Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

J. of Cardiovasc. Trans. Res.

(2013) 6:729–739
DOI 10.1007/s12265-013-9501-0

Ultrasound Contrast Materials in Cardiovascular Medicine:


from Perfusion Assessment to Molecular Imaging
Alexander L. Klibanov

Received: 3 June 2013 / Accepted: 8 July 2013 / Published online: 3 August 2013
# Springer Science+Business Media New York 2013

Abstract Ultrasound imaging is widely used in cardiovas- Introduction


cular diagnostics. Contrast agents expand the range of tasks
that ultrasound can perform. In the clinic in the USA, endo- Ultrasound imaging is widely applied in cardiovascular med-
cardial border delineation and left ventricle opacification icine, especially as a tool for rapid diagnostics in case of
have been an approved indication for more than a decade. urgent decision-making conditions. Ultrasound imaging sys-
However, myocardial perfusion contrast ultrasound studies are tems are now easily available and affordable worldwide.
still at the clinical trials stage. Blood pool contrast and perfusion They have always been portable; hand-held and pocket
in other tissues might be an easier indication to achieve: general equipment versions have now become a convenient option.
blood pool ultrasound contrast is in wider use in Europe, Ultrasound provides real-time anatomic and functional (e.g.,
Canada, Japan, and China. Targeted (molecular) contrast wall motion abnormality) information that could be applied
microbubbles will be the next generation of ultrasound imaging for diagnostics and image-guided interventions. Ultrasound
probes, capable of specific delineation of the areas of disease by imaging is usually performed by a transducer probe placed
adherence to molecular targets. The shell of targeted on the skin above the organ or tissue of interest. For higher
microbubbles (currently in the preclinical research and early image quality the probe can be inserted in the body (e.g.,
stage clinical trials) is decorated with the ligands (antibodies, intra-cavity—such as trans-esophageal, for high-resolution
peptides or mimetics, hormones, and carbohydrates) that ensure heart imaging, or intravascular, thin catheter-based, for high-
firm binding to the molecular markers of disease. frequency observation of stent placement and vascular
plaque monitoring with spatial resolution as low as tens of
Keywords Ultrasound contrast . Microbubbles . Perfusion . micrometers).
Molecular imaging . Inflammation . Ischemia–reperfusion Doppler ultrasound has been used for decades to as-
injury . P-selectin . E-selectin . VCAM-1 sess blood flow—but that technique is limited to large
vessels. Ultrasound imaging benefits from the develop-
Associate Editor Angela Taylor oversaw the review of this article. ment of contrast agents, as is common for all other
imaging modalities. Such agents, based on gas-filled
Electronic supplementary material The online version of this article
(doi:10.1007/s12265-013-9501-0) contains supplementary material,
microspheres, were proposed decades ago [1, 2], but
which is available to authorized users. are only now starting to gain wider clinical acceptance.
Blood pool agents represent the first [3, 4] and second
A. L. Klibanov (*)
Division of Cardiovascular Medicine and Cardiovascular Research [5–7] generations of ultrasound contrast. Targeted
Center, University of Virginia School of Medicine, Charlottesville, microbubbles that provide contrast for ultrasound molec-
VA 22908, USA ular imaging have already reached clinical trials stage
e-mail: sasha@virginia.edu
[8]. This review will focus on the design, logic, and
A. L. Klibanov potential translational use of the injectable microbubble
e-mail: SKLIB1@gmail.com materials for contrast ultrasound imaging.
730 J. of Cardiovasc. Trans. Res. (2013) 6:729–739

Design of Microbubbles for Intravascular Contrast to combine anatomy and microbubble data. Most effective
Ultrasound microbubble detection routines apply multi-pulse tech-
niques, which combine phase inversion with power modula-
The concept of ultrasound contrast is based on the difference tion of transmitted ultrasound [12]. Sensitivity of contrast
between compressibility of water (extremely low) and gas detection is excellent, with the ability to observe individual
(very high). When ultrasound (it is a pressure wave) travels microbubble particles of ultrasound contrast, in real-time,
through the uniform biological tissue (which consists of without destruction, many centimeters deep in the tissues
water to a significant extent and is almost incompressible), [13]. Contrast microbubbles typically are ∼1–3 μm in diam-
it propagates with minimal loss of energy. When an ultra- eter, and mass of each particle is ∼0.1–1 pg. A typical dose of
sound pulse reaches a microbubble in a tissue (e.g., in the these microbubbles does not exceed several billion particles
bloodstream), its gas core compresses and expands in accor- in case of intravenous administration in humans. Therefore,
dance with the respective positive or negative acoustic pres- very small amount of material, sub-milligram dose in
sure [9]. As a result, the excursion of the gas–liquid interface humans, is sufficient to provide successful ultrasound con-
results in the generation of a secondary ultrasound wave: trast. It is several orders of magnitude less than the required
each bubble becomes a small point source “speaker”, which dose of X-ray or MRI contrast. This is a significant charac-
effectively scatters ultrasound. Luckily, resonance frequency teristic, which is especially important for the design of con-
of micrometer-size gas bubbles is the same as the frequency trast agents useful for molecular imaging, which may gain
applied for medical ultrasound imaging. Therefore ultra- clinical use in the future. However, the initial success for
sound imaging can detect intravascular microbubbles that contrast ultrasound practical application in the clinic was
have size smaller than red blood cells and can freely circulate achieved with the non-targeted, blood pool contrast agents.
in the bloodstream without being trapped in the blood
capillaries.
Expansion and compression of the gas void is crucial for Perfusion Studies with Contrast Ultrasound
generation of acoustic backscatter signal from microbubbles.
This implies the need for a very thin (several nanometers) In the initial early-stage ultrasound contrast experiments,
shell, either a simple lipid or surfactant monolayer [3] or a microbubbles were manufactured from air immediately be-
thin coat of denatured albumin [4] which could be easily fore injection and did not have any stabilizer shell; these
expanded or compressed even by the moderate acoustic early generation air particles were extremely unstable. They
pressure (ultrasound Mechanical Index (MI) <0.1–0.2, de- were not able to pass through the capillaries and could not be
pending on the frequency), without destroying the bubbles, used as blood pool imaging agents following intravenous
so that repeated real-time imaging at tens of frames per administration, only intra-arterial. Next, air-filled bubbles
second can be performed. An alternative approach, where with a stabilizer shell were introduced [3, 4]. These agents
the microbubbles are subjected to ultrasound with higher were able to provide transpulmonary blood pool contrast
acoustic pressure, still lower than the approved maximum useful for cardiac imaging: after intravenous injection,
of the transmit pressure, results in a destruction of micro- microbubble contrast signal from the blood in the left ven-
bubbles within a single imaging frame. Backscatter generat- tricle was obtained. Circulation lifetime of these micro-
ed during microbubble destruction is extremely intense and bubbles in the bloodstream was rather short (seconds), so
nonlinear, so it can be easily detected even by regular B- the second generation of microbubbles was brought to clin-
mode ultrasound imaging [10]. In some of the ultrasound ical practice, which contained biocompatible fluorinated
contrast agent designs, e.g., those entrapped in a thick poly- gases with low solubility in blood [5–7]. Attempts to expand
mer shell [11], destruction is required for bubble imaging: the indication of second generation microbubble agents from
the shell has to crack to ensure efficient vibration of the gas– the approved “endocardial border delineation and left ven-
liquid interface. This scenario is only implemented at higher tricle opacification” to myocardial perfusion brought a vari-
acoustic pressures (MI>0.6), and imaging acoustic pressure ety of clinical trials, large and small scale [14, 15], but are yet
threshold depends on the thickness of the polymer shell to gain an approved indication status. In part this is due to the
surrounding the gas core. fact that the heart is a difficult organ to image: the heart is
Some of the clinical ultrasound imaging systems (e.g., located rather deep beneath the skin surface, and tissue (e.g.,
Sequoia 512 with CPS) are outfitted with low power contrast fat) attenuation of ultrasound signal may be significant.
agent detection capabilities for some of the available probes Ultrasound propagation is hindered by the ribcage, and by
(Fig. 1). These contrast-specific presets enhance the signal the fact that high concentration of contrast microbubbles
from ultrasound contrast agents and efficiently suppress the present in the heart chambers during the perfusion study in
background signal from the tissue itself (Fig. 2); fusion the immediate proximity to the heart muscle will attenuate
frames of contrast and grey-scale images can be generated ultrasound. To make myocardial perfusion imaging even
J. of Cardiovasc. Trans. Res. (2013) 6:729–739 731

Fig. 1 Ultrasound contrast Dilute microbubble dispersion imaging


imaging of individual
microbubbles at high dilution, before and after destructive ultrasound.
suspended in normal saline. Left
panel imaging at Mechanical
Index (MI) 0.2 (nondestructive,
low power). Right panel imaging
MI 1.9
following 2-s exposure to MI 1.9
ultrasound
leads to destruction of
microbubbles within the beam
elevation. Imaging performed on
Siemens Sequoia with 15 L8
probe, 7 MHz, CPS mode, 2 cm
depth, 1 cm focus
MI 0.2 nondestructive imaging: following 2 seconds of MI 1.9
detection of individual ultrasound: most of the insonated
microbubbles, ~1-3 um diameter, bubbles destroyed quickly
<1pg mass

more complicated, heart muscle is in constant 3D motion, In other organs, where tissue motion is not a significant
both in a cardiac cycle, and a displacement due to breathing. issue, destruction-replenishment perfusion studies can be
A robust approach to myocardial perfusion by ultrasound performed in real-time, where an intense ultrasound pulse
contrast imaging was suggested very early by Wei and Kaul quickly destroys the bubbles in the interrogated area first
[16], where imaging of circulating microbubbles was (e.g., Fig. 1, right frame), and is then followed by real-time
performed as intermittent high-power destructive frames, (up to ∼20–50 Hz) non-destructive low-power imaging of
synchronized with the heart cycle (e.g., images taken at microbubble contrast (Supplement Video 1). This technique
end-systole). The method used in that study was to perform allows observation of the influx of microbubbles into the
destruction-replenishment imaging at the next contraction, vasculature of interest. Modern low-power ultrasound detec-
then missing one heartbeat, missing two heartbeats, then tion schemes provide tracking of bubble trajectories and thus
three, four, etc., so that replenishment kinetics of micro- obtaining the maps of the feeding vasculature [18]. This
bubbles in the blood perfused through the interrogated area approach is also useful for kidney perfusion assessment [19].
could be deduced. Destruction of microbubbles can provide Real-time high sensitivity non-destructive ultrasound
superb acoustic signal for contrast detection even for contrast imaging may be quite useful for assessment of
polymer-coated bubbles [17]. Synchronization with heart low-perfusion vasculature. One important example is imag-
cycle can provide alignment of the interrogated tissue for ing of vasa vasorum within the active atherosclerotic plaque in
all the frames in the sequence, so that the blood replenish- the carotid arteries [20]. High-sensitivity and high-resolution
ment rate could be computed for every pixel of the imaged nondestructive contrast ultrasound imaging capability is nec-
tissue. essary to perform vasa vasorum contrast imaging: vasculature

Fig. 2 Ultrasound imaging of a Greyscale B-mode ultrasound vs. tissue suppression by a


tissue immersed in a water tank.
Left panel anatomy imaging (B- multipulse phase inversion power modulation.
mode gray-scale). Right panel
CPS contrast mode suppresses
the signal from the tissue lacking
microbubbles in the
bloodstream—thus improving
the signal/noise ratio for contrast
ultrasound imaging. Imaging
performed on Siemens Sequoia
with 15 L8 probe, 7 MHz, 2 cm
depth, 1 cm focus, MI 0.2

1 cm 1 cm
Greyscale-B mode imaging of a tissue CPS contrast mode: almost complete
sample in a water tank. suppression of tissue signal; no
microbubbles present in the tissue.
732 J. of Cardiovasc. Trans. Res. (2013) 6:729–739

in the active plaque has very low density, therefore the move- target receptor. Due to microbubble size (micrometer range)
ment of individual microbubbles can provide information we do not expect contrast agent extravasation. Therefore, to
whether the plaque has active vasculature. Imaging is compli- achieve microbubble adhesion, target receptors have to be
cated by the fact that the large artery lumen in an immediate located within the vascular bed, e.g., on vascular endotheli-
proximity to the plaque is filled with microbubbles, which um. Within 5–15 min the recirculating non-bound micro-
would cause ultrasound attenuation. Consequently, intra- bubbles are cleared from the bloodstream almost completely
plaque vessels can be imaged effectively only in the plaque (mostly by collapse, via gas loss from the core; gas is
located between the skin (and transducer face) and the artery removed from the body by exhalation, via the lungs). After
lumen; monitoring the status of a distal plaque may be com- this dwell period, molecular ultrasound imaging can be
plicated. Even more complicated will be to assess the plaque performed by detecting the bubbles retained by the target
status in the coronary arteries. Most likely, intravascular con- receptor molecules in the region of interest (Fig. 3 cartoon of
trast ultrasound capability will be required for the latter [21]. the concept; also see Fig. 4, left panel as an example of
Possibly, many of these issues could be resolved by the use of targeted microbubbles in vitro). To confirm binding speci-
targeted microbubble imaging, where targeting ligands against ficity, and to verify that circulating microbubbles do not
a vascular marker of disease is immobilized on the micro- contribute to the signal, acoustic power of the imaging sys-
bubble shell [22]. tem can be increased to destroy the adherent (target-bound)
bubbles, and imaging repeated to observe the contrast from
the few circulating microbubbles and/or occasional tissue
Ultrasound Molecular Imaging with Ultrasound speckle signals (Fig. 1, right panel).
Contrast
Microbubble Design: Specific Formulations
Targeted Ultrasound Imaging: General Principles
Microbubble formulation strategy relies generally on three
Molecular imaging with ultrasound contrast is performed in options. First, contrast agent microparticles (gas-filled
a manner similar to other imaging modalities (e.g., radio- microbubbles surrounded by a thin shell) can be generated
labeled ligands or fluorescent probes). The shells of micro- ahead of time and packaged in the vials for extended storage
bubbles are decorated with a targeting ligand directed to- [13, 22]. The shell should be thin, typically, monomolecular,
wards a molecular marker of disease. Following intravenous to allow successful vibration of the bubble in the ultrasound
administration, targeted microbubbles circulate with the pressure field. At the same time, the shell should be sturdy
blood flow, and during this period they have a chance to enough to withstand long-term storage in the aqueous media,
adhere to the zones of the vasculature that overexpress the without a significant loss of a gas core, for several years of

Fig. 3 Targeted microbubble: an


ultrasound molecular imaging
agent. Typical microbubble size:
1–3 μm. Coating with a
monomolecular shell (e.g.,
∼2 nm thick lipid monolayer)
and a grafter brush of PEG
polymer, decorated with a
targeting ligand. Microbubble
will selectively attach to the
target surfaces that carry as little
as ten target molecules per
square micrometer
J. of Cardiovasc. Trans. Res. (2013) 6:729–739 733

Microbubbles on a target surface:


imaging before and after destructive ultrasound.

MI 1.9
ultrasound
pulses

before after
Fig. 4 Ultrasound imaging of microbubbles attached to the Petri dish of microbubbles in the patch (confirmed by microscopy). Imaging
surface (∼0.5 cm patch). Imaging performed in a saline tank, non- performed on Siemens Sequoia with 15 L8 probe, 7 MHz, CPS mode,
destructively, at MI 0.2 (left panel) and following destructive ultrasound 2 cm depth, 1 cm focus
treatment (MI 1.9), right panel, leading to almost complete destruction

refrigerated storage in sealed vials under the headspace of the formation of gas microbubbles stabilized with a lipid shell
same gas as is inside microbubbles. This can be achieved by dispersed in the aqueous phase [7]. Manufacturing process is
using a denatured protein shell (albumin) as in Albunex and easy and quick (Fig. 5 and Supplement Video 2): bubbles can
Optison microbubbles, or by using a phospholipid monolay- be prepared by the patient bedside, in the doctor’s office or in
er decorated with an additional PEG polymer brush coat, so the hospital pharmacy, to be used within hours.
that microbubbles would not fuse when they form a dense
“cake” at the top of the aqueous phase in the vial upon Microbubble Shell and Gas Core Selection: Imaging
storage. Microbubble cake needs to be dissociated into indi- Considerations
vidual bubble aqueous dispersion by gentle mixing, e.g., by
hand rotation, immediately before administration. The microbubble shell is a crucial component of the particle
Second approach is based on the dry powder matrix design. If it is too thick and non-compliant polymer (e.g.,
precursor, which is water-soluble and can be reconstituted polylactide [11]), the only way to obtain efficient acoustic
with the aqueous medium immediately before use. The solid response it to crack it with high acoustic power (see
formulation is exceptionally stable on storage: all the com- Section “Design of Microbubbles for Intravascular Contrast
ponents are in the dry state, lacking any water, so there is Ultrasound”)—but that will lead to bubble destruction and
almost no chance of microbubble fusion, ligand denatur- inability to repeat imaging of the same area and obtain
ation, or a hydrolytic/chemical degradation. The vial with targeted contrast signal. For thin (several nanometers) shells
the dry matrix is sealed, in most cases with a non-reactive there is another limiting factor: gas exchange can happen
(e.g., perfluorinated) gas atmosphere inside. A soluble pre- quickly through a thin membrane. Air-filled microbubbles
cursor often is PEG [6, 23] or carbohydrate [3]. When water lose their gas, and thus the ultrasound contrast capability,
is added to the vial that contains dry matrix with embedded quite quickly after intravenous administration. This happens
lipid or polymer shell components, the matrix is quickly within seconds if oxygen-only breathing gas is applied (ni-
penetrated by the aqueous phase and dissolved, leaving trogen is extracted from the bubbles into nitrogen-depleted
behind the pockets of gas phase that form bubbles blood) [26]. Perfluorocarbon bubbles circulate for much
surrounded by the insoluble stabilizer shell in the aqueous longer period (minutes) because of low solubility of these
phase, ready for injection. Several non-targeted ultrasound gases in water or blood, even if the shell is quite thin and
contrast formulations are prepared this way, such as Sonovue acoustic response without bubble destruction is possible.
[6] or Imagent [24], as well as targeted formulations, such as
BR55 [23] or Sonazoid [25]. This formulation implies the Ligand Attachment to the Microbubble Shell. Choice
use of expensive lyophilizers or spray drying equipment. of Ligands for Efficient Targeting
Third approach is based on an amalgamation principle,
where a sealed 2-ml vial that contains gas and sterile aqueous Molecular ultrasound imaging succeeds only when ligand-
liquid phase with a stabilizer lipid mixture is placed in the coated microbubbles attach to the target receptor and are
holder arm of a small desktop apparatus (a slightly modified retained on the target surface for the duration of the imaging
version of a dental amalgamator), and subjected to high- exam (Fig. 3). For this scenario to take place, multiple
shear mixing. Vibration occurs with an amplitude of ∼2 cm ligand-receptor binding events need to occur on the contact
and ∼4,000 rpm, for less than a minute. It results in the zone between the bubble and target surface; otherwise, the
734 J. of Cardiovasc. Trans. Res. (2013) 6:729–739

Fig. 5 Microbubble preparation Aqueous lipid mixture with perfluorocarbon gas


by amalgamation. Left panel
aqueous lipid mixture under headspace: before and after microbubble preparation
perfluorocarbon atmosphere in a
sealed vial. Right panel aqueous
dispersion of perfluorocarbon
microbubbles (>109 particles per
milliliter) following 45 s of
amalgamator treatment at
4,000 rpm. Microbubbles are
stable for many hours after
preparation
45 seconds
amalgamation

before after

interaction may be too weak to retain the bubble in place in improvement of microbubble targeting efficacy [32, 33].
the significant shear created by the blood flow. The higher Microscopic structures (folds) on the surface of the lipid
the ligand density, the better the resistance of adherent monolayer shell ensure more efficient adhesion of micro-
microbubbles to the shear flow [27]. But the main issue is bubbles to the target [34]. A combination of several of those
not with the efficacy of retention of adherent microbubbles: approaches may be useful for targeting enhancement, as well
to dislodge the bubbles already bound and retained on the as a combination of two [35, 36] or more ligands on the
target surface, very fast flow is needed, well in excess of bubble surface.
physiological conditions [28]. The main problem is the low
efficacy of initial microbubble adhesion in the faster flow
conditions. As microbubbles pass by the target surface with Molecular Ultrasound Imaging: Applications
the flow of blood quite quickly, the ligand on the bubble shell
has very limited time to bind to the surface receptor (for Targeted Microbubbles in Inflammation Imaging
meter per second flow speed, estimated contact time for
ligand and target molecules can be as short as nanoseconds). Targeted microbubble imaging of inflammatory conditions
Therefore, many antibodies, which are selected by overall may rely on two scenarios: (1) imaging of activated leuko-
affinity but not the rate of association with the target recep- cytes and (2) imaging of the markers of activated vascular
tor, are unable to achieve effective targeting in fast flow [28]. endothelium. The first approach is quite simple, and it is
The simplest approach is to increase the ligand surface somewhat surprising that it has not been reported in the
density on the microbubble exterior, which may not be clinical setting so far. It relies on the ability of leukocytes
possible for antibodies (typically, only ∼105 molecules per to actively bind apoptotic cells and particles and internalize
microbubble), but is quite feasible for smaller peptide or them via phagocytosis, due to phosphatidylserine recogni-
peptide mimetic ligand molecules [23]. Nature addresses this tion [37]. When microbubbles are prepared as mimics of
problem by combining rapidly adhering ligands, such as apoptotic cells, by simply incorporating phosphatidylserine
sialyl Lewis X, with extended molecular structures, such as in their shell, they adhere to phagocytic cells quickly and
PSGL-1 or L-selectin, and micron-size extensions on the efficiently [38, 39]. Bubbles bind mostly to Kupffer cells in
surface of leukocytes. Same approaches can be applied for the liver and to spleen macrophages, but in case of signifi-
the microbubble ligand attachment. Polymeric sialyl Lewis cant presence of activated neutrophils adherent to the vascu-
X (or Lewis A) oligosaccharide derivatives [29, 30] or short lature in the inflamed tissue, then phosphatidylserine
glycosulfopeptides [31] provide efficient microbubble microbubbles would specifically adhere to neutrophils and
targeting in fast flow, as rapidly binding selectin ligands. mark the inflamed area for targeted imaging. Control bubbles
An extended PEG spacer arm, ∼20–30 nm long, provides that lack phosphatidylserine do not demonstrate this effective
J. of Cardiovasc. Trans. Res. (2013) 6:729–739 735

adherence. This approach has been successful for detecting targeted microbubbles successfully visualized tissue re-
neutrophils within the inflamed vasculature in a variety of jection in a heterotrophic transplant model in rats [51],
inflammation models [39, 40]. so contrast ultrasound molecular imaging may serve as a
The second approach, more complicated in terms of con- non-invasive tool to alert of the need for an aggressive
trast agent design and formulation, relies on the direct mo- intervention.
lecular ultrasound imaging of the specific biomarkers on the
surface of activated vascular endothelium. A set of mole-
cules upregulated on the surface of vascular endothelium in Molecular Ultrasound Imaging of Thrombi or Clot
response to inflammation is relatively narrow as reported at Components
this point: ICAM-1 [41], VCAM-1 [42], P-selectin [43], or E-
selectin [44, 45]. Initially, for preclinical proof-of-principle It is generally assumed that clot imaging by ultrasound may
studies, monoclonal antibodies against these receptors were not require contrast or targeting: a large dark mass of the clot
successfully applied [41, 43]. These ligands offer excellent in a vessel is often detectable by non-contrast ultrasound,
binding affinity to the target receptors, and outstanding spec- with the aid of Doppler. Contrast in the bloodstream may aid
ificity, with low nonspecific background in normal tissues. A in the delineation of the clot contours. However, detection of
significant disadvantage of these molecules is their large size small clot on the surface of active plaque, or imaging of
(typical molecular mass of an IgG is ∼150 KDa, with only a activated platelets adherent on the vessel wall might not be
small fragment useful for targeting) and origin (mostly mouse feasible without targeting. Molecular ultrasound imaging
or rat, foreign proteins immunogenic to a human). Humanized can be useful in these situations. Microbubbles can be out-
or fully human antibodies are gradually gaining ground as fitted with antibodies against GPIIb/IIIa [52] or RGD pep-
injectable therapeutic and diagnostic agents, as are novel tides [53] to achieve clot targeting. Alternatively, P-selectin
methods of their discovery [46]. Preparation and design of expressed on the surface of activated platelets can be targeted
shorter fragments (such as nanobodies [47] or affibodies [48]) [30]. In the latter approach, application of rapidly adhering
may also be an option. However, translation of targeted targeting ligand Sialyl Lewis A for microbubble coating
microbubbles to clinical practice will most likely occur first ensures efficient targeting in the conditions of very fast flow,
with even smaller molecules, such as peptides, peptide mi- with the ultimate goal to mark arterial vascular injuries and
metics, or oligosaccharide (see Section “Ligand Attachment to possibly detection of vulnerable plaque. Targeting fibrin in
the Microbubble Shell. Choice of Ligands for Efficient the clot (e.g., with anti-fibrin antibodies) might also be
Targeting”). feasible [54].
Ischemia–reperfusion injury is an important target subset
of inflammatory imaging: when the blood flow to certain
tissue is transiently restricted and eventually restored, signif- Atherosclerotic Plaque Imaging: Targeting Vascular
icant inflammatory response of the affected tissue takes Inflammation Markers
place. Vascular endothelium in these tissues overexpresses
P- and E-selectin and other inflammatory markers. There- It is well established that atherosclerotic plaque develop-
fore, targeted bubbles can efficiently bind to endothelium in ment, and especially plaque vulnerability, are directly related
these areas, so that delineation of the areas of disease can be to inflammation. Markers of endothelium activation, such as
achieved [44]. Obviously, a prerequisite for the success of VCAM-1, are known to be widely expressed in the plaque
targeted microbubble contrast imaging is the existence of and may boost leukocyte accumulation in the plaque. There-
blood flow: in case of no-reflow [49], if the vasculature is fore, they may serve as receptors for microbubble targeting
severely damaged, microbubbles will not have a chance to [55]. Targeting of microbubbles to inflammatory markers
enter the affected tissue. (e.g., VCAM-1) with antibodies is therefore possible and
A “milder” case of ischemia–reperfusion injury is repre- has been demonstrated in murine models, such as apoE−/−
sented as “ischemic memory”: a transient ischemic event is mice on high-cholesterol diet [42, 56]. In this (rather acute)
resolved, so by the time the diagnosis needs to be performed, model, all the vasculature of the animal is essentially in-
perfusion is normal. However, transient ischemia still leads flamed and VCAM-1 is overexpressed systemically just
to upregulation of the inflammatory markers on the endothe- ∼2–3 months from the initiation of high-cholesterol diet, so
lium, therefore, molecular ultrasound imaging of the risk microbubble adhesion is observed in many areas, not specif-
area can be performed with microbubbles targeted to the ically in the atherosclerotic plaque lesions in aorta or carotid
activated endothelium markers [44, 50]. arteries (Fig. 6, right panel). Next step is to investigate if
Inflammation is implicated in one other condition that selective detection of plaque and especially distinguishing
may require monitoring: transplant rejection. Microbubbles active plaque will be possible by molecular ultrasound im-
might play a useful role in that setting as well: ICAM-1- aging in the setting of human atherosclerotic disease.
736 J. of Cardiovasc. Trans. Res. (2013) 6:729–739

Fig. 6 Targeted imaging of microbubbles: visualization of inflamma- targeted microbubbles is observed in the vasculature of apoE−/− mice on
tion in a murine atherosclerosis model. Left panel inflammation- a Western diet. Imaging performed 10 min after intravenous bolus of
targeted microbubbles do not accumulate in the carotid vasculature of 107 bubbles, with Siemens Sequoia apparatus (15 L8 probe, 7 MHz,
control mice (bright specular reflections on the bottom of the image CPS mode, image frame 1.5 cm tall)
represent non-tissue response). Right panel significant accumulation of

Imaging of Bacterial Biofilms tissue plasminogen activator. All these procedures can be
performed under ultrasound guidance, with ultrasound probes
Bacterial biofilms in the infective endocarditis setting are that combine low-power contrast ultrasound imaging with
mostly caused by streptococci or staphylococci. These bacteria high acoustic pressure therapeutic transducers. Alternatively,
(often resistant to antibiotic treatment) can form embedded MRI-guided focused ultrasound equipment is gaining popu-
structures with fibrin-platelet complexes and deposit on heart larity and interest, due to the low frequency (sub-megahertz)
valves. These complexes are easily accessible from the blood- ultrasound penetration capabilities and an excellent imaging
stream, therefore microbubble targeting of bacteria for targeted quality of MRI, especially in the transcranial setting, e.g., in
imaging of biofilm location, by targeting surface markers of accelerated drug delivery across blood–brain barrier [65].
bacteria, is feasible [57]. Even more useful, a combination of
microbubbles and ultrasound treatment may aid antibacterial
therapy [58]. Conclusion

Microbubble contrast agents are on a verge of wider expan-


Approaches to Image-Guided Therapy in the Vascular sion into the cardiovascular ultrasound imaging arena. Initial
Setting successful approvals for endocardial border delineation and
left ventricle opacification took place more than a decade ago
In the presence of microbubbles the acoustic energy brought in the USA and Europe. However, those accomplishments
by the ultrasound waves can be deposited locally in the were not yet followed by more complex milestones, such as
immediate proximity to the microbubble. This energy deposi- myocardial perfusion indication approval. In Europe, China,
tion can be applied for a wide range of image-guided inter- Canada, and Japan contrast ultrasound imaging is quite pop-
ventions, from assisting transfection [59–61] to siRNA deliv- ular and approved for general radiology use (mostly for liver
ery [62] to removal of bacterial biofilms on the surface of imaging). In the USA no such clinical indication is approved
implanted devices [58] to accelerated thrombolysis [63]. The yet. Use of microbubble contrast targeted towards the mo-
latter approach has already reached clinical trial stage [64], lecular markers of disease will expand the diagnostic capa-
where it can reduce the dose of a thrombolytic agent needed bilities of ultrasound to the area of molecular imaging; this
for stroke therapy, or, in some instances, provide clot dissolu- will be especially useful in the urgent diagnostics cardiovascu-
tion without the administration of an exceptionally expensive lar environment and beyond, as well as in image-guided biopsy
thrombolytic enzyme. Ultrasound-assisted thrombolysis could and therapy setting. Focused ultrasound in combination with
minimize the risk of hemorrhage associated with the current microbubble agents might also find applications in targeted
clot dissolution therapy that requires a significant dose of drug and gene delivery.
J. of Cardiovasc. Trans. Res. (2013) 6:729–739 737

Acknowledgment The author is grateful to numerous colleagues and myocardial perfusion using intravenous myocardial contrast echo-
collaborators, students, and fellows, at UVA and collaborating institu- cardiography in healthy volunteers: comparison with positron emis-
tions, especially to Jonathan Lindner, Klaus Ley, and Joshua Rychak. sion tomography. Journal of the American Society of Echocardiog-
Support by NIH (R21/33CA102880), earlier R01EB002185, SBIR raphy, 19(3), 285–293.
subcontract via R43/44 EB007857, and a research grant from Philips 16. Wei, K., Jayaweera, A. R., Firoozan, S., Linka, A., Skyba, D. M., &
Research are gratefully acknowledged. Kaul, S. (1998). Quantification of myocardial blood flow with
ultrasound-induced destruction of microbubbles administered as a
constant venous infusion. Circulation, 97(5), 473–483.
17. Senior, R., Monaghan, M., Main, M. L., Zamorano, J. L., Tiemann,
References K., Agati, L., et al. (2009). Detection of coronary artery disease
with perfusion stress echocardiography using a novel ultrasound
imaging agent: two Phase 3 international trials in comparison with
1. Gramiak, R., & Shah, P. M. (1968). Echocardiography of the aortic radionuclide perfusion imaging. European Journal of Echocardi-
root. Investigative Radiology, 3(5), 356–366. ography, 10(1), 26–35.
2. Gramiak, R., & Shah, P. M. (1971). Detection of intracardiac blood 18. Wilson, S. R., Jang, H. J., Kim, T. K., Iijima, H., Kamiyama, N., &
flow by pulsed echo-ranging ultrasound. Radiology, 100(2), 415– Burns, P. N. (2008). Real-time temporal maximum-intensity-
418. projection imaging of hepatic lesions with contrast-enhanced so-
3. Schurmann, R., & Schlief, R. (1994). Saccharide-based contrast nography. AJR. American Journal of Roentgenology, 190(3), 691–
agents. Characteristics and diagnostic potential. La Radiologia 695.
Medica, 87(5 Suppl 1), 15–23. 19. Kalantarinia, K., Belcik, J. T., Patrie, J. T., & Wei, K. (2009). Real-
4. Keller, M. W., Glasheen, W., & Kaul, S. (1989). Albunex: a safe time measurement of renal blood flow in healthy subjects using
and effective commercially produced agent for myocardial contrast contrast-enhanced ultrasound. American Journal of Physiology.
echocardiography. Journal of the American Society of Echocardi- Renal Physiology, 297(4), F1129–F1134.
ography, 2(1), 48–52. 20. Staub, D., Patel, M. B., Tibrewala, A., Ludden, D., Johnson, M.,
5. Skyba, D. M., Camarano, G., Goodman, N. C., Price, R. J., Skalak, Espinosa, P., et al. (2011). Vasa vasorum and plaque
T. C., & Kaul, S. (1996). Hemodynamic characteristics, myocardial neovascularization on contrast-enhanced carotid ultrasound imag-
kinetics and microvascular rheology of FS-069, a second- ing correlates with cardiovascular disease and past cardiovascular
generation echocardiographic contrast agent capable of producing events. Stroke, 41(1), 41–47.
myocardial opacification from a venous injection. Journal of the 21. Vavuranakis, M., Kakadiaris, I. A., O'Malley, S. M., Papaioannou,
American College of Cardiology, 28(5), 1292–1300. T. G., Sanidas, E. A., Naghavi, M., et al. (2008). A new method for
6. Schneider, M., Arditi, M., Barrau, M. B., Brochot, J., Broillet, A., assessment of plaque vulnerability based on vasa vasorum imaging,
Ventrone, R., et al. (1995). BR1: a new ultrasonographic contrast by using contrast-enhanced intravascular ultrasound and differen-
agent based on sulfur hexafluoride-filled microbubbles. Investigative tial image analysis. International Journal of Cardiology, 130(1),
Radiology, 30(8), 451–457. 23–29.
7. Grauer, S. E., Pantely, G. A., Xu, J., Ge, S., Giraud, G. D., Shiota, 22. Klibanov, A. L., Hughes, M. S., Marsh, J. N., Hall, C. S., Miller, J.
T., et al. (1996). Myocardial imaging with a new transpulmonary G., Wible, J. H., et al. (1997). Targeting of ultrasound contrast
lipid-fluorocarbon echo contrast agent: experimental studies in material. An in vitro feasibility study. Acta Radiologica. Supple-
pigs. American Heart Journal, 132(5), 938–945. ment, 412, 113–120.
8. Wijkstra H., Smeenge M., Rosette Jdl. , Pochon S., Tardy-Cantalupi 23. Pochon, S., Tardy, I., Bussat, P., Bettinger, T., Brochot, J., von
I., Tranquart F. (2012) Targeted microbubble prostate cancer imagign Wronski, M., et al. (2010). BR55: a lipopeptide-based VEGFR2-
with BR55. In: Cate Ft, Jong Nd, Leen E (eds) The 17th European targeted ultrasound contrast agent for molecular imaging of angio-
Symposium on Ultrasound Contrast Imaging., Rotterdam, p 6. genesis. Investigative Radiology, 45(2), 89–95.
9. Leighton, T. G. (1994). The acoustic bubble. London: Academic 24. Pelura, T. J., & Pelura, T. J. (1998). Clinical experience with
Press. AF0150 (Imagent US), a new ultrasound contrast agent. Academic
10. de Jong, N., Emmer, M., van Wamel, A., & Versluis, M. (2009). Radiology, 1(5 Suppl), S69–S71. discussion S72-64.
Ultrasonic characterization of ultrasound contrast agents. Medical 25. Sugimoto, K., Moriyasu, F., Saito, K., Taira, J., Saguchi, T.,
and Biological Engineering and Computing, 47(8), 861–873. Yoshimura, N., et al. (2012). Comparison of Kupffer-phase
11. Bouakaz, A., Versluis, M., & de Jong, N. (2005). High-speed Sonazoid-enhanced sonography and hepatobiliary-phase gadoxetic
optical observations of contrast agent destruction. Ultrasound in acid-enhanced magnetic resonance imaging of hepatocellular car-
Medicine and Biology, 31(3), 391–399. cinoma and correlation with histologic grading. Journal of Ultra-
12. Phillips, P., & Gardner, E. (2004). Contrast-agent detection and sound in Medicine, 31(4), 529–538.
quantification. European Radiology, 14(Suppl 8), P4–P10. 26. Wible, J. H., Jr., Wojdyla, J. K., Bales, G. L., McMullen, W. N.,
13. Klibanov, A. L., Rasche, P. T., Hughes, M. S., Wojdyla, J. K., Geiser, E. A., & Buss, D. D. (1996). Inhaled gases affect the
Galen, K. P., Wible, J. H., Jr., et al. (2004). Detection of individual ultrasound contrast produced by Albunex in anesthetized dogs.
microbubbles of ultrasound contrast agents: imaging of free- Journal of the American Society of Echocardiography, 9(4), 442–
floating and targeted bubbles. Investigative Radiology, 39(3), 451.
187–195. 27. Klibanov, A. L., Hughes, M. S., Villanueva, F. S., Jankowski, R. J.,
14. Senior, R., Lepper, W., Pasquet, A., Chung, G., Hoffman, R., Wagner, W. R., Wojdyla, J. K., et al. (1999). Targeting and ultra-
Vanoverschelde, J. L., et al. (2004). Myocardial perfusion assess- sound imaging of microbubble-based contrast agents. Magma,
ment in patients with medium probability of coronary artery disease 8(3), 177–184.
and no prior myocardial infarction: comparison of myocardial 28. Takalkar, A. M., Klibanov, A. L., Rychak, J. J., Lindner, J. R., &
contrast echocardiography with 99mTc single-photon emission Ley, K. (2004). Binding and detachment dynamics of microbubbles
computed tomography. American Heart Journal, 147(6), 1100– targeted to P-selectin under controlled shear flow. Journal of Con-
1105. trolled Release, 96(3), 473–482.
15. Dijkmans, P. A., Knaapen, P., Sieswerda, G. T., Aiazian, E., Visser, 29. Klibanov, A. L., Rychak, J. J., Yang, W. C., Alikhani, S., Li, B.,
C. A., Lammertsma, A. A., et al. (2006). Quantification of Acton, S., et al. (2006). Targeted ultrasound contrast agent for
738 J. of Cardiovasc. Trans. Res. (2013) 6:729–739

molecular imaging of inflammation in high-shear flow. Contrast characterization of targeted ultrasound microbubbles for diagnostic
Media & Molecular Imaging, 1(6), 259–266. use. Ultrasound in Medicine and Biology, 37(1), 136–150.
30. Guenther, F., von zur Muhlen, C., Ferrante, E. A., Grundmann, S., 46. Hust, M., Frenzel, A., Meyer, T., Schirrmann, T., & Dubel, S.
Bode, C., & Klibanov, A. L. (2010). An ultrasound contrast agent (2012). Construction of human naive antibody gene libraries.
targeted to P-selectin detects activated platelets at supra-arterial Methods in Molecular Biology, 907, 85–107.
shear flow conditions. Investigative Radiology, 45(10), 586–591. 47. Hernot, S., Unnikrishnan, S., Du, Z., Shevchenko, T., Cosyns, B.,
31. Rychak, J. J., Li, B., Acton, S. T., Leppanen, A., Cummings, R. D., Broisat, A., et al. (2012). Nanobody-coupled microbubbles as
Ley, K., et al. (2006). Selectin ligands promote ultrasound contrast novel molecular tracer. Journal of Controlled Release, 158(2),
agent adhesion under shear flow. Molecular Pharmaceutics, 3(5), 346–353.
516–524. 48. Feldwisch, J., & Tolmachev, V. (2012). Engineering of affibody
32. Kim, D. H., Klibanov, A. L., & Needham, D. (2000). The influence molecules for therapy and diagnostics. Methods in Molecular Biol-
of tiered layers of surface-grafted poly(ethylene glycol) on recep- ogy, 899, 103–126.
tor–ligand-mediated adhesion between phospholipid monolayer- 49. Hayat, S. A., & Senior, R. (2008). Myocardial contrast echocardi-
stabilized microbubbles and coated glass beads. Langmuir, 16(6), ography in ST elevation myocardial infarction: ready for prime
2808–2817. time? European Heart Journal, 29(3), 299–314.
33. Ham, A. S., Klibanov, A. L., & Lawrence, M. B. (2009). Action at a 50. Villanueva, F. S., Lu, E., Bowry, S., Kilic, S., Tom, E., Wang, J.,
distance: lengthening adhesion bonds with poly(ethylene glycol) et al. (2007). Myocardial ischemic memory imaging with molecular
spacers enhances mechanically stressed affinity for improved vas- echocardiography. Circulation, 115(3), 345–352.
cular targeting of microparticles. Langmuir, 25(17), 10038–10044. 51. Weller, G. E., Lu, E., Csikari, M. M., Klibanov, A. L., Fischer, D.,
34. Rychak, J. J., Lindner, J. R., Ley, K., & Klibanov, A. L. (2006). Wagner, W. R., et al. (2003). Ultrasound imaging of acute cardiac
Deformable gas-filled microbubbles targeted to P-selectin. Journal transplant rejection with microbubbles targeted to intercellular ad-
of Controlled Release, 114(3), 288–299. hesion molecule-1. Circulation, 108(2), 218–224.
35. Ferrante, E. A., Pickard, J. E., Rychak, J., Klibanov, A., & Ley, K. 52. Wang, X., Hagemeyer, C. E., Hohmann, J. D., Leitner, E., Armstrong,
(2009). Dual targeting improves microbubble contrast agent adhe- P. C., Jia, F., et al. (2012). Novel single-chain antibody-targeted
sion to VCAM-1 and P-selectin under flow. Journal of Controlled microbubbles for molecular ultrasound imaging of thrombosis: vali-
Release, 140(2), 100–107. dation of a unique noninvasive method for rapid and sensitive detec-
36. Maul, T. M., Dudgeon, D. D., Beste, M. T., Hammer, D. A., Lazo, J. tion of thrombi and monitoring of success or failure of thrombolysis in
S., Villanueva, F. S., et al. (2010). Optimization of ultrasound mice. Circulation, 125(25), 3117–3126.
contrast agents with computational models to improve selection 53. Unger, E. C., McCreery, T. P., Sweitzer, R. H., Shen, D., & Wu, G.
of ligands and binding strength. Biotechnology and Bioengineer- (1998). In vitro studies of a new thrombus-specific ultrasound
ing, 107(5), 854–864. contrast agent. The American Journal of Cardiology, 81(12A),
37. Fadok, V. A., de Cathelineau, A., Daleke, D. L., Henson, P. M., & 58G–61G.
Bratton, D. L. (2001). Loss of phospholipid asymmetry and surface 54. Korosoglou, G., Behrens, S., Bekeredjian, R., Hardt, S.,
exposure of phosphatidylserine is required for phagocytosis of Hagenmueller, M., Dinjus, E., et al. (2006). The potential of a
apoptotic cells by macrophages and fibroblasts. The Journal of new stable ultrasound contrast agent for site-specific targeting. An
Biological Chemistry, 276(2), 1071–1077. in vitro experiment. Ultrasound Medicine Biology, 32(10), 1473–
38. Yanagisawa, K., Moriyasu, F., Miyahara, T., Yuki, M., & Iijima, H. 1478.
(2007). Phagocytosis of ultrasound contrast agent microbubbles by 55. Lindner, J. R. (2002). Detection of inflamed plaques with contrast
Kupffer cells. Ultrasound in Medicine and Biology, 33(2), 318– ultrasound. The American Journal of Cardiology, 90(10C), 32L–
325. 35L.
39. Lindner, J. R., Song, J., Xu, F., Klibanov, A. L., Singbartl, K., Ley, 56. Wu, J., Leong-Poi, H., Bin, J., Yang, L., Liao, Y., Liu, Y., et al.
K., et al. (2000). Noninvasive ultrasound imaging of inflammation (2011). Efficacy of contrast-enhanced US and magnetic
using microbubbles targeted to activated leukocytes. Circulation, microbubbles targeted to vascular cell adhesion molecule-1 for
102(22), 2745–2750. molecular imaging of atherosclerosis. Radiology, 260(2), 463–471.
40. Christiansen, J. P., Leong-Poi, H., Klibanov, A. L., Kaul, S., & 57. Anastasianides P., Mojica K., Matter M. L., Allen J. S. (http://www.
Lindner, J. R. (2002). Noninvasive imaging of myocardial reperfu- acoustics.org/press/157th/anastasiadis.html) Targeted ultrasound
sion injury using leukocyte-targeted contrast echocardiography. contrast agents for the imaging of biofilm infections. Accessed
Circulation, 105(15), 1764–1767. 2013.05.26
41. Villanueva, F. S., Jankowski, R. J., Klibanov, S., Pina, M. L., Alber, 58. Dong, Y., Chen, S., Wang, Z., Peng, N., & Yu, J. (2013). Synergy of
S. M., Watkins, S. C., et al. (1998). Microbubbles targeted to ultrasound microbubbles and vancomycin against Staphylococcus
intercellular adhesion molecule-1 bind to activated coronary artery epidermidis biofilm. The Journal of Antimicrobial Chemotherapy,
endothelial cells. Circulation, 98(1), 1–5. 68(4), 816–826.
42. Kaufmann, B. A., Sanders, J. M., Davis, C., Xie, A., Aldred, P., 59. Christiansen, J. P., French, B. A., Klibanov, A. L., Kaul, S., &
Sarembock, I. J., et al. (2007). Molecular imaging of inflammation Lindner, J. R. (2003). Targeted tissue transfection with ultrasound
in atherosclerosis with targeted ultrasound detection of vascular cell destruction of plasmid-bearing cationic microbubbles. Ultrasound
adhesion molecule-1. Circulation, 116(3), 276–284. in Medicine and Biology, 29(12), 1759–1767.
43. Lindner, J. R., Song, J., Christiansen, J., Klibanov, A. L., Xu, F., & 60. Leong-Poi, H., Kuliszewski, M. A., Lekas, M., Sibbald, M.,
Ley, K. (2001). Ultrasound assessment of inflammation and renal Teichert-Kuliszewska, K., Klibanov, A. L., et al. (2007). Therapeu-
tissue injury with microbubbles targeted to P-selectin. Circulation, tic arteriogenesis by ultrasound-mediated VEGF165 plasmid gene
104(17), 2107–2112. delivery to chronically ischemic skeletal muscle. Circulation Re-
44. Davidson, B. P., Kaufmann, B. A., Belcik, J. T., Xie, A., Qi, Y., & search, 101(3), 295–303.
Lindner, J. R. (2012). Detection of antecedent myocardial ischemia 61. Vannan, M., McCreery, T., Li, P., Han, Z., Unger, E., Kuersten, B.,
with multiselectin molecular imaging. Journal of the American et al. (2002). Ultrasound-mediated transfection of canine myocar-
College of Cardiology, 60(17), 1690–1697. dium by intravenous administration of cationic microbubble-linked
45. Myrset, A. H., Fjerdingstad, H. B., Bendiksen, R., Arbo, B. E., plasmid DNA. Journal of the American Society of Echocardiography,
Bjerke, R. M., Johansen, J. H., et al. (2011). Design and 15(3), 214–218.
J. of Cardiovasc. Trans. Res. (2013) 6:729–739 739

62. Carson, A. R., McTiernan, C. F., Lavery, L., Grata, M., Leng, X., 64. Alexandrov, A. V. (2006). Ultrasound enhanced thrombolysis for
Wang, J., et al. (2012). Ultrasound-targeted microbubble destruc- stroke. International Journal of Stroke, 1(1), 26–29.
tion to deliver siRNA cancer therapy. Cancer Research, 72(23), 65. Hynynen, K., McDannold, N., Vykhodtseva, N., Raymond, S.,
6191–6199. Weissleder, R., Jolesz, F. A., et al. (2006). Focal disruption of the
63. Wu, Y., Unger, E. C., McCreery, T. P., Sweitzer, R. H., Shen, D., blood–brain barrier due to 260-kHz ultrasound bursts: a method for
Wu, G., et al. (1998). Binding and lysing of blood clots using molecular imaging and targeted drug delivery. Journal of Neuro-
MRX-408. Investigative Radiology, 33(12), 880–885. surgery, 105(3), 445–454.

You might also like