere once they pass close
starting point is not the polymer but a monomer. As the polymer- to the vessel wall, microbubbles can be driven to the vessel wall
ization reaction leads to the shell formation directly, incorporation actively.
of a drug at this stage is difficult. Compared to other imaging modalities targeted contrast ultra-
Recent delivery experiments with a new, emerging class of gene sound has the advantage that imaging can be performed relatively
therapeutics, small interfering RNA (siRNA), show promise to over- fast at a high sensitivity. The microbubble size, however, also brings
come the inherent in vivo delivery obstacles of nucleic acids in a drawback as it is more subject to shear forces in the blood flow
general and siRNA in particular, such as rapid excretion via the [32]. Although this has only be shown in a flow cell, the effects
liver, serum instability, non-specific distribution, tissue and cell of shear flow and microbubble displacement upon application of
barricades [68]. The major limitation for the use of siRNA, both in ultrasound are aspects that need further study before ultrasound
vitro and in vivo, is the inability of naked siRNA to passively diffuse imaging with targeted microbubbles can be used for more than
through cellular membranes due to the strong anionic charge of the qualitative purposes.
phosphate backbone and consequent electrostatic repulsion from The limited circulation time of microbubbles also has con-
the anionic cell membrane surface. To deliver siRNA with microbub- sequences for their use as drug delivery vehicles. Treatment
bles, siRNA was either directly attached to the microbubble surface preferably takes place shortly after injection of microbubbles and
or simply mixed with microbubbles prior to administration. In some will be restricted to well-perfused areas. Repeated injections and
studies these vehicles showed enhanced transfection efficiency ultrasound treatments are normally used to evaluate the effect of
both in vitro and in vivo. Further they provide a better protection ultrasound triggered release of drugs from microbubbles in terms
against degradation by serum nucleases. [60,69,70] of tumor growth reduction.
248 M.R. Böhmer et al. / European Journal of Radiology 70 (2009) 242–253

An approach to increase circulation times and aid extravasa-


tion is using nanoparticles, however, this will come at the expense
of decreased imaging possibilities. Nanoparticles, such as micelles
made of blockcopolymers, have been used for ultrasound mediated
drug delivery and are reviewed by Husseini and Pitt [6]. In most
studies, the frequencies used are much lower than in those used in
combination with microbubbles. The mechanism of drug delivery
is also in this case related to cavitation. Rapoport [77] created dox-
orubicin containing polylactide nanoparticles, which, at least for a
fraction of them, contain perfluoropentane. Perfluoropentane has
a boiling point at 27 ◦ C, but in the form of emulsion droplets it is
superheated as shown by Giesecke and Hynynen [78]. Therefore
at body temperature they can still be in the liquid state and phase- Fig. 6. (a) Phase behavior of poly(N-isopropylacrylamide)-based polymers (b) in
converted by a trigger, such as an ultrasound pulse. The doxorubicin water. At temperatures below the LCST the polymer is water soluble (random coil
containing particles are so small that they escape the vasculature configuration), while the chain collapses at temperatures above the LCST.
in a tumor by the enhanced permeation and retention effect and,
when exposed to ultrasound, cause pronounced tumor regression.
sensitive polymers. One strategy pursues micelles formed from
Long time imaging of remaining gas bubbles in tumor tissue was
amphiphilic diblocks made up from a hydrophobic inert block-
possible.
polymer, like polylactic acid, polystyrene, etc. and a PNIPAAm
block [92,93]. The micelle can be loaded with drugs. Below the
4. Temperature sensitive drug delivery systems LCST, this diblock self-assembles into micelles with a hydrophilic
PNIPAAm corona. Heating induces a hydrophilic–hydrophobic tran-
While thermal ablation requires a substantial thermal dose to sition of the PNIPAAm block polymer, leading to a destabilization
induce tissue necrosis, a more subtle temperature increase can and morphology change such as aggregation of the micelles. The
be used to support treatment with conventional chemotherapeu- latter can significantly enhance drug release compared to tem-
tics and drug delivery systems [79]. Mild hyperthermia enhances, peratures below the LCST. The LCST can be fine-tuned in a wider
for example, extravasation of drug loaded liposomes like Doxil© temperature range by designing end-functionalized NIPAAm-based
[80,81] or enhances anti-angiogenic treatment [82]. Hyperther- polymers, copolymer or block polymers [94,95]. However, the com-
mia can also increase local drug concentrations in conjunction plex dependence of the LCST on intramolecular hydrogen bonding
with temperature-induced drug delivery [83]. Temperature sensi- and electrostatic interactions and interactions with water makes
tive drug delivery systems were already explored in combination the LCST susceptible to pH, ionic strength of the solvent and inter-
with hyperthermia induced by radiofrequency (RF) [84] magnetic actions with other molecules. The advantage of delivery systems
particles [85], or by heating with light in the infrared regime [86,87]. where the drug release can be fine-tuned with respect to tempera-
Only recently, some of these temperature sensitive drug delivery ture and pH comes with the disadvantage that drug release becomes
carriers were explored in combination with ultrasound induced also more complex and difficult to control in vivo as the LCST in
drug delivery [88]. The more efficient uptake of drug delivery sys- vitro and in vivo can significantly differ. Though many systems were
tems in tumors at elevated temperatures, together with the local investigated in vitro, little work has been done in preclinical studies
temperature triggered release of drugs, makes ultrasound induced in general or in combination with ultrasound induced hyperthermia
drug delivery a very promising field. in specific. One of the few preclinical studies exploits the interde-
Temperature sensitive drug delivery systems can be designed pendency of pH and temperature to enhance drug delivery in the
following two different approaches. One class of agents is based on more acidic environment of a tumor [96]. Other preclinical studies
amphiphilic temperature sensitive polymers showing a lower crit- showed the feasibility of temperature-induced drug delivery using
ical solution temperature (LCST) in aqueous solution. The second temperature sensitive polymeric micelles [97,98].
class of temperature sensitive drug delivery carriers is based on
liposomes. Here, lipids are used that show a phase transition above
4.2. Temperature sensitive liposomes
body temperature. Upon passing the phase transition temperature,
the liposomal bilayer becomes leaky for drugs encapsulated in the
Liposomal drug delivery systems are a well-studied field and
inner lumen of the liposome.
found their applications in cancer therapy in the clinic [99]. Lipo-
somes can be loaded with different hydrophilic drugs in the inner
4.1. Polymer-based systems water compartment. A particularly high drug payload is achievable
with drugs like doxorubicin or daunorubicin that precipitate in an
Temperature sensitive polymeric drug delivery systems are inner lumen loading mechanism based on a pH gradient. The conve-
usually based on polymers that undergo upon heating a phase nient method of drug loading and the achievable high drug payload
transition associated with a change in polymer–solvent interac- probably explain the dominant role of these drug delivery systems.
tion [89–91]. The solvent properties change from a good solvent Drug release from conventional liposomal formulations is usually
at temperatures below the LCST to a poor solvent at temperatures diffusion controlled, showing little difference in drug release kinetic
above, leading to a morphology change from an extended random at body temperature compared to temperatures slightly above in
coil to a collapsed chain (Fig. 6a). Most polymers studied in this con- the hyperthermia regime. The concept of drug delivery using tem-
text are based on N-isopropylacrylamide (NIPAAm) (Fig. 1b). The perature sensitive liposomes was introduced by Yatvin et al. more
LCST behavior of this polymer is due to a loss of hydrogen bonding than 25 years ago using cis-platinum as a drug compound [100].
between the amino-group and surrounding water, and increased The field of liposome-based drug delivery under hyperthermia was
hydrophobic interactions of the N-isopropylgroups above the tran- reviewed several times [79,101].
sition temperature Temperature sensitive liposomes (TSL) are composed of lipids
Different strategies were followed to design temperature sensi- that show a melting transition of the acyl-chains in the bilayer [102].
tive drug delivery systems based on above mentioned temperature TSLs show a strong increase in drug release around the melting tran-
M.R. Böhmer et al. / European Journal of Radiology 70 (2009) 242–253 249

Fig. 7. Temperature sensitive liposomes containing a drug (red dots) and an imaging or contrast agent that allows visualizing and quantifying the drug delivery process.

sition temperature Tm of the lipids associated with the formation the vascular system, released drugs still need to extravasate, possi-
of transient pores in the bilayer [103,104]. The release temperature bly aided by sonoporation to reach the target tissue. Blood flow
and the release kinetic can be controlled by choosing suitable lipids in the capillaries may carry away the released drug, diminish-
with a Tm in the desired range of around 1–4 K above body temper- ing the effect high local drug concentration. Here, the smaller
ature and the incorporation of lysolipids [104,105], which induce size of temperature sensitive systems potentially offers the advan-
efficient pore formation after the melting transition of the mem- tage that the place of release can be chosen. At early times after
brane. Temperature-induced delivery of doxorubicin encapsulated administration, drug release can take place in the microvascular
in TSLs was thoroughly investigated in preclinical studies and is system when passing through a tissue with elevated temperature.
now in clinical trials [106]. At later time points after administration, the drug delivery car-
rier may have accumulated first in a lesion either by passive or
active targeting mechanisms (e.g., EPR effect or using specific tar-
4.3. Temperature triggered release
geting ligands). Drug release in the interstitial space is triggered
subsequently by heating the lesion using, for example, ultrasound.
In all temperature-induced drug delivery application, the chal-
However, also in the microvasculature an increased concentration is
lenge remains to quantify the amount of release drugs and
found. For example, studies with doxorubicin loaded TSLs revealed
eventually control the release quantitatively. The achieved drug
a 30-fold peak concentration of doxorubicin in the microvascula-
concentration within a treated lesion can be evaluated in preclinical
ture. Pronounced effects on tumor growth are also found if the
studies using standard analytical means, however, imaging tech-
heat-treatment is given shortly after injection [108], therefore we
niques such as nuclear imaging or MRI offer the advantage of being
can only conclude that effective mechanism needs further study.
applicable to humans in a clinical setting. Fig. 7 shows the under-
Treatment response may be a result of anti-vascular effects and
lying idea of incorporating a drug and contrast or imaging agent
anti-neoplastic mode of action [101,113].
inside a thermo-sensitive liposome. Upon heating the drug and
Future work will aim at designing new temperature sensitive
the contrast agents are released. The observable contrast originates
drug carriers that show a sharp and rapid drug release at temper-
from a change of the local environment of the release contrast
atures slightly above body temperature. However, as much work
agents, which correlates with the drug release. Examples are T1 MR
will be needed to optimize the treatment protocol with respect to
contrast agents that are incorporated inside the liposome at high
timing of injection, rate of extravasation, drug release kinetic, and,
concentration, and provide a strong signal once they are released
finally, the timing of ultrasound induced hyperthermia itself with
from the liposome.
existing drug delivery systems. The possibility of controlling the
Nuclear imaging is the method of choice to quantify the amount
drug delivery in real time under image guidance and quantifying
of drug delivery systems accumulated in the lesion [107], or even-
the drug release using, for example, MRI will be essential to estab-
tually also the released drug within the tissue. The disadvantage is
lish a treatment protocol that is superior to today’s approach with
the required radiolabeling of the drug and/or drug carrier and prob-
better therapeutic value.
lematic integration of nuclear imaging in the treatment protocol as
a standard technique. Thus, nuclear imaging will probably keep its
role for validation in a research phase but will not get a major role in 5. Therapeutic applications of ultrasound triggered drug
image-guided drug delivery. More promising is MR imaging of drug delivery
release, especially in the combined setting of an HIFU/MRI system.
Here, MR contrast agents can be incorporated in the drug delivery 5.1. Cardiology
carriers that provide a signal change upon drug release. The value of
this approach was shown using for example temperature sensitive Ultrasound contrast agents have been approved for diagnostic
liposomes filled with Gd-based T1 contrast agents or manganese purposes in the filed of cardiology to better visualize the left ven-
based agents [86,87,102,108–110]. tricle. Therefore many ultrasound contrast agent mediated drug
Temperature sensitive liposomes can also be designed by con- delivery studies have been performed in the heart as recently
jugating PNIPAAm polymers to the liposomal membrane [111,112]. reviewed by Mayer and Bekeradjian [8]. The paper summarizes
Below the LTCS, the hydrophilic PNIPAAm polymer extends into the field of gene delivery using ultrasound mediated delivery tech-
solution and stabilizes the liposome. Upon heating above the LCST, niques and gives examples of studies where the expression of
the hydrophilic to hydrophobic transition leads to a destabiliza- reporter genes was, in rats and mice, enhanced with a factor 3–300
tion of the liposome and in vivo to a very different interaction with using ultrasound frequencies around 1 MHz. Also for therapeutic
cells. genes an overview of a number of studies is given. The majority
No matter which approach is used, all temperature sensi- of work in the cardiology field has focused on gene delivery but
tive drug delivery systems can be prepared in the size range of the ultimate therapeutic aim is to induce angiogenesis and car-
10–300 nm, which presents a major difference with respect to pres- diac repair. Side effects by low molecular weight drugs are less
sure sensitive micro bubbles. As most microbubbles stay within of an issue in cardiology compared to oncology and have, there-
250 M.R. Böhmer et al. / European Journal of Radiology 70 (2009) 242–253

fore, received less attention in the cardiology research on triggered Furthermore, diminished oxygen delivery and hypoxic condi-
delivery. tions cause reduced efficacy of radiation therapy, high level of
As described previously, gas-filled microbubbles are an impor- metabolic products (e.g., carbonic and lactic acid), lower extracel-
tant ultrasonic contrast agent used to enhance edge detection and lular pH and may potentially affect the cellular uptake of some
evaluate myocardial perfusion [114,115]. It was shown in preclinical drugs.
experiments that ultrasound mediated drug delivery can directly Hyperthermia mediated liposomal drug delivery has shown
enhance the expression of adenoviral vectors and plasmid DNA to promise for enhancing local drug deployment while minimizing
the heart [70,116,117,118]. drug distribution outside targeted tissues [101] and is currently
USDD can successfully deliver plasmid DNA to myocardium. In being applied clinically in the treatment of various types of cancer.
optimization experiments, the levels of reporter gene luciferase In cancer therapy, the studies, recently reviewed by Frenkel [3] on
expression were similar to that obtained using adenovirus but temperature sensitive delivery vehicles, are more advanced [106],
without the profound liver uptake associated with adenovirus however, radiofrequency ablation is the heating method, which is
[16]. more invasive, and has less well-defined spatial temperature con-
In addition USDD has also been applied for organ-specific deliv- trol than ultrasound.
ery of other bioactive agents [69] and could facilitate the delivery of The clinical relevance of such controlled and triggered release
protein therapeutics to ultrasound-accessible organs while keeping concepts for drug delivery systems having been demonstrated,
systemic concentrations and side effects low. Vascular endothe- research in this area focuses currently on optimization of cell
lial growth factor (VEGF) bound to albumin microbubbles was specific targeting. These more advanced targeted nanocarriers in
delivered to the heart using USDD. A more than 10-fold increase general have clearly shown their potential in various animal tumor
of cardiac VEGF uptake was seen compared with systemic VEGF models and await clinical application.
administration [118]. Some preclinical studies have demonstrated A more novel approach is to use gene therapy in cancer treat-
that ultrasound alone can facilitate uptake of various substances ment. A crucial requirement for gene therapy is tight control of
including biologics. transgene expression, both spatial and temporal to enhance the
Striking are the examples of Kondo et al. [18] to treat an acute spatial targeting and efficiency of gene delivery. Tissue specific pro-
myocardial infarction resulting in enhanced angiogenesis and lim- moters may also be used to limit transgene expression to targeted
itation of the infarct size. Also, Korpanty et al. [119,120] have tissues, and in that way add a layer of targeting and safety to gene
shown increased density of arterioles and capillaries after ultra- delivery procedures [132].
sound mediated gene transfer. Therapeutic effects could be demonstrated in vivo with var-
ious targeted nucleic acid formulations, such as tumor-targeted
5.2. Oncology DNA plasmids expressing p53 or tumor necrosis factor alpha, small
interfering RNAs knocking down gene expression from tumor spe-
In the last decades cancer has moved from a deadly to a man- cific chromosomal translocations or gene expression of tumor
ageable chronic disease. Cancer in the breast, prostate, liver and neoangiogenic processes, as well as double stranded RNA poly
other organs can be imaged quite accurately with diagnostic ultra- inosine–cytosine which triggers apoptosis in targeted tumor cells
sound [121] and if these carcinomas can be segmented, targeted and [133].
treated with therapeutic ultrasound, a new non-invasive, blood- In a wider sense, gene therapy is experiencing an unprece-
less approach to the treatment of such diseases can be developed. dented renaissance through the emerging field of the novel,
However, after surgical removal and/or treatment with radiation or innovative drug format of small interfering (siRNA). Since the
HIFU of a primary tumor, management of the residual tumor includ- discovery that double stranded (dsRNA) can specifically inhibit
ing metastasis is typically carried out using a variety of systemic expression of homologous genes, RNA interference (RNAi) has
therapies that include small organic molecules, and increasingly become one of the most widely used methods for studying
innovative therapies such as biologics and the emerging siRNA ther- loss-of-function phenotypes in model organisms and is increas-
apeutics [122,123]. ingly used across the whole pharmaceutical research process
For advanced tumor stages, chemotherapy remains the treat- including therapeutics. RNAi has been used to target dominant
ment of choice. Despite the fact that such anticancer agents have a mutant or amplified oncogenes, translocation products, signal-
very effective tumor killing potential in vitro and in animal cancer ing molecules and viral oncogenes such as bcr-abl, mutated ras,
models, they often fail in patients as they are unable to reach all or over expressed Bcl-2. Therapies based upon RNAi may have
tumor cells that are able to regenerate the tumors [124] and, there- a number of inherent, fundamental benefits, such as harnessing
fore, chemotherapy is rarely curative but rather palliative, especially natural pathways and the potential to target virtually any pro-
for solid tumors [125]. tein, i.e., no limitation to “drugable” proteins. In a number of
The microenvironment of a tumor is critical in tumor initiation studies it could be demonstrated that it is possible to deliver
and promotion, and there is increasing evidence that this may be an siRNA intracellularly via microbubble-enhanced focused ultra-
important factor in developing therapeutic approaches [126]. The sound [62,133].
tumor microenvironment, or stroma, influences the growth of the
tumor and its ability to progress and metastasize. It also can limit
the access of therapeutics to the tumor, alter drug metabolism and 6. Conclusions and outlook
contribute to the development of drug resistance. As opposed to
normal tissues, blood vessels in tumors are leaky [127] and vascu- The integration of therapeutic interventions with diagnostic
lature is less spatially organized [128], resulting in the abnormal imaging, to allow for local image-guided delivery, calls for devel-
function of vessels [129]. The combination of impaired blood flow opments in equipment and agents including new therapeutics.
through blockage by neoplasmatic tumor tissue, a leaky vascula- Focused ultrasound in combination with MRI and ultrasound imag-
ture, and lack of functional lymphatics leads to increased interstitial ing has great potential to bring ultrasound triggered drug release
fluid pressures. In addition, the plasma to interstitial gradient of to the clinic, while employing pressure and temperature sensi-
osmotic pressure in tumors is also generally reduced limiting the tive delivery vehicles. The preclinical data demonstrate the specific
extravasation and creating a major obstacle against delivery of ther- solutions that are emerging for local drug and gene delivery in both
apeutic agents [130,131] oncology and cardiology.
M.R. Böhmer et al. / European Journal of Radiology 70 (2009) 242–253 251

Acknowledgement [30] Wallez Y, Huber P. Endothelial adherens and tight junctions in vascu-
lar homeostasis, inflammation and angiogenesis. Biochim Biophys Acta
2008;1778(3):794–809.
The assistance of Dr. Sander Langereis in preparing the figures [31] Walsh DA. Pathophysiological mechanisms of angiogenesis. Adv Clin Chem
is gratefully acknowledged. 2007;44:187–221.
[32] Schmidt B, Souza I, van Beek A, Bohmer M. Adhesion and ultrasound-induced
delivery from monodisperse microbubbles in a parallel plate flow cell. J Con-
trol Release 2008;131:19–26.
References [33] Korpanty G, Carbon JG, Grayburn PA, Fleming JB, Brekken RA. Monitoring
response to anticancer therapy by targeting microbubbles to tumor vascu-
[1] Clement GT. Perspectives in clinical uses of high-intensity focused ultrasound. lature. Clin Cancer Res 2007;13(1):323–30.
Ultrasonics 2004;42(10):1087–93. [34] Villanueva FS, Jankowski RJ, Klibanov S, et al. Microbubbles targeted to inter-
[2] Moyer HR, Delman KA. The role of hyperthermia in optimizing tumor response cellular adhesion molecule-1 bind to activated coronary artery endothelial
to regional therapy. Int J Hyperthermia 2008;24(3):251–61. cells. Circulation 1998;98(1):1–5.
[3] Frenkel V. Ultrasound mediated delivery of drugs and genes to solid tumors. [35] Klibanov AL, Hughes MS, Marsh JN, et al. Targeting of ultrasound contrast
Adv Drug Deliv Rev 2008;60(10):1193–208. material, an in vitro feasibility study. Acta Radiol Suppl 1997;412:113–20.
[4] Ferrara K, Pollard R, Borden M. Ultrasound microbubble contrast agents: fun- [36] Schumann PA, Christiansen JP, Quigley RM, et al. Targeted-microbubble bind-
damentals and application to gene and drug delivery. Annu Rev Biomed Eng ing selectively to GPIIb IIIa receptors of platelet thrombi. Invest Radiol
2007;9:415–47. 2002;37(11):587–93.
[5] Hernot S, Klibanov AL. Microbubbles in ultrasound-triggered drug and gene [37] Lindner JR, Song J, Christiansen J, Klibanov AL, Xu F, Ley K. Ultrasound assess-
delivery. Adv Drug Deliv Rev 2008;60(10):1153–66. ment of inflammation and renal tissue injury with microbubbles targeted to
[6] Husseini GA, Pitt WG. Micelles and nanoparticles for ultrasonic drug and gene P-selectin. Circulation 2001;104(17):2107–12.
delivery. Adv Drug Deliv Rev 2008;60(10):1137–52. [38] Weller GE, Lu E, Csikari MM, et al. Ultrasound imaging of acute cardiac
[7] Hynynen K. Ultrasound for drug and gene delivery to the brain. Adv Drug Deliv transplant rejection with microbubbles targeted to intercellular adhesion
Rev 2008;60(10):1209–17. molecule-1. Circulation 2003;108(2):218–24.
[8] Mayer CR, Bekeredjian R. Ultrasonic gene and drug delivery to the cardiovas- [39] Ellegala DB, Leong-Poi H, Carpenter JE, et al. Imaging tumor angiogenesis with
cular system. Adv Drug Deliv Rev 2008;60(10):1177–92. contrast ultrasound and microbubbles targeted to alpha(v)beta3. Circulation
[9] Mitragotri S. Healing sound: the use of ultrasound in drug delivery and other 2003;108(3):336–41.
therapeutic applications. Nat Rev Drug Discov 2005;4(3):255–60. [40] Leong-Poi H, Christiansen J, Heppner P, et al. Assessment of endoge-
[10] Palussiere J, Salomir R, Le Bail B, et al. Feasibility of MR-guided focused nous and therapeutic arteriogenesis by contrast ultrasound molec-
ultrasound with real-time temperature mapping and continuous sonica- ular imaging of integrin expression. Circulation 2005;111(24):3248–
tion for ablation of VX2 carcinoma in rabbit thigh. Magn Reson Med 54.
2003;49(1):89–98. [41] Willmann JK, Paulmurugan R, Chen K, et al. US imaging of tumor angiogenesis
[11] Vimeux FC, De Zwart JA, Palussiere J, et al. Real-time control of with microbubbles targeted to vascular endothelial growth factor receptor
focused ultrasound heating based on rapid MR thermometry. Invest Radiol type 2 in mice. Radiology 2008;246(2):508–18.
1999;34(3):190–3. [42] Schneider M, Pochon S, Tardy I, et al. Angiogenic molecular imaging with KDR-
[12] Hokland SL, Pedersen M, Salomir R, Quesson B, Stodkilde-Jorgensen H, Moo- targeted microbubbles. Contrast Media Mol Imaging 2007;2:274–304.
nen CTW. MRI-Guided focused ultrasound: Methodology and applications. [43] Della Martina A, Allemann E, Bettinger T, et al. Grafting of abciximab to
IEEE Trans Med Imaging 2006;25(6):723–31. a microbubble-based ultrasound contrast agent for targeting to platelets
[13] Moonen CT. Spatio-temporal control of gene expression and cancer treatment expressing GP IIb/IIIa—characterization and in vitro testing. Eur J Pharm Bio-
using magnetic resonance imaging-guided focused ultrasound. Clin Cancer pharm 2008;68(3):555–64.
Res 2007;13(12):3482–9. [44] Takalkar AM, Klibanov AL, Rychak JJ, Lindner JR, Ley K. Binding and detachment
[14] Moonen CT, Quesson B, Salomir R, et al. Thermal therapies in inter- dynamics of microbubbles targeted to P-selectin under controlled shear flow.
ventional MR imaging. Focused ultrasound. Neuroimaging Clin N Am J Control Release 2004;96(3):473–82.
2001;11(4):737–47, xi. [45] Rychak JJ, Li B, Acton ST, et al. Selectin ligands promote ultrasound contrast
[15] Treat LH, McDannold N, Vykhodtseva N, Zhang Y, Tam K, Hynynen K. Targeted agent adhesion under shear flow. Mol Pharm 2006;3(5):516–24.
delivery of doxorubicin to the rat brain at therapeutic levels using MRI-guided [46] Weller GE, Villanueva FS, Tom EM, Wagner WR. Targeted ultrasound contrast
focused ultrasound. Int J Cancer 2007;121(4):901–7. agents: in vitro assessment of endothelial dysfunction and multi-targeting to
[16] Chen S, Shohet RV, Bekeredjian R, Frenkel P, Grayburn PA. Optimization of ICAM-1 and sialyl Lewisx. Biotechnol Bioeng 2005;92(6):780–8.
ultrasound parameters for cardiac gene delivery of adenoviral or plasmid [47] Klibanov AL, Rychak JJ, Yang WC, et al. Targeted ultrasound contrast agent for
deoxyribonucleic acid by ultrasound-targeted microbubble destruction. J Am molecular imaging of inflammation in high-shear flow. Contrast Media Mol
Coll Cardiol 2003;42(2):301–8. Imaging 2006;1(6):259–66.
[17] Li P, Cao LQ, Dou CY, Armstrong WF, Miller D. Impact of myocardial contrast [48] Price RJ, Skyba DM, Kaul S, Skalak TC. Delivery of colloidal particles and
echocardiography on vascular permeability: an in vivo dose response study red blood cells to tissue through microvessel ruptures created by targeted
of delivery mode, pressure amplitude and contrast dose. Ultrasound Med Biol microbubble destruction with ultrasound. Circulation 1998;98(13):1264–7.
2003;29(9):1341–9. [49] Marmottant P, Hilgenfeldt S. Controlled vesicle deformation and lysis by single
[18] Kondo I, Ohmori K, Oshita A, et al. Treatment of acute myocardial infarction by oscillating bubbles. Nature 2003;423(6936):153–6.
hepatocyte growth factor gene transfer: the first demonstration of myocardial [50] Marmottant P, Hilgenfeldt S. A bubble-driven microfluidic transport element
transfer of a “functional” gene using ultrasonic microbubble destruction. J Am for bioengineering. Proc Natl Acad Sci U S A 2004;101(26):9523–7.
Coll Cardiol 2004;44(3):644–53. [51] van Wamel A, Kooiman K, Emmer M, ten Cate FJ, Versluis M, de Jong N. Ultra-
[19] Cosgrove D. Ultrasound contrast agents: an overview. Eur J Radiol sound microbubble induced endothelial cell permeability. J Control Release
2006;60(3):324–30. 2006;116(2):e100–2.
[20] Schneider M. Molecular imaging and ultrasound-assisted drug delivery. J [52] van Wamel A, Kooiman K, Harteveld M, et al. Vibrating microbubbles poking
Endourol 2008;22(4):795–801. individual cells: drug transfer into cells via sonoporation. J Control Release
[21] Lencioni R, Della Pina C, Crocetti L, Bozzi E, Cioni D. Clinical management of 2006;112(2):149–55.
focal liver lesions: the key role of real-time contrast-enhanced US. Eur Radiol [53] van Wamel A, Bouakaz A, Bernard B, ten Cate F, de Jong N. Radionu-
2007;17(Suppl. 6):F73–9. clide tumour therapy with ultrasound contrast microbubbles. Ultrasonics
[22] Postema M, van Wamel A, Lancee CT, de Jong N. Ultrasound-induced encap- 2004;42(1–9):903–6.
sulated microbubble phenomena. Ultrasound Med Biol 2004;30(6):827– [54] Mehier-Humbert S, Yan F, Frinking P, Schneider M, Guy RH, Bettinger T.
40. Ultrasound-mediated gene delivery: influence of contrast agent on transfec-
[23] Emmer M, van Wamel A, Goertz DE, de Jong N. The onset of microbubble tion. Bioconjug Chem 2007;18(3):652–62.
vibration. Ultrasound Med Biol 2007;33(6):941–9. [55] Hynynen K, McDannold N, Sheikov NA, Jolesz FA, Vykhodtseva N. Local
[24] van der Meer SM, Dollet B, Voormolen MM, et al. Microbubble spectroscopy and reversible blood-brain barrier disruption by noninvasive focused ultra-
of ultrasound contrast agents. J Acoust Soc Am 2007;121(1):648–56. sound at frequencies suitable for trans-skull sonications. Neuroimage
[25] Bloch SH, Wan M, Dayton PA, Ferrara KW. Optical observation of lipid- and 2005;24(1):12–20.
polymer-shelled ultrasound microbubble contrast agents. Appl Phys Lett [56] Miller DL, Song JM. Tumor growth reduction and DNA transfer by cavitation-
2004;84(4):631–3. enhanced high-intensity focused ultrasound in vivo. Ultrasound Med Biol
[26] Bouakaz A, Versluis M, de Jong N. High-speed optical observations of contrast 2003;29(6):887–93.
agent destruction. Ultrasound Med Biol 2005;31(3):391–9. [57] Miller DL, Dou C. The potential for enhancement of mouse melanoma metas-
[27] Chin CT, Lancee C, Borsboom J, et al. Brandaris 128: a digital 25 million tasis by diagnostic and high-amplitude ultrasound. Ultrasound Med Biol
frames per second camera with 128 highly sensitive frames. Rev Sci Instrum 2006;32(7):1097–101.
2003;74(12):5026–34. [58] Shi WT, Bohmer M, van Wamel A, et al. Ultrasound therapy with drug loaded
[28] Kooiman K, Bohmer M, Emmer M, et al. Oil-filled polymer microcapsules microcapsules. In: 2007 IEEE Ultrasonics Symposium. 2007. p. 773–6.
for ultrasound-mediated delivery of lipophilic drugs. J Controlled Release [59] Unger EC, McCreery TP, Sweitzer RH, Caldwell VE, Wu Y. Acoustically active
2009;133:109–18. lipospheres containing paclitaxel: a new therapeutic ultrasound contrast
[29] Marmottant P. unpublished results. agent. Invest Radiol 1998;33(12):886–92.
252 M.R. Böhmer et al. / European Journal of Radiology 70 (2009) 242–253

[60] Hauff P, Seemann S, Reszka R, et al. Evaluation of gas-filled microparticles [91] Meyer DE, Shin BC, Kong GA, Dewhirst MW, Chilkoti A. Drug targeting using
and sonoporation as gene delivery system: feasibility study in rodent tumor thermally responsive polymers and local hyperthermia. J Control Release
models. Radiology 2005;236(2):572–8. 2001;74(1–3):213–24.
[61] Lentacker I, De Smedt SC, Demeester J, Sanders NN. Microbubbles which bind [92] Ganta S, Devalapally H, Shahiwala A, Amiji M. A review of stimuli-
and protect DNA against nucleases. J Control Release 2006;116(2):e73–5. responsive nanocarriers for drug and gene delivery. J Control Release
[62] Vandenbroucke RE, Lentacker I, Demeester J, De Smedt SC, Sanders NN. Ultra- 2008;126(3):187–204.
sound assisted siRNA delivery using PEG-siPlex loaded microbubbles. J Control [93] Oh KT, Yin HQ, Lee ES, Bae YH. Polymeric nanovehicles for anticancer drugs
Release 2008;126(3):265–73. with triggering release mechanisms. J Mater Chem 2007;17:3987–4001.
[63] Borden MA, Caskey CF, Little E, Gillies RJ, Ferrara KW. DNA and polylysine [94] Liu XM, Wang LS. A one-pot synthesis of oleic acid end-capped temperature-
adsorption and multilayer construction onto cationic lipid-coated microbub- and pH-sensitive amphiphilic polymers. Biomaterials 2004;25(10):
bles. Langmuir 2007;23(18):9401–8. 1929–36.
[64] Bohmer MR, Schroeders R, Steenbakkers JAM, et al. Preparation of monodis- [95] Liu XM, Pramoda KP, Yang YY, Chow SY, He CB. Cholesteryl-grafted functional
perse polymer particles and capsules by ink-jet printing. Colloids Surfaces amphiphilic poly(N-isopropylacrylamide-co-N-hydroxylmethylacrylamide):
a-Physicochem Eng Aspects 2006;289(1–3):96–104. synthesis, temperaturesensitivity, self-assembly and encapsulation of a
[65] Straub JA, Chickering DE, Church CC, Shah B, Hanlon T, Bernstein H. Porous hydrophobic agent. Biomaterials 2004;25(13):2619–28.
PLGA microparticles: AI-700, an intravenously administered ultrasound con- [96] Liu SQ, Wiradharma N, Gao SJ, Tong YW, Yang YY. Bio-functional micelles self-
trast agent for use in echocardiography. J Control Release 2005;108(1):21–32. assembled from a folate-conjugated block copolymer for targeted intracellular
[66] Palmowski M, Huppert J, Ladewig G, et al. Molecular profiling of angiogenesis delivery of anticancer drugs. Biomaterials 2007;28(7):1423–33.
with targeted ultrasound imaging: early assessment of antiangiogenic therapy [97] Liu BR, Yang M, Li RT, et al. The antitumor effect of novel docetaxel-loaded
effects. Mol Cancer Ther 2008;7(1):101–9. thermosensitive micelles. Eur J Pharm Biopharm 2008;69(2):527–34.
[67] Palmowski M, Morgenstern B, Hauff P, et al. Pharmacodynamics of [98] Na K, Lee KH, Lee DH, Bae YH. Biodegradable thermo-sensitive nanopar-
streptavidin-coated cyanoacrylate microbubbles designed for molecular ticles from poly(L-lactic acid)/poly(ethylene glycol) alternating multi-block
ultrasound imaging. Invest Radiol 2008;43(3):162–9. copolymer for potential anti-cancer drug carrier. Eur J Pharm Sci
[68] Xie FY, Woodle MC, Lu PY. Harnessing in vivo siRNA delivery for drug discovery 2006;27(2–3):115–22.
and therapeutic development. Drug Discov Today 2006;11(1–2):67–73. [99] Drummond DC, Meyer O, Hong K, Kirpotin DB, Papahadjopoulos D. Optimizing
[69] Unger EC, Hersh E, Vannan M, Matsunaga TO, McCreery T. Local drug and gene liposomes for delivery of chemotherapeutic agents to solid tumors. Pharm Rev
delivery through microbubbles. Prog Cardiovasc Dis 2001;44(1):45–54. 1999;51(4):691–743.
[70] Frenkel PA, Chen S, Thai T, Shohet RV, Grayburn PA. DNA-loaded albumin [100] Yatvin MB, Muhlensiepen H, Porschen W, Weinstein JN, Feinendegen LE. Selec-
microbubbles enhance ultrasound-mediated transfection in vitro. Ultrasound tive delievery of liposome-associated cis-diclorodiammineplatium (II) by heat
Med Biol 2002;28(6):817–22. and its influence on tumor uptake and growth. Cancer Res 1981;41(5):1602–7.
[71] Dayton PA, Pearson D, Clark J, et al. Ultrasonic analysis of peptide- and [101] Ponce AM, Vujaskovic Z, Yuan F, Needham D, Dewhirst MW. Hyperthermia
antibody-targeted microbubble contrast agents for molecular imaging of mediated liposomal drug delivery. Int J Hyperthermia 2006;22(3):205–13.
alphavbeta3-expressing cells. Mol Imaging 2004;3(2):125–34. [102] Viglianti BL, Ponce AM, Michelich CR, et al. Chemodosimetry of in
[72] Vos HJ, Dollet B, Bosch JG, Versluis M, de Jong N. Nonspherical vibrations of vivo tumor liposomal drug concentration using MRI. Magn Reson Med
microbubbles in contact with a wall: a pilot study at low mechanical index. 2006;56(5):1011–8.
Ultrasound Med Biol 2008;34(4):685–8. [103] Needham D, Dewhirst MW. The development and testing of a new
[73] Talu E, Hettiarachchi K, Powell RL, Lee AP, Dayton PA, Longo ML. Maintain- temperature-sensitive drug delivery system for the treatment of solid tumors.
ing monodispersity in a microbubble population formed by flow-focusing. Adv Drug Deliv Rev 2001;53(3):285–305.
Langmuir 2008;24(5):1745–9. [104] Mills JK, Needham D. The materials engineering of temperature-sensitive lipo-
[74] Dayton P, Klibanov A, Brandenburger G, Ferrara K. Acoustic radiation force in somes. Methods Enzymol 2004;387:82–113.
vivo: a mechanism to assist targeting of microbubbles. Ultrasound Med Biol [105] Anyarambhatla GR, Needham D. Enhancement of the phase transition
1999;25(8):1195–201. permeability of DPPC liposomes by incorporation of MPPC: a new
[75] Rychak JJ, Klibanov AL, Ley KF, Hossack JA. Enhanced targeting of ultra- temperature-sensitive liposome for use with mild hyperthermia. J Liposome
sound contrast agents using acoustic radiation force. Ultrasound Med Biol Res 1999;9(4):491–506.
2007;33(7):1132–9. [106] Celsion. www.celsioncom.
[76] Wu J, Nyborg WL. Ultrasound, cavitation bubbles and their interaction with [107] Kleiter MM, Yu DH, Mohammadian LA, et al. A tracer dose of technetium-99m-
cells. Adv Drug Deliv Rev 2008;60(10):1103–16. labeled liposomes can estimate the effect of hyperthermia on intratumoral
[77] Rapoport N, Gao Z, Kennedy A. Multifunctional nanoparticles for combin- Doxil extravasation. Clin Cancer Res 2006;12(22):6800–7.
ing ultrasonic tumor imaging and targeted chemotherapy. J Natl Cancer Inst [108] Ponce AM, Viglianti BL, Yu D, et al. Magnetic resonance imaging of
2007;99(14):1095–106. temperature-sensitive liposome release: drug dose painting and antitumor
[78] Giesecke T, Hynynen K. Ultrasound-mediated cavitation thresholds of liquid effects. J Natl Cancer Inst 2007;99(1):53–63.
perfluorocarbon droplets in vitro. Ultrasound Med Biol 2003;29(9):1359–65. [109] Viglianti BL, Abraham SA, MacFall JR, Bally MB, Dewhirst MW. In vivo mon-
[79] Kong G, Dewhirst MW. Hyperthermia and liposomes. Int J Hyperthermia itoring of real time tissue pharmacokinetics of liposome/drug concentration
1999;15(5):345–70. using MRI. Radiology 2002;225:647–8.
[80] Frenkel V, Etherington A, Greene M, et al. Delivery of liposomal doxorubicin [110] Viglianti BL, Abraham SA, Michelich CR, et al. In vivo monitoring of tissue phar-
(Doxil) in a breast cancer tumor model: investigation of potential enhance- macokinetics of liposome/drug using MRI: illustration of targeted delivery.
ment by pulsed-high intensity focused ultrasound exposure. Acad Radiol Magn Reson Med 2004;51(6):1153–62.
2006;13(4):469–79. [111] Kono K, Yoshino K, Takagishi T. Effect of poly(ethylene glycol) grafts on
[81] Yuh EL, Shulman SG, Mehta SA, et al. Delivery of systemic chemotherapeu- temperature-sensitivity of thermosensitive polymer-modified liposomes. J
tic agent to tumors by using focused ultrasound: study in a murine model. Control Release 2002;80(1–3):321–32.
Radiology 2005;234(2):431–7. [112] Kono K. Thermosensitive polymer-modified liposomes. Adv Drug Deliv Rev
[82] Horsman MR. Angiogenesis and vascular targeting: relevance for hyperther- 2001;53(3):307–19.
mia. Int J Hyperthermia 2008;24(1):57–65. [113] Chen Q, Tong S, Dewhirst MW, Yuan F. Targeting tumor microvessels using
[83] Dromi S, Frenkel V, Luk A, et al. Pulsed-high intensity focused ultrasound and doxorubicin encapsulated in a novel thermosensitive liposome. Mol Cancer
low temperature-sensitive liposomes for enhanced targeted drug delivery and Ther 2004;3(10):1311–7.
antitumor effect. Clin Cancer Res 2007;13(9):2722–7. [114] Amyot R, Morales MA, Rovai D. Contrast echocardiography for myocardial
[84] Friedman M, Mikityansky I, Kam A, et al. Radiofrequency ablation of cancer. perfusion imaging using intravenous agents: progress and promises. Eur J
Cardiovasc Int Radiol 2004;27(5):427–34. Echocardiogr 2000;1(4):233–43.
[85] Pankhurst QA, Connolly J, Jones SK, Dobson J. Applications of magnetic [115] Mayer S, Grayburn PA. Myocardial contrast agents: recent advances and future
nanoparticles in biomedicine. J Phys D-Appl Phys 2003;36(13):R167–81. directions. Prog Cardiovasc Dis 2001;44(1):33–44.
[86] Salomir R, Palussiere J, Fossheim SL, et al. Local delivery of magnetic reso- [116] Shohet RV, Chen S, Zhou YT, et al. Echocardiographic destruction of albu-
nance (MR) contrast agent in kidney using thermosensitive liposomes and min microbubbles directs gene delivery to the myocardium. Circulation
MR imaging-guided local hyperthermia: a feasibility study in vivo. J Magn 2000;101(22):2554–6.
Reson Imaging 2005;22(4):534–40. [117] Bekeredjian R, Chen S, Frenkel PA, Grayburn PA, Shohet RV. Ultrasound-
[87] Bos C, Lepetit-Coiffe M, Quesson B, Moonen CTW. Simultaneous monitoring targeted microbubble destruction can repeatedly direct highly specific
of temperature and T-1: methods and preliminary results of applica- plasmid expression to the heart. Circulation 2003;108(8):1022–6.
tion to drug delivery using thermosensitive liposomes. Magn Reson Med [118] Bekeredjian R, Grayburn PA, Shohet RV. Use of ultrasound contrast agents
2005;54(4):1020–4. for gene or drug delivery in cardiovascular medicine. J Am Coll Cardiol
[88] Deckers R, Rome C, Moonen CT. The role of ultrasound and magnetic resonance 2005;45(3):329–35.
in local drug delivery. J Magn Reson Imaging 2008;27(2):400–9. [119] Korpanty G, Chen S, Shohet RV, et al. Targeting of VEGF-mediated angiogenesis
[89] Rijcken CJ, Soga O, Hennink WE, van Nostrum CF. Triggered destabilisation of to rat myocardium using ultrasonic destruction of microbubbles. Gene Ther
polymeric micelles and vesicles by changing polymers polarity: an attractive 2005;12(17):1305–12.
tool for drug delivery. J Control Release 2007;120(3):131–48. [120] Korpanty G, Grayburn PA, Shohet RV, Brekken RA. Targeting vascular
[90] Chilkoti A, Dreher MR, Meyer DE, Raucher D. Targeted drug delivery by ther- endothelium with avidin microbubbles. Ultrasound Med Biol 2005;31(9):
mally responsive polymers. Adv Drug Deliv Rev 2002;54(5):613–30. 1279–83.
M.R. Böhmer et al. / European Journal of Radiology 70 (2009) 242–253 253

[121] Thuroff S, Chaussy C, Vallancien G, et al. High-intensity focused ultrasound [128] McDonald DM, Choyke PL. Imaging of angiogenesis: from microscope to clinic.
and localized prostate cancer: efficacy results from the European multicentric Nat Med 2003;9(6):713–25.
study. J Endourol 2003;17(8):673–7. [129] Jain RK. Antiangiogenic therapy for cancer: current and emerging concepts.
[122] Haussecker D. The business of RNAi therapeutics. Hum Gene Ther Oncology (Williston Park) 2005;19(4 Suppl 3):7–16.
2008;19(5):451–62. [130] Boucher Y, Baxter LT, Jain RK. Interstitial pressure gradients in tissue-
[123] Reischl D, Zimmer A. Drug delivery of siRNA therapeutics: potentials and limits isolated and subcutaneous tumors: implications for therapy. Cancer Res
of nanosystems. Nanomedicine 2008. 1990;50(15):4478–84.
[124] Minchinton AI, Tannock IF. Drug penetration in solid tumours. Nat Rev Cancer [131] Di Paolo A, Bocci G. Drug distribution in tumors: mechanisms, role in drug
2006;6(8):583–92. resistance, and methods for modification. Curr Oncol Rep 2007;9(2):109–
[125] Lake RA, Robinson BW. Immunotherapy and chemotherapy—a practical part- 14.
nership. Nat Rev Cancer 2005;5(5):397–405. [132] Greenberger S, Shaish A, Varda-Bloom N, et al. Transcription-controlled
[126] Witz IP. Tumor-microenvironment interactions: dangerous liaisons. Adv Can- gene therapy against tumor angiogenesis. J Clin Invest 2004;113(7):1017–
cer Res 2008;100:203–29. 24.
[127] Galmarini CM, Galmarini FC. Multidrug resistance in cancer therapy: role of [133] Russ V, Wagner E. Cell and tissue targeting of nucleic acids for cancer gene
the microenvironment. Curr Opin Investig Drugs 2003;4(12):1416–21. therapy. Pharm Res 2007;24(6):1047–57.

Вам также может понравиться