Böhmer et al 2009

Embed Size (px)

Citation preview

  • 8/7/2019 Bhmer et al 2009

    1/12

    European Journal of Radiology 70 (2009) 242253

    Contents lists available at ScienceDirect

    European Journal of Radiology

    j o u r n a l h o m e p a g e : w w w . e l s e v i e r . c o m / l o c a t e / e j r a d

    Ultrasound triggered image-guided drug delivery

    Marcel R. Bhmer 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 Netherlandsb Cardiovascular Division, Department of Medicine, Cobb Hall, University of Virginia School of Medicine, Hospital Drive, Cobb Hall RM 1026, Charlottesville, VA 22908-158, USAc Department of Cardiology and Angiology, University Hospital Mnster, Albert Schweitzerstrasse 33, 48149 Mnster, Germanyd 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

    Article history:

    Received 13 January 2009

    Accepted 14 January 2009

    Keywords:

    Ultrasound

    Image guidance

    MRI

    Microbubbles

    Temperature sensitive liposome

    a b s t r a c t

    The integration of therapeutic interventions with diagnostic imaging has been recognized as one of

    the next technological developments that will have a major impact on medical treatments. Important

    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

    depth understanding of the mechanisms of the cellular and molecular basis of diseases. Recent research

    demonstrates that both pressure sensitive and temperature sensitive delivery systems hold promise for

    local treatment. The use of ultrasound for the delivery of drugs has been demonstrated in particular

    the field of cardiology and oncology for a variety of therapeutics ranging from small drug molecules to

    biologics and nucleic acids.

    2008 Elsevier Ireland Ltd. All rights reserved.

    1. Introduction

    Theroleof medical imaging technologies in medical care is shift-

    ing from a tool for diagnosis of a disease to being an integral part

    of therapeutic interventions such as in image-guided treatments.

    Stereotactic systems use images obtained before surgery, e.g., MR

    and CT, foraccurateguidanceof a surgical toolto the target anatomy.

    Instead of tissue removal, one can use high intensity focused ultra-

    sound (HIFU) as a surgery tool. Using HIFU, energy can be focused

    precisely to a small volume of interest. HIFU allows ablation of

    tissue by local administration of thermal dosages. Image-guided

    therapy offers the potential to direct therapeutic action precisely to

    the point in the tissue where it is needed and not to other tissues.

    When this is possible, a high and local thermal dose can be admin-

    istered. Image-guided delivery using HIFU requires the integration

    of imaging for diagnosis andtreatment planning anda therapy that

    can be accurately directed and controlled by simultaneous imageguidance, resulting in less side effects.

    The use of ultrasound for local hyperthermia was recognized

    early as reviewed by Moyer and Clement [1,2]. Direct exposure

    to therapeutic ultrasound produces irreversible cell death through

    coagulative necrosis, and is currently being clinically evaluated in

    breast, kidney, and liver tumors [3]. There is an increasing level of

    literatureevidence [39] that demonstrateshow ultrasound energy

    Corresponding author.

    E-mail address: [email protected](M.R. Bhmer).

    can also be used non-destructively for increasing the efficacy for

    delivery of drugs and genetic material. Especially for chemothera-

    peutic regimens to be successful in cancer treatment, the particular

    drug must be effective in the tumor environment and administered

    in doses that cause tumor eradication while keeping severe side

    effects within acceptable limits, commonly called the therapeutic

    window.

    Performing minimally invasive therapy, such as ultrasound

    mediated drug delivery (USDD), under image guidance requires

    adequate definition of the region of interest and accurate compen-

    sation for motion. Especially in the heart the feedback provided is

    necessary to target the therapy accurately.The region of interestcan

    be identified by detection of an abnormal morphology. Molecular

    imaging holds promise to apply minimally invasive therapy in an

    early stage of a disease as malignancies can be detected in an early

    stage. Molecular imaging uses targeted contrast agents, which are

    agents decoratedwith, for instance,antibodies or fragmentsthereofthat specifically interact with specific markers such as endothelial

    markers of inflammation or angiogenesis.

    New methods in ultrasound and magnetic resonance (MR)

    provide higher resolution information in two and three spatial

    dimensions, with acquisition and display occurring nearly in real

    time. Computer image processing methods offer ways of clarifying,

    highlighting, or detecting specific regions in tissue. Developments

    in MR thermometryprovidea technical solution tofollow thedeliv-

    ery of a thermal dose to a lesion. Fortreatment, a volumeof interest

    inside a patient is delineated based on MR imaging, and subse-

    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

    http://www.sciencedirect.com/science/journal/0720048Xhttp://www.elsevier.com/locate/ejradmailto:[email protected]://dx.doi.org/10.1016/j.ejrad.2009.01.051http://dx.doi.org/10.1016/j.ejrad.2009.01.051mailto:[email protected]://www.elsevier.com/locate/ejradhttp://www.sciencedirect.com/science/journal/0720048X
  • 8/7/2019 Bhmer et al 2009

    2/12

    M.R. Bhmer et al. / European Journal of Radiology 70 (2009) 242253 243

    mapped by MR thermometry and fed back into the control of the

    ultrasound transducer to provide full temporal and spatial control

    of the heating [1012]. Therefore, the technique gives more than a

    feedback on the location of the region of interest, it also provides

    information on a physical parameter, which can be used to guide

    and control the therapy [13,14]. MRI can also be used to monitor

    changes in the permeability of the vasculature, as shown by Treat

    et al. [15]

    Contrast ultrasound imaging, using microbubbles, also pro-

    vides useful information for image-guided drug delivery. For these

    agents, optimized detection algorithms are available on ultrasound

    diagnostic imaging systems. With respect to therapy transducer

    design, developments in electronic steering of the beam improve

    the size of focal region and reduce grating lobes while maintaining

    a small number of elements and a compact size.

    In this review we focus on the use of ultrasound for therapy

    and provide examples in cardiology and oncology. We will review

    drug delivery vehicles based on temperature and pressure sensitive

    systems. Such systems are either modified slow release systems

    with a temperature sensitive component or contrast agents that

    have been modified to include or enhance drug delivery. Develop-

    ments in ultrasound and MRI imaging, and new agents to follow

    and quantify drug release, will be described.

    2. Ultrasound mediated drug delivery systemsequipment

    design considerations

    The equipment for ultrasound mediated delivery varies widely

    dependent on the application and often on the clinical availabil-

    ity of ultrasound imaging or therapy devices. The following section

    will describe the components parts of ultrasound therapy devices

    for drug delivery with a discussion on the relevant importance and

    design limitations. We will then follow with a discussion of spe-

    cific application requirements that depend on the target volume

    within the subject. As diagnostic imaging systems are not designed

    for therapy, which is in particular reflected in the focusing of the

    ultrasound beam, we will not consider these studies here.

    2.1. Signal excitation

    The two primary physical mechanisms for activating particle

    based therapies can be separated into heat- and pressure-activated

    particles. In the case of heat-activated particles, typically the

    goal is to deposit enough acoustic energy into the targeted vol-

    ume to raise the temperature in order to release an encapsulated

    drug. In these cases, the duty cycle of the acoustic signal is of

    paramount importance and so the electrical excitation and ampli-

    fication will consist of pure tone (long pulse lengths) signal sources

    and high power amplifiers (class D op-amps are popular). In the

    case of pressure-mediated release, the acoustic signal is often of

    shorter duration and lower overall energy deposition than thatused in heat-release applications. Pressure based release uses par-

    ticles, mostly microbubbles, which deposit or release a drug when

    encountering a peak negative pressure beyond a particular thresh-

    old (usually) between 0.5 and 5 MPa peak negative pressure. These

    signals are quite short in duration but high in acoustic pressure. A

    typical approach is to transmita short (

  • 8/7/2019 Bhmer et al 2009

    3/12

    244 M.R. Bhmer et al. / European Journal of Radiology 70 (2009) 242253

    of the organs, and amount of peripheral blood flow for heat con-

    vection among other factors are not simple linear scale factors. For

    example, thedepth of thetreatment zone will determinethe choice

    of ultrasonic frequency which canlead toinadvertentheating 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

    be addressed. In many applications this is appropriate and cost-

    effective, but it is not fast. Electronic steering is accomplished by

    dividing a therapy transducer into multiple, individually address-

    able parts. The electronic signalapplied to each element is retardedby a phase shift in such a way to control the location in the tissue

    wherethe acoustic waves coherentlyinterfere. Such steering allows

    for rapid (on order of milliseconds) changes in treatment location.

    These arrays havebeen used indiagnostic imaging andareknown as

    theclassof phasedarrays. In therapyapplications, theuse of phased

    arrays has been more limited because of technical challenges as

    mentioned in the following sections.

    Several issuesmust be addressed when using arrays of elements

    in therapeutic drug delivery. In particular, the size of the array can

    often be large because of the needed pressures or acoustic energy.

    As a result, when dividing into multiple elements, the element

    count can be quite largeoften into the thousands in order to avoid

    effects such as grating lobes. Grating lobes occur because of the

    inadvertentphasecoherence occurringin theacousticfield in unin-

    tended areas. This complication has implications for drug delivery

    especially in cases where exposure or release of a drug in a sensitive

    organ may lead to undesired side effects. Clever approaches have

    been suggested to avoid these grating lobes without requiring a

    large number of elements,includingthe use of sparse arrays,irregu-

    larshapedand spaced elements,and limiting the number of phases

    to be applied to the elements to simplify the driving electronics.

    3. Pressure-mediated delivery

    3.1. Ultrasound contrast agents

    Microbubbles used as ultrasound contrast agents are tiny gas

    bubbles, small enough to pass the lung capillary bed. To preventdissolution of the gas they have a shell made from a lipid, a pro-

    tein or a biodegradable polymer. Lipid-shelled microbubbles are

    used in clinical practice and have a monolayer of phospholipid.

    An albumin-shelled agent, Optison, is also clinically available and

    polymer-shelled agents have reached the end of phase III clini-

    cal trials. Microbubbles are used for left ventricle opacification;

    the ultrasound contrast between the blood in the left ventricle

    and the myocardium is low and can be increased significantly by

    intravenous injection of a small number of microbubbles. Typically

    108109 microbubbles are injected for a diagnosticultrasound scan.

    Microbubbles can be used to improve endocardial border delin-

    eation and, thereby wallmotion abnormalities. In addition it allows

    for analysis of myocardial perfusion, which further helps to iden-

    tify the myocardiumat risk. Perfusion cannotonly be applied to the

    Fig. 1. TEM picture of 2m (polylactide)-shelled microbubbles prepared by emul-

    sification and freeze-drying technology.

    myocardium but also to other tissues. As noted by Cosgrove [19] andSchneider [20], microbubbles can be used for dynamic detection of

    macro and microvascular flow in many organs. Microbubbles are

    also used to study the blood supply to the liver. Primary hepatocel-

    lular carcinomas (HCC) are supplied by the hepatic artery. After a

    bolus injection of ultrasound contrastagent, these lesions arehigh-

    lighted by the perfusionof contrastagentbefore the rest of the liver

    is fully perfused. Contrast liver imaging has been the subject of a

    multi-center study and described by Lencioni et al. [21]

    3.2. Thin-shelled and hard-shelled microbubbles

    Microbubble agents can be classified as soft- or thin-shelled

    and hard-shelled agents. Ultrasound contrast agents do not only

    scatter ultrasound efficiently, they also react to low energy ultra-sound by emitting specific frequencies. Thin-shelled agents are

    microbubbles having a lipid monolayer with a thickness of about

    23nm. They undergo volume expansions and contractions that

    generate an acoustic signal [22], of which non-linear components

    give the most specific information for imaging [23,24]. As the shell

    of these microbubbles is so thin, fluorinated gases are needed to

    keep the microbubble stable for a sufficient time in the circula-

    tion. Hard-shelled microbubbles have typical shell thicknesses in

    the range of 20100 nm.An example of polymer-shelled microbub-

    bles is given in Fig. 1. They hardly show volume expansions at low

    acoustic pressure [25,26]. Nevertheless some of these agents do

    generate acoustic signals as well without losing gas. Studies using

    an extremely fast camera [27] have given first indications that they

    often indent like a badly inflated ball, which is a way to conservetheir surface area and allow for a change in the volume [28,29]. At

    higherpressures the microbubbles are destroyed showing dramatic

    changes in the gas volume as shown in Fig. 2, where the activa-

    tion of a polymer-shelled microbubble is given. Polymer-shelled

    microbubbles do not need very hydrophobic gases to be stable in

    circulation.

    3.3. Targeted microbubbles

    The use of microbubbles is currently being extended to targeted

    imaging and drug delivery applications. For molecular imaging

    applications, the shell is coated with specific ligands. A typical

    example is the targetingof endothelial markers of angiogenesis and

    inflammation [30,31].

  • 8/7/2019 Bhmer et al 2009

    4/12

    M.R. Bhmer et al. / European Journal of Radiology 70 (2009) 242253 245

    Fig. 2. Series of events upon insonation of polymer-shelled microcapsule, frames 1416, 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 phaseof research,targeting ligands

    are conveniently bound to the shell via biotinstreptavidinbiotin

    bridges, see Fig. 3. Biotinylated lipids and biotinylated biodegrad-

    able polymers [32] used for microbubble shell preparation are

    available or can be synthesized. Microbubbles targeted to vascu-

    lar endothelial growth factors and selectins have shown strong

    enhanced ultrasound images in the areas of upregulation of these

    markers in the vasculature [33].

    Direct coupling of targeting ligands to the microbubble shell

    using peptide bond formation chemistry [34], see Fig. 3, is nec-

    essary at the next step towards clinical trials, when the presence

    of non-endogenous proteins, such as avidin or streptavidin, is not

    desired.

    Initial model system studies [35] showed that biotinylatedmicrobubbles can be targeted to avidin-coated surface in vitro,

    and ultrasound imaging of these targeted bubbles was success-

    ful. Most of the experimental targeted ultrasound imaging efforts

    have been focused on the various in vivo animal models, from

    thrombus targeting [36] to the ultrasound imaging of a variety of

    molecules upregulated on vascular endothelium. Anti-P-selectin-

    Fig.3. Inclusionof drugs in lipid-shelled microbubbles. (A)Lipid-shelledmicrobub-

    bleconsistingof gas encapsulated bya lipid monolayer,a fractionof thelipidcan be

    pegylated (not shown). (B) Lipid-shelled microbubble with an additional oil-phase

    to increase the reservoir size to incorporate hydrophilic drugs {Unger, 1998 #122}.

    (C) Lipid-shelled decorated with liposomes via biotin streptavidin bridges.

    antibody-carrying microbubbles have been successfully used for

    targeted ultrasound contrast imaging in the areas of TNF-induced

    inflammation or ischemia-reperfusion injury [37]. Detection of

    ICAM-1 upregulation in transplant rejectionmodel was achieved by

    targeting microbubbles with biotinylated anti-ICAM-1 antibodies

    [38]. A large set of studies was conducted at imaging angio-

    genic endothelium, via biotinylated antibodies against v3 in thetumor vasculature setting [39] as well as therapeutic angiogene-

    sis [40]. Tumor vasculature status can be evaluated by targeting

    streptavidin-carrying lipid microbubbles decorated with biotiny-

    lated antibodies against VEGF receptor 2 [41]. The ease of use of

    ultrasound imaging allows comparative targeted imaging of two

    markers in the tumor vasculature of the same animals, for instance

    endoglin versus VEGF receptor [33].At this time, covalent coupling methods, lacking avidinbiotin

    scheme, aregaining wider acceptance, showing the successful cou-

    pling of small ligands [42] or antibodies [43] with good yield. This

    covalent approach will be more applicable in the clinic.

    Binding of antibodiesto the vascular endothelium targets is very

    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-

    mentof microbubblesto the target is aidedby firmantibodybinding

    [46]. An alternative is to increase the ligand concentration on the

    microbubble surface, e.g., by using polymeric version of sialyl Lewis

    X, which is available commercially, polymeric sialyl Lewis X, is

    capable of firmbut rapid cooperative multipointbindingwith the P-

    selectin target surface, and providesefficientmicrobubble targeting

    [47].

  • 8/7/2019 Bhmer et al 2009

    5/12

    246 M.R. Bhmer et al. / European Journal of Radiology 70 (2009) 242253

    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 emulsionmethod [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

    For ultrasound induced drug delivery based on microbubbles

    two approaches are distinguished, see for instance the review by

    Hernot and Klibanov [5] where the distinction is made between

    co-administration of drugs and microbubbles and drug-loading of

    the microbubbles themselves. In the case of co-administration the

    function of the microbubbles is to enhance the permeability of

    the endothelial wall. This can either be affected by rupture of the

    endothelial wall, leading to extravasation of relatively large enti-

    ties such as red blood cells and polymer particles [48] or in a more

    subtle way, at lower pressures, by microbubbles causing temporaryopening of cell membranes.Many of the properties of cell mem-

    branes are shear dependent. Marmottant and Hilgenfeldt [49,50]

    demonstrated that the oscillations of microbubbles induce local

    shear stress by altering the flow of liquid near the cell surface. Van

    Wamelet al.[51,52] haveshown the deformation of cell membranes

    in the presence of an oscillating microbubble directlyusing an ultra-

    fast camera. Sonoporation is the term that is used to describe the

    formation of pores by ultrasound. If the pores are too large they

    cannot reseal leading to cell death, however, if the pores can seal

    again they will stay open for a time and, in principle allow passage

    of therapeutics, such as radionuclides [53] or plasmid DNA (see ref.

    [8] for an overview). Mehier-Humbert et al. [54] investigated the

    percentage of GFP positive Matt-B III cells following plasmid DNA

    delivery using lipid- and polymer-shelled microbubbles.A great challenge in sonoporation is to open the blood brain

    barrier in a reversible way. Treat et al. [15] and Hynynen et al. [55]

    have shown that pores made in a rabbit brain close again in about

    6 h. They also demonstrated that contrast enhanced MRI is a very

    suitable method to follow sonoporation. The method has been used

    to deliver doxorubicin across the blood brain barrier in rats, as well

    as to deliver genes under MRI guidance.

    As stressed in this paragraph the combination of microbubbles

    and ultrasound have an effect on the properties of adjacent cells. A

    number of authors have found that this effect extends to tumor

    growth. For instance Miller and Song [56] reported that tumor

    growth of renal carcinoma in mice in the presence of Optison and

    ultrasound is reduced. Damaging tumors by ultrasound could have

    an effect on metastasis because of the disintegration of the tumor.

    Miller and Dou [57] investigated the enhancement of lung metas-

    tasis from an implanted mouse melanoma tumor after application

    of ultrasound in the presence of microbubbles. At high pressure

    (5MPa) and a 1 Hzrate toavoidheating morelungmetastases were

    indeed found in the presence of microbubble than in their absence.

    However at lower pressure (2 MPa) no enhancement was found. In

    the absence of microbubbles an elevated level of metastasis was

    already found in at a peak negative pressure of 5 MPa.

    3.5. Incorporation of drugs and genetic material in microbubbles

    Instead of co-injecting drugs and microbubbles, microbubblescan also be modified to contain drugs [58,59] or DNA [6062]. The

    advantage, as explicitly shown by Lentacker et al. [61] in an in

    vitro setting is that the therapeutic molecule is close to where the

    acoustic action is, and therefore opens the opportunity for a dose

    reduction while maintaining its therapeutic efficacy.

    If drugs have sufficient affinity for the lipid monolayer they can,

    in principle, be incorporated directly in lipid-shelled microbub-

    bles (Fig. 4A). However, as the monolayer is very thin, the amount

    that can be incorporated is extremely low. Unger [59] has added

    an additional oil, triacetin, to make a thicker hydrophobic layer

    to increase the incorporation of paclitaxel, as shown schemati-

    cally in Fig. 4B. This approach is limited to hydrophobic drugs; a

    more general applicable route is to attach liposomes or lipoplexes

    to the microbubble (Fig. 4C). Another option is to bind the drugto the outside, for instance in a form associated with lipids, such

    a drug carrying liposomes or lipoplexes: positively charged lipid

    complexed to negatively charged nucleic acid [61]. They have

    demonstrated high transfection efficiency by showing luciferase

    activity in vitro. Attachment of liposomes can allow the incorpo-

    ration of both hydrophilic molecules into the aqueous core of the

    liposome or hydrophobic drugs into the lipid bilayers. Finally mul-

    tilayer technology [63] can be used to deposit large therapeutic

    molecules layer by layer on a microbubble surface.

    In principle layer-by-layer deposition or attachment of lipo-

    somes is also possible for polymer-shelled agents, see Fig. 5.

    However, different routes are also available to prepare drug loaded

    polymer-shelled agents. Emulsification of a polymer solution con-

    taining a carrier solvent and an alkane in an aqueous phase is a

  • 8/7/2019 Bhmer et al 2009

    6/12

    M.R. Bhmer et al. / European Journal of Radiology 70 (2009) 242253 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

    is removed from the emulsion droplets and the polymer phase

    separates from the shell closing in the alkane in the center. The

    alkane is subsequently removed by freeze-drying. This prepara-

    tion route is suitable for hydrophobic drugs. Fabrication techniques

    starting from monodispersed emulsions, such as a technique based

    on submerged inkjet-printing [64] allow for a precise incorpora-

    tion of a known amount of drug per particle and therefore improve

    the control over the doseage. Direct emulsification techniques can

    also yield a narrow size distribution with a maximum between 1

    and 2m and >95% of the microbubbles smaller than 3m [28].Hydrophilic drugs cannot be incorporated directly in an emulsi-

    fication procedure. One possibility is to employ double emulsiontechnology. In a double emulsion, an aqueous phase, containing

    the drug, is first emulsified in an organic phase (containing the

    shell-forming polymer), and this first emulsion is subsequently

    emulsified in a second aqueous phase. Poly-lactide-co glycolide

    microbubblescontainingplasmidDNA have beenpreparedthis way

    [60]. Gene delivery from these microbubbles was shown in rats and

    also an effect on tumor growth was demonstrated.

    An alternative way to prepare polymer-shelled microbubbles is

    spray-drying [65] Hydrophobic drugs can in principle be included

    in the spray. In the preparation method of Palmowski [66,67], the

    starting point is not the polymer but a monomer. As the polymer-

    ization reaction leads to the shell formation directly, incorporation

    of a drug at this stage is difficult.

    Recentdelivery experiments with a new, emerging class of genetherapeutics, small interfering RNA (siRNA), show promise to over-

    come the inherent in vivo delivery obstacles of nucleic acids in

    general and siRNA in particular, such as rapid excretion via the

    liver, serum instability, non-specific distribution, tissue and cell

    barricades [68]. The major limitation for the use of siRNA, both in

    vitro and in vivo, is the inability of naked siRNA to passively diffuse

    through cellular membranes dueto thestrong anionic chargeof the

    phosphate backbone and consequent electrostatic repulsion from

    the anionic cellmembranesurface.To deliversiRNA withmicrobub-

    bles, siRNA was either directly attached to the microbubble surface

    or simply mixed withmicrobubblesprior toadministration.In some

    studies these vehicles showed enhanced transfection efficiency

    both in vitro and in vivo. Further they provide a better protection

    against degradation by serum nucleases. [60,69,70]

    3.6. Targeted imaging and drug delivery with microbubbles

    Microbubbles typically do not exhibit long circulation times.

    The ReticuloEndothelial System takes them out of the circula-

    tion and contrast is observed over time periods of about 20 min

    in humans and a few minutes in mice and rats. The limited cir-

    culation has consequences for targeted imaging and therapy as

    compared to nanomedicine formulations. To achieve a long circu-

    lationtime, small particles, around100200nm, have to be chosen,

    and their surface charges have to be screened, for which espe-

    cially poly-ethylene glycol is used. Although microbubbles can be

    surface-modified with PEG, their size is optimized for acoustic

    activity and therefore in the micron range. Fortunately the imag-ing of microbubbles is very sensitive and as the non-adhering

    microbubbles disappear rapidly from the blood stream only the

    few remaining adhering microbubbles can be imaged. Secondly

    the acoustic signature of a microbubble differs, it shifts to lower

    frequency, if the bubble adheres [71,72]. To exploit this for imag-

    ing, however, monodisperse microbubbles are needed [73]. Finally

    the use of radiation forces has been explored. These are long, low

    amplitude acoustic pulses that drive the bubbles to the vessel wall

    [7476]. Instead of long circulating small agents that pass by the

    region of interest manytimes and may adhere once they pass close

    to the vessel wall, microbubbles can be driven to the vessel wall

    actively.

    Compared to other imaging modalities targeted contrast ultra-

    sound has the advantage that imaging can be performed relativelyfast at a high sensitivity. The microbubblesize, however, also brings

    a drawback as it is more subject to shear forces in the blood flow

    [32]. Although this has only be shown in a flow cell, the effects

    of shear flow and microbubble displacement upon application of

    ultrasound are aspects that need further study before ultrasound

    imaging with targeted microbubbles can be used for more than

    qualitative purposes.

    The limited circulation time of microbubbles also has con-

    sequences for their use as drug delivery vehicles. Treatment

    preferably takes place shortly after injection of microbubbles and

    will be restricted to well-perfused areas. Repeated injections and

    ultrasound treatments are normally used to evaluate the effect of

    ultrasound triggered release of drugs from microbubbles in terms

    of tumor growth reduction.

  • 8/7/2019 Bhmer et al 2009

    7/12

    248 M.R. Bhmer et al. / European Journal of Radiology 70 (2009) 242253

    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 forultrasound 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-

    convertedby a trigger,such as an ultrasound pulse. The doxorubicin

    containing particles are so small that they escape the vasculature

    in a tumor by the enhanced permeation and retention effect and,

    when exposed to ultrasound, cause pronounced tumor regression.

    Long time imaging of remaining gas bubbles in tumor tissue was

    possible.

    4. Temperature sensitive drug delivery systems

    While thermal ablation requires a substantial thermal dose toinduce tissue necrosis, a more subtle temperature increase can

    be used to support treatment with conventional chemotherapeu-

    tics and drug delivery systems [79]. Mild hyperthermia enhances,

    for example, extravasation of drug loaded liposomes like Doxil

    [80,81] or enhances anti-angiogenic treatment [82]. Hyperther-

    mia can also increase local drug concentrations in conjunction

    with temperature-induced drug delivery [83]. Temperature sensi-

    tive drug delivery systems were already explored in combination

    with hyperthermia induced by radiofrequency (RF) [84] magnetic

    particles [85], or byheatingwithlightin theinfrared regime [86,87].

    Only recently, some of these temperature sensitive drug delivery

    carriers were explored in combination with ultrasound induced

    drug delivery [88]. The more efficient uptake of drug delivery sys-

    tems in tumors at elevated temperatures, together with the localtemperature triggered release of drugs, makes ultrasound induced

    drug delivery a very promising field.

    Temperature sensitive drug delivery systems can be designed

    following two different approaches. One class of agents is based on

    amphiphilic temperature sensitive polymers showing a lower crit-

    ical solution temperature (LCST) in aqueous solution. The second

    class of temperature sensitive drug delivery carriers is based on

    liposomes. Here, lipids are used that show a phase transition above

    body temperature. Upon passing the phase transition temperature,

    the liposomal bilayer becomes leaky for drugs encapsulated in the

    inner lumen of the liposome.

    4.1. Polymer-based systems

    Temperature sensitive polymeric drug delivery systems are

    usually based on polymers that undergo upon heating a phase

    transition associated with a change in polymersolvent interac-

    tion [8991]. The solvent properties change from a good solvent

    at temperatures below the LCST to a poor solvent at temperatures

    above, leading to a morphology change from an extended random

    coil toa collapsedchain(Fig. 6a). Most polymersstudied in this con-

    text are based on N-isopropylacrylamide (NIPAAm) (Fig. 1b). The

    LCST behavior of this polymer is due to a loss of hydrogen bonding

    between the amino-group and surrounding water, and increased

    hydrophobic interactions of the N-isopropylgroups above the tran-

    sition temperature

    Different strategies were followed to design temperature sensi-

    tive drug delivery systems based on above mentioned temperature

    Fig. 6. (a) Phase behavior of poly(N-isopropylacrylamide)-based polymers (b) in

    water. At temperatures below the LCST the polymer is water soluble (random coil

    configuration), while the chain collapses at temperatures above the LCST.

    sensitive polymers. One strategy pursues micelles formed from

    amphiphilic diblocks made up from a hydrophobic inert block-

    polymer, like polylactic acid, polystyrene, etc. and a PNIPAAm

    block [92,93]. The micelle can be loaded with drugs. Below the

    LCST, this diblock self-assembles into micelles with a hydrophilic

    PNIPAAmcorona. Heating induces a hydrophilichydrophobictran-

    sition of the PNIPAAm block polymer, leading to a destabilizationand morphology change such as aggregation of the micelles. The

    latter can significantly enhance drug release compared to tem-

    peratures below the LCST. The LCST can be fine-tuned in a wider

    temperature range by designing end-functionalized NIPAAm-based

    polymers,copolymer or block polymers [94,95]. However, the com-

    plex dependence of the LCST on intramolecular hydrogen bonding

    and electrostatic interactions and interactions with water makes

    the LCST susceptible to pH, ionic strength of the solvent and inter-

    actions with other molecules. The advantage of delivery systems

    where the drug release can be fine-tuned with respect to tempera-

    tureand pH comeswith thedisadvantagethat drugreleasebecomes

    also more complex and difficult to control in vivo as the LCST in

    vitro and in vivo can significantly differ.Though many systems were

    investigatedin vitro,little work hasbeen done in preclinicalstudiesin general or in combination withultrasound induced hyperthermia

    in specific. One of the few preclinical studies exploits the interde-

    pendency of pH and temperature to enhance drug delivery in the

    more acidic environment of a tumor [96]. Other preclinical studies

    showed the feasibility of temperature-induced drug delivery using

    temperature sensitive polymeric micelles [97,98].

    4.2. Temperature sensitive liposomes

    Liposomal drug delivery systems are a well-studied field and

    found their applications in cancer therapy in the clinic [99]. Lipo-

    somes can be loaded with different hydrophilic drugs in the inner

    water compartment. A particularly high drug payload is achievable

    with drugs like doxorubicin or daunorubicin that precipitate in aninner lumen loading mechanismbasedon a pHgradient. Theconve-

    nient method of drug loading and the achievable high drug payload

    probably explain the dominant role of these drug delivery systems.

    Drug release from conventional liposomal formulations is usually

    diffusioncontrolled, showing little differencein drugreleasekinetic

    at body temperature compared to temperatures slightly above in

    the hyperthermia regime. The concept of drug delivery using tem-

    perature sensitive liposomes was introduced by Yatvin et al. more

    than 25 years ago using cis-platinum as a drug compound [100].

    The field of liposome-based drug delivery under hyperthermia was

    reviewed several times [79,101].

    Temperature sensitive liposomes (TSL) are composed of lipids

    thatshow a melting transition of the acyl-chains in the bilayer[102].

    TSLs show a strongincrease indrug release around themeltingtran-

  • 8/7/2019 Bhmer et al 2009

    8/12

    M.R. Bhmer et al. / European Journal of Radiology 70 (2009) 242253 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

    of transient pores in the bilayer [103,104]. The release temperature

    andthe release kinetic can be controlledby choosing suitable lipids

    with a Tm in the desired range of around 14 K above body temper-

    ature and the incorporation of lysolipids [104,105], which induce

    efficient pore formation after the melting transition of the mem-

    brane. Temperature-induced delivery of doxorubicin encapsulated

    in TSLs was thoroughly investigated in preclinical studies and is

    now in clinical trials [106].

    4.3. Temperature triggered release

    In all temperature-induced drug delivery application, the chal-

    lenge remains to quantify the amount of release drugs and

    eventually control the release quantitatively. The achieved drug

    concentration within a treated lesion can be evaluatedin preclinical

    studies using standard analytical means, however, imaging tech-

    niques such as nuclear imaging or MRI offer the advantage of being

    applicable to humans in a clinical setting. Fig. 7 shows the under-

    lying idea of incorporating a drug and contrast or imaging agent

    inside a thermo-sensitive liposome. Upon heating the drug and

    the contrast agents are released. The observable contrast originates

    from a change of the local environment of the release contrast

    agents, which correlates with thedrug release. Examples are T1 MRcontrast agents that are incorporated inside the liposome at high

    concentration, and provide a strong signal once they are released

    from the liposome.

    Nuclear imaging is the method of choice to quantify the amount

    of drug delivery systems accumulated in the lesion [107], or even-

    tually also the released drug within the tissue. The disadvantage is

    therequired radiolabeling of thedrug and/ordrug carrier andprob-

    lematic integration of nuclear imaging in the treatment protocol as

    a standard technique. Thus, nuclear imaging will probably keep its

    role forvalidationin a research phase butwill notget a majorrole in

    image-guided drug delivery. More promising is MR imaging of drug

    release, especially in the combined setting of an HIFU/MRI system.

    Here, MR contrast agents can be incorporated in the drug delivery

    carriers that provide a signalchange upon drug release. The valueofthis approach was shown using for example temperature sensitive

    liposomes filled with Gd-based T1 contrast agents or manganese

    based agents [86,87,102,108110].

    Temperature sensitive liposomes can also be designed by con-

    jugating PNIPAAm polymers to the liposomal membrane [111,112].

    Below the LTCS, the hydrophilic PNIPAAm polymer extends into

    solution and stabilizes the liposome. Upon heating above the LCST,

    the hydrophilic to hydrophobic transition leads to a destabiliza-

    tion of the liposome and in vivo to a very different interaction with

    cells.

    No matter which approach is used, all temperature sensi-

    tive drug delivery systems can be prepared in the size range of

    10300nm, which presentsa majordifferencewith respect to pres-

    sure sensitive micro bubbles. As most microbubbles stay within

    the vascular system, released drugs still need to extravasate, possi-

    bly aided by sonoporation to reach the target tissue. Blood flow

    in the capillaries may carry away the released drug, diminish-

    ing the effect high local drug concentration. Here, the smaller

    size of temperature sensitive systems potentially offers the advan-

    tage that the place of release can be chosen. At early times after

    administration, drug release can take place in the microvascular

    system when passing through a tissue with elevated temperature.

    At later time points after administration, the drug delivery car-

    rier may have accumulated first in a lesion either by passive oractive targeting mechanisms (e.g., EPR effect or using specific tar-

    geting ligands). Drug release in the interstitial space is triggered

    subsequently by heating the lesion using, for example, ultrasound.

    However, alsoin the microvasculaturean increasedconcentrationis

    found. For example, studies with doxorubicin loaded TSLs revealed

    a 30-fold peak concentration of doxorubicin in the microvascula-

    ture. Pronounced effects on tumor growth are also found if the

    heat-treatment is given shortly after injection [108], therefore we

    can only conclude that effective mechanism needs further study.

    Treatment response may be a result of anti-vascular effects and

    anti-neoplastic mode of action [101,113].

    Future work will aim at designing new temperature sensitive

    drug carriers that show a sharp and rapid drug release at temper-

    atures slightly above body temperature. However, as much workwill be needed to optimize the treatment protocol with respect to

    timing of injection, rate of extravasation, drug release kinetic, and,

    finally, the timing of ultrasound induced hyperthermia itself with

    existing drug delivery systems. The possibility of controlling the

    drug delivery in real time under image guidance and quantifying

    the drug release using, for example, MRI will be essential to estab-

    lish a treatment protocol that is superior to todays approach with

    better therapeutic value.

    5. Therapeutic applications of ultrasound triggered drug

    delivery

    5.1. Cardiology

    Ultrasound contrast agents have been approved for diagnostic

    purposes in the filed of cardiology to better visualize the left ven-

    tricle. Therefore many ultrasound contrast agent mediated drug

    delivery studies have been performed in the heart as recently

    reviewed by Mayer and Bekeradjian [8]. The paper summarizes

    the field of gene delivery using ultrasound mediated delivery tech-

    niques and gives examples of studies where the expression of

    reporter genes was, in rats and mice, enhanced with a factor 3300

    using ultrasound frequencies around 1 MHz. Also for therapeutic

    genes an overview of a number of studies is given. The majority

    of work in the cardiology field has focused on gene delivery but

    the ultimate therapeutic aim is to induce angiogenesis and car-

    diac repair. Side effects by low molecular weight drugs are less

    of an issue in cardiology compared to oncology and have, there-

  • 8/7/2019 Bhmer et al 2009

    9/12

    250 M.R. Bhmer et al. / European Journal of Radiology 70 (2009) 242253

    fore, received less attention in the cardiology research on triggered

    delivery.

    As described previously, gas-filled microbubbles are an impor-

    tant ultrasonic contrast agent used to enhance edge detection and

    evaluate myocardial perfusion [114,115]. It was shown in preclinical

    experiments that ultrasound mediated drug delivery can directly

    enhance the expression of adenoviral vectors and plasmid DNA to

    the heart [70,116,117,118].

    USDD can successfully deliver plasmid DNA to myocardium. In

    optimization experiments, the levels of reporter gene luciferase

    expression were similar to that obtained using adenovirus but

    without the profound liver uptake associated with adenovirus

    [16].

    In addition USDD has also been applied for organ-specific deliv-

    ery of other bioactive agents [69] and could facilitate thedelivery of

    protein therapeutics to ultrasound-accessible organs whilekeeping

    systemic concentrations and side effects low. Vascular endothe-

    lial growth factor (VEGF) bound to albumin microbubbles was

    delivered to the heart using USDD. A more than 10-fold increase

    of cardiac VEGF uptake was seen compared with systemic VEGF

    administration [118]. Some preclinical studies have demonstrated

    that ultrasound alone can facilitate uptake of various substances

    including biologics.

    Striking are the examples of Kondo et al. [18] to treat an acutemyocardial infarction resulting in enhanced angiogenesis and lim-

    itation of the infarct size. Also, Korpanty et al. [119,120] have

    shown increased density of arterioles and capillaries after ultra-

    sound mediated gene transfer.

    5.2. Oncology

    In the last decades cancer has moved from a deadly to a man-

    ageable chronic disease. Cancer in the breast, prostate, liver and

    other organs can be imaged quite accurately with diagnostic ultra-

    sound [121] and ifthesecarcinomas canbe segmented, targeted and

    treated with therapeutic ultrasound, a new non-invasive, blood-

    less approach to the treatment of such diseases can be developed.

    However, after surgical removal and/or treatment with radiation orHIFUof a primary tumor,management of the residual tumor includ-

    ing metastasis is typically carried out using a variety of systemic

    therapies that include small organic molecules, and increasingly

    innovative therapiessuch as biologics and the emerging siRNA ther-

    apeutics [122,123].

    For advanced tumor stages, chemotherapy remains the treat-

    ment of choice. Despite the fact that such anticancer agents have a

    very effective tumor killing potential in vitro and in animal cancer

    models, they often fail in patients as they are unable to reach all

    tumor cells that are able to regenerate the tumors [124] and, there-

    fore,chemotherapy is rarelycurative butrather palliative,especially

    for solid tumors [125].

    The microenvironment of a tumor is critical in tumor initiation

    andpromotion, andthere is increasing evidence that this may be animportant factor in developing therapeutic approaches [126]. The

    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

    contribute to the development of drug resistance. As opposed to

    normal tissues, blood vessels in tumors are leaky [127] and vascu-

    lature is less spatially organized [128], resulting in the abnormal

    function of vessels [129]. The combination of impaired blood flow

    through blockage by neoplasmatic tumor tissue, a leaky vascula-

    ture, and lackof functional lymphatics leadsto increasedinterstitial

    fluid pressures. In addition, the plasma to interstitial gradient of

    osmotic pressure in tumors is also generally reduced limiting the

    extravasation and creating a major obstacle against delivery of ther-

    apeutic agents [130,131]

    Furthermore, diminished oxygen delivery and hypoxic condi-

    tions cause reduced efficacy of radiation therapy, high level of

    metabolic products (e.g., carbonic and lactic acid), lower extracel-

    lular pH and may potentially affect the cellular uptake of some

    drugs.

    Hyperthermia mediated liposomal drug delivery has shown

    promise for enhancing local drug deployment while minimizing

    drug distribution outside targeted tissues [101] and is currently

    being applied clinically in the treatment of various types of cancer.

    In cancer therapy, the studies, recently reviewed by Frenkel [3] on

    temperature sensitive delivery vehicles, are more advanced [106],

    however, radiofrequency ablation is the heating method, which is

    more invasive, and has less well-defined spatial temperature con-

    trol than ultrasound.

    The clinical relevance of such controlled and triggered release

    concepts for drug delivery systems having been demonstrated,

    research in this area focuses currently on optimization of cell

    specific targeting. These more advanced targeted nanocarriers in

    general have clearly shown their potential in various animal tumor

    models and await clinical application.

    A more novel approach is to use gene therapy in cancer treat-

    ment. A crucial requirement for gene therapy is tight control of

    transgene expression, both spatial and temporal to enhance the

    spatial targeting and efficiency of gene delivery. Tissuespecific pro-moters may also be used to limit transgene expression to targeted

    tissues, and in that way add a layer of targeting and safety to gene

    delivery procedures [132].

    Therapeutic effects could be demonstrated in vivo with var-

    ious targeted nucleic acid formulations, such as tumor-targeted

    DNA plasmids expressing p53 or tumor necrosis factor alpha, small

    interfering RNAs knocking down gene expression from tumor spe-

    cific chromosomal translocations or gene expression of tumor

    neoangiogenic processes, as well as double stranded RNA poly

    inosinecytosine which triggers apoptosis in targeted tumor cells

    [133].

    In a wider sense, gene therapy is experiencing an unprece-

    dented renaissance through the emerging field of the novel,

    innovative drug format of small interfering (siRNA). Since thediscovery that double stranded (dsRNA) can specifically inhibit

    expression of homologous genes, RNA interference (RNAi) has

    become one of the most widely used methods for studying

    loss-of-function phenotypes in model organisms and is increas-

    ingly used across the whole pharmaceutical research process

    including therapeutics. RNAi has been used to target dominant

    mutant or amplified oncogenes, translocation products, signal-

    ing molecules and viral oncogenes such as bcr-abl, mutated ras,

    or over expressed Bcl-2. Therapies based upon RNAi may have

    a number of inherent, fundamental benefits, such as harnessing

    natural pathways and the potential to target virtually any pro-

    tein, i.e., no limitation to drugable proteins. In a number of

    studies it could be demonstrated that it is possible to deliver

    siRNA intracellularly via microbubble-enhanced focused ultra-sound [62,133].

    6. Conclusions and outlook

    The integration of therapeutic interventions with diagnostic

    imaging, to allow for local image-guided delivery, calls for devel-

    opments in equipment and agents including new therapeutics.

    Focused ultrasound in combination with MRI and ultrasound imag-

    ing has great potential to bring ultrasound triggered drug release

    to the clinic, while employing pressure and temperature sensi-

    tive delivery vehicles. The preclinical data demonstrate the specific

    solutions that are emerging for local drug and gene delivery in both

    oncology and cardiology.

  • 8/7/2019 Bhmer et al 2009

    10/12

    M.R. Bhmer et al. / European Journal of Radiology 70 (2009) 242253 251

    Acknowledgement

    The assistance of Dr. Sander Langereis in preparing the figures

    is gratefully acknowledged.

    References

    [1] ClementGT.Perspectives in clinical usesof high-intensity focused ultrasound.

    Ultrasonics 2004;42(10):108793.[2] MoyerHR, DelmanKA. Theroleof hyperthermiain optimizing tumor response

    to regional therapy. Int J Hyperthermia 2008;24(3):25161.[3] Frenkel V. Ultrasound mediated delivery of drugs and genes to solid tumors.

    Adv Drug Deliv Rev 2008;60(10):1193208.[4] Ferrara K, Pollard R, Borden M. Ultrasound microbubble contrast agents: fun-

    damentals and application to gene and drug delivery. Annu Rev Biomed Eng2007;9:41547.

    [5] Hernot S, Klibanov AL. Microbubbles in ultrasound-triggered drug and genedelivery. Adv Drug Deliv Rev 2008;60(10):115366.

    [6] HusseiniGA, PittWG. Micelles andnanoparticles for ultrasonic drugand genedelivery. Adv Drug Deliv Rev 2008;60(10):113752.

    [7] HynynenK. Ultrasound fordrug andgene deliveryto thebrain.Adv DrugDelivRev 20 08;60(10):120917.

    [8] Mayer CR, Bekeredjian R. Ultrasonic gene and drug delivery to the cardiovas-cular system. Adv Drug Deliv Rev 2008;60(10):117792.

    [9] Mitragotri S. Healing sound: the use of ultrasound in drug delivery and othertherapeutic applications. Nat Rev Drug Discov 2005;4(3):25560.

    [10] Palussiere J, Salomir R, Le Bail B, et al. Feasibility of MR-guided focusedultrasound with real-time temperature mapping and continuous sonica-tion for ablation of VX2 carcinoma in rabbit thigh. Magn Reson Med2003;49(1):8998.

    [11] Vimeux FC, De Zwart JA, Palussiere J, et al. Real-time control offocused ultrasound heating based on rapid MR thermometry. Invest Radiol1999;34(3):1903.

    [12] Hokland SL, Pedersen M, Salomir R, Quesson B, Stodkilde-Jorgensen H, Moo-nen CTW. MRI-Guided focused ultrasound: Methodology and applications.IEEE Trans Med Imaging 2006;25(6):72331.

    [13] Moonen CT. Spatio-temporalcontrolof gene expression andcancer treatmentusing magnetic resonance imaging-guided focused ultrasound. Clin CancerRes 2007;13(12):34829.

    [14] Moonen CT, Quesson B, Salomir R, et al. Thermal therapies in inter-ventional MR imaging. Focused ultrasound. Neuroimaging Clin N Am2001;11(4):73747, xi.

    [15] Treat LH, McDannold N, VykhodtsevaN, Zhang Y,Tam K, HynynenK. Targeteddeliveryof doxorubicinto therat brain at therapeutic levels using MRI-guidedfocused ultrasound. Int J Cancer 2007;121(4):9017.

    [16] Chen S, Shohet RV, Bekeredjian R, Frenkel P, Grayburn PA. Optimization ofultrasound parameters for cardiac gene delivery of adenoviral or plasmiddeoxyribonucleic acid by ultrasound-targeted microbubble destruction. J AmColl Cardiol 20 03;42(2):3018.

    [17] Li P, Cao LQ, Dou CY, Armstrong WF, Miller D. Impact of myocardial contrastechocardiography on vascular permeability: an in vivo dose response studyof deliverymode, pressure amplitudeand contrast dose. Ultrasound Med Biol2003;29(9):13419.

    [18] Kondo I, Ohmori K, Oshita A, et al. Treatmentof acute myocardial infarction byhepatocyte growthfactorgene transfer: thefirst demonstration of myocardialtransferof a functionalgene using ultrasonic microbubbledestruction.J AmColl Cardiol 2004;44(3):64453.

    [19] Cosgrove D. Ultrasound contrast agents: an overview. Eur J Radiol2006;60(3):32430.

    [20] Schneider M. Molecular imaging and ultrasound-assisted drug delivery. JEndourol 2008;22(4):795801.

    [21] Lencioni R, Della Pina C, Crocetti L, Bozzi E, Cioni D. Clinical management offocal liver lesions: the key role of real-time contrast-enhanced US. Eur Radiol2007;17(Suppl. 6):F739.

    [22] Postema M, van Wamel A, Lancee CT, de Jong N. Ultrasound-induced encap-sulated microbubble phenomena. Ultrasound Med Biol 2004;30(6):82740.

    [23] Emmer M, van Wamel A, Goertz DE, de Jong N. The onset of microbubblevibration. Ultrasound Med Biol 2007;33(6):9419.

    [24] van der Meer SM, Dollet B, Voormolen MM, et al. Microbubble spectroscopyof ultrasound contrast agents. J Acoust Soc Am 2007;121(1):64856.

    [25] Bloch SH, Wan M, Dayton PA, Ferrara KW. Optical observation of lipid- andpolymer-shelled ultrasound microbubble contrast agents. Appl Phys Lett2004;84(4):6313.

    [26] Bouakaz A, Versluis M, de JongN. High-speed optical observations of contrastagent destruction. Ultrasound Med Biol 2005;31(3):3919.

    [27] Chin CT, Lancee C, Borsboom J, et al. Brandaris 128: a digital 25 millionframes per second camera with 128 highly sensitive frames. Rev Sci Instrum2003;74(12):502634.

    [28] Kooiman K, Bohmer M, Emmer M, et al. Oil-filled polymer microcapsulesfor ultrasound-mediated delivery of lipophilic drugs. J Controlled Release2009;133:10918.

    [29] Marmottant P. unpublished results.

    [30] Wallez Y, Huber P. Endothelial adherens and tight junctions in vascu-lar homeostasis, inflammation and angiogenesis. Biochim Biophys Acta2008;1778(3):794809.

    [31] Walsh DA. Pathophysiological mechanisms of angiogenesis. Adv Clin Chem2007;44:187221.

    [32] Schmidt B, Souza I, van Beek A, BohmerM. Adhesionand ultrasound-induceddelivery from monodisperse microbubbles in a parallel plate flow cell. J Con-trol Release 2008;131:1926.

    [33] Korpanty G, Carbon JG, Grayburn PA, Fleming JB, Brekken RA. Monitoringresponse to anticancer therapy by targeting microbubbles to tumor vascu-lature. Clin Cancer Res 2007;13(1):32330.

    [34] Villanueva FS, Jankowski RJ, Klibanov S, et al. Microbubbles targeted to inter-cellular adhesion molecule-1 bind to activated coronary artery endothelialcells. Circulation 1998;98(1):15.

    [35] Klibanov AL, Hughes MS, Marsh JN, et al. Targeting of ultrasound contrastmaterial, an in vitro feasibility study. Acta Radiol Suppl 1997;412:11320.

    [36] Schumann PA, Christiansen JP, Quigley RM, et al. Targeted-microbubble bind-ing selectively to GPIIb IIIa receptors of platelet thrombi. Invest Radiol2002;37(11):58793.

    [37] Lindner JR, Song J, Christiansen J, Klibanov AL, Xu F, Ley K. Ultrasound assess-ment of inflammation and renal tissue injury with microbubbles targeted toP-selectin. Circulation 2001;104(17):210712.

    [38] Weller GE, Lu E, Csikari MM, et al. Ultrasound imaging of acute cardiactransplant rejection with microbubbles targeted to intercellular adhesionmolecule-1. Circulation 2003;108(2):21824.

    [39] Ellegala DB,Leong-Poi H, Carpenter JE,et al.Imagingtumor angiogenesis withcontrast ultrasound and microbubbles targeted to alpha(v)beta3. Circulation2003;108(3):33641.

    [40] Leong-Poi H, Christiansen J, Heppner P, et al. Assessment of endoge-nous and therapeutic arteriogenesis by contrast ultrasound molec-

    ular imaging of integrin expression. Circulation 2005;111(24):324854.

    [41] WillmannJK, PaulmuruganR, ChenK, et al. US imaging of tumor angiogenesiswith microbubbles targeted to vascular endothelial growth factor receptortype 2 in mice. Radiology 2008;246(2):50818.

    [42] Schneider M, Pochon S, TardyI, et al.Angiogenic molecularimaging withKDR-targeted microbubbles. Contrast Media Mol Imaging 2007;2:274304.

    [43] Della Martina A, Allemann E, Bettinger T, et al. Grafting of abciximab toa microbubble-based ultrasound contrast agent for targeting to plateletsexpressing GP IIb/IIIacharacterization and in vitro testing. Eur J Pharm Bio-pharm 2008;68(3):55564.

    [44] Takalkar AM,KlibanovAL, RychakJJ, LindnerJR, LeyK. Bindingand detachmentdynamics of microbubbles targetedto P-selectin under controlledshear flow.J Control Release 2004;96(3):47382.

    [45] Rychak JJ, Li B, Acton ST, et al. Selectin ligands promote ultrasound contrastagent adhesion under shear flow. Mol Pharm 2006;3(5):51624.

    [46] Weller GE, Villanueva FS, Tom EM, Wagner WR. Targeted ultrasound contrastagents: in vitro assessment of endothelial dysfunction and multi-targeting to

    ICAM-1 and sialyl Lewisx. Biotechnol Bioeng 20 05;92(6):7808.[47] Klibanov AL, Rychak JJ, Yang WC, et al. Targeted ultrasound contrast agent formolecular imaging of inflammation in high-shear flow. Contrast Media MolImaging 2006;1(6):25966.

    [48] Price RJ, Skyba DM, Kaul S, Skalak TC. Delivery of colloidal particles andred blood cells to tissue through microvessel ruptures created by targetedmicrobubble destruction with ultrasound. Circulation 1998;98(13):12647.

    [49] MarmottantP,HilgenfeldtS. Controlled vesicledeformationand lysis bysingleoscillating bubbles. Nature 2003;423(6936):1536.

    [50] Marmottant P, Hilgenfeldt S. A bubble-driven microfluidic transport elementfor bioengineering. Proc Natl Acad Sci U S A 2004;101(26):95237.

    [51] van Wamel A, Kooiman K, Emmer M, ten Cate FJ, Versluis M, de Jong N. Ultra-sound microbubble induced endothelial cell permeability. J Control Release2006;116(2):e1002.

    [52] van Wamel A, Kooiman K, Harteveld M, et al. Vibrating microbubbles pokingindividual cells: drug transfer into cells via sonoporation. J Control Release2006;112(2):14955.

    [53] van Wamel A, Bouakaz A, Bernard B, ten Cate F, de Jong N. Radionu-clide tumour therapy with ultrasound contrast microbubbles. Ultrasonics

    2004;42(19):9036.[54] Mehier-Humbert S, Yan F, Frinking P, Schneider M, Guy RH, Bettinger T.

    Ultrasound-mediated gene delivery: influence of contrast agent on transfec-tion. Bioconjug Chem 2007;18(3):65262.

    [55] Hynynen K, McDannold N, Sheikov NA, Jolesz FA, Vykhodtseva N. Localand reversible blood-brain barrier disruption by noninvasive focused ultra-sound at frequencies suitable for trans-skull sonications. Neuroimage2005;24(1):1220.

    [56] Miller DL, Song JM. Tumor growth reduction and DNA transfer by cavitation-enhanced high-intensity focused ultrasound in vivo. Ultrasound Med Biol2003;29(6):88793.

    [57] Miller DL, Dou C. The potential for enhancement of mouse melanoma metas-tasis by diagnostic and high-amplitude ultrasound. Ultrasound Med Biol2006;32(7):1097101.

    [58] Shi WT, Bohmer M, van Wamel A, et al. Ultrasound therapy with drug loadedmicrocapsules. In: 2007 IEEE Ultrasonics Symposium. 2007. p. 7736.

    [59] Unger EC, McCreery TP, Sweitzer RH, Caldwell VE, Wu Y. Acoustically activelipospheres containing paclitaxel: a new therapeutic ultrasound contrastagent. Invest Radiol 1998;33(12):88692.

  • 8/7/2019 Bhmer et al 2009

    11/12

    252 M.R. Bhmer et al. / European Journal of Radiology 70 (2009) 242253

    [60] Hauff P, Seemann S, Reszka R, et al. Evaluation of gas-filled microparticlesand sonoporation as gene delivery system: feasibility study in rodent tumormodels. Radiology 2005;236(2):5728.

    [61] LentackerI, De Smedt SC, Demeester J, Sanders NN. Microbubbleswhich bindand protect DNA against nucleases. J Control Release 2006;116(2):e735.

    [62] Vandenbroucke RE, Lentacker I, Demeester J, De SmedtSC, Sanders NN.Ultra-soundassisted siRNAdelivery usingPEG-siPlex loaded microbubbles. J ControlRelease 2008;126(3):26573.

    [63] Borden MA, Caskey CF, Little E, Gillies RJ, Ferrara KW. DNA and polylysineadsorption and multilayer construction onto cationic lipid-coated microbub-bles. Langmuir 2007;23(18):94018.

    [64] Bohmer MR, Schroeders R, Steenbakkers JAM, et al. Preparation of monodis-perse polymer particles and capsules by ink-jet printing. Colloids Surfacesa-Physicochem Eng Aspects 2006;289(13):96104.

    [65] Straub JA, Chickering DE, Church CC, Shah B, Hanlon T, Bernstein H. PorousPLGA microparticles: AI-700, an intravenously administered ultrasound con-trast agent for use in echocardiography. J Control Release 2005;108(1):2132.

    [66] Palmowski M, HuppertJ, Ladewig G, et al. Molecular profiling of angiogenesiswith targetedultrasoundimaging: earlyassessment of antiangiogenic therapyeffects. Mol Cancer Ther 20 08;7(1):1019.

    [67] Palmowski M, Morgenstern B, Hauff P, et al. Pharmacodynamics ofstreptavidin-coated cyanoacrylate microbubbles designed for molecularultrasound imaging. Invest Radiol 2008;43(3):1629.

    [68] XieFY,Woodle MC,Lu PY. Harnessing in vivo siRNA deliveryfor drugdiscoveryand therapeutic development. Drug Discov Today 2006;11(12):6773.

    [69] Unger EC,Hersh E, Vannan M, Matsunaga TO,McCreery T.Local drugand genedelivery through microbubbles. Prog Cardiovasc Dis 2001;44(1):4554.

    [70] Frenkel PA, Chen S, Thai T, Shohet RV, Grayburn PA. DNA-loaded albuminmicrobubbles enhance ultrasound-mediated transfectionin vitro. UltrasoundMed Biol 2002;28(6):81722.

    [71] Dayton PA, Pearson D, Clark J, et al. Ultrasonic analysis of peptide- andantibody-targeted microbubble contrast agents for molecular imaging ofalphavbeta3-expressing cells. Mol Imaging 2004;3(2):12534.

    [72] Vos HJ, Dollet B, Bosch JG, Versluis M, de Jong N. Nonspherical vibrations ofmicrobubbles in contact with a wall: a pilot study at low mechanical index.Ultrasound Med Biol 2008;34(4):6858.

    [73] Talu E, Hettiarachchi K, Powell RL, Lee AP, Dayton PA, Longo ML. Maintain-ing monodispersity in a microbubble population formed by flow-focusing.Langmuir 2008;24(5):17459.

    [74] Dayton P, Klibanov A, Brandenburger G, Ferrara K. Acoustic radiation force invivo: a mechanism to assist targeting of microbubbles. Ultrasound Med Biol1999;25(8):1195201.

    [75] Rychak JJ, Klibanov AL, Ley KF, Hossack JA. Enhanced targeting of ultra-sound contrast agents using acoustic radiation force. Ultrasound Med Biol2007;33(7):11329.

    [76] Wu J, Nyborg WL. Ultrasound, cavitation bubbles and their interaction withcells. Adv Drug Deliv Rev 2008;60(10):110316.

    [77] Rapoport N, Gao Z, Kennedy A. Multifunctional nanoparticles for combin-

    ing ultrasonic tumor imaging and targeted chemotherapy. J Natl Cancer Inst2007;99(14):1095106.[78] Giesecke T, Hynynen K. Ultrasound-mediated cavitation thresholds of liquid

    perfluorocarbon droplets in vitro. Ultrasound Med Biol 2003;29(9):135965.[79] Kong G, Dewhirst MW. Hyperthermia and liposomes. Int J Hyperthermia

    1999;15(5):34570.[80] Frenkel V, Etherington A, Greene M, et al. Delivery of liposomal doxorubicin

    (Doxil) in a breast cancer tumor model: investigation of potential enhance-ment by pulsed-high intensity focused ultrasound exposure. Acad Radiol2006;13(4):46979.

    [81] Yuh EL, Shulman SG, Mehta SA, et al. Delivery of systemic chemotherapeu-tic agent to tumors by using focused ultrasound: study in a murine model.Radiology 2005;234(2):4317.

    [82] Horsman MR. Angiogenesis and vascular targeting: relevance for hyperther-mia. Int J Hyperthermia 2008;24(1):5765.

    [83] Dromi S, Frenkel V, Luk A, et al. Pulsed-high intensity focused ultrasound andlow temperature-sensitive liposomesfor enhancedtargeted drug deliveryandantitumor effect. Clin Cancer Res 2007;13(9):27227.

    [84] Friedman M, Mikityansky I, Kam A, et al. Radiofrequency ablation of cancer.

    Cardiovasc Int Radiol 2004;27(5):42734.[85] Pankhurst QA, Connolly J, Jones SK, Dobson J. Applications of magnetic

    nanoparticles in biomedicine. J Phys D-Appl Phys 2003;36(13):R16781.[86] Salomir R, Palussiere J, Fossheim SL, et al. Local delivery of magnetic reso-

    nance (MR) contrast agent in kidney using thermosensitive liposomes andMR imaging-guided local hyperthermia: a feasibility study in vivo. J MagnReson Imaging 2005;22(4):53440.

    [87] Bos C, Lepetit-Coiffe M, Quesson B, Moonen CTW. Simultaneous monitoringof temperature and T-1: methods and preliminary results of applica-tion to drug delivery using thermosensitive liposomes. Magn Reson Med2005;54(4):10204.

    [88] DeckersR, Rome C,MoonenCT.The roleof ultrasoundand magneticresonancein local drug delivery. J Magn Reson Imaging 2008;27(2):4009.

    [89] Rijcken CJ, Soga O, Hennink WE, van Nostrum CF. Triggered destabilisation ofpolymeric micelles and vesicles by changing polymers polarity: an attractivetool for drug delivery. J Control Release 2007;120(3):13148.

    [90] Chilkoti A, Dreher MR, Meyer DE, Raucher D. Targeted drug delivery by ther-mally responsive polymers. Adv Drug Deliv Rev 2002;54(5):61330.

    [91] Meyer DE, Shin BC, Kong GA, Dewhirst MW, Chilkoti A. Drug targeting usingthermally responsive polymers and local hyperthermia. J Control Release2001;74(13):21324.

    [92] Ganta S, Devalapally H, Shahiwala A, Amiji M. A review of stimuli-responsive nanocarriers for drug and gene delivery. J Control Release2008;126(3):187204.

    [93] Oh KT, Yin HQ, Lee ES, Bae YH. Polymeric nanovehicles for anticancer drugswith triggering release mechanisms. J Mater Chem 2007;17:39874001.

    [94] Liu XM, Wang LS. A one-pot synthesis of oleic acid end-capped temperature-and pH-sensitive amphiphilic polymers. Biomaterials 2004;25(10):192936.

    [95] Liu XM, Pramoda KP, Yang YY, Chow SY, He CB. Cholesteryl-grafted functionalamphiphilic poly(N-isopropylacrylamide-co-N-hydroxylmethylacrylamide):synthesis, temperaturesensitivity, self-assembly and encapsulation of ahydrophobic agent. Biomaterials 2004;25(13):261928.

    [96] Liu SQ, Wiradharma N, GaoSJ, Tong YW, Yang YY. Bio-functional micelles self-assembledfrom a folate-conjugatedblock copolymerfor targeted intracellulardelivery of anticancer drugs. Biomaterials 2007;28(7):142333.

    [97] Liu BR, Yang M, Li RT, et al. The antitumor effect of novel docetaxel-loadedthermosensitive micelles. Eur J Pharm Biopharm 2008;69(2):52734.

    [98] Na K, Lee KH, Lee DH, Bae YH. Biodegradable thermo-sensitive nanopar-ticles from poly(L-lactic acid)/poly(ethylene glycol) alternating multi-blockcopolymer for potential anti-cancer drug carrier. Eur J Pharm Sci2006;27(23):11522.

    [99] DrummondDC, MeyerO, HongK, KirpotinDB, PapahadjopoulosD. Optimizingliposomes fordelivery ofchemotherapeutic agentsto solidtumors. Pharm Rev1999;51(4):691743.

    [100] Yatvin MB,MuhlensiepenH, PorschenW,WeinsteinJN,FeinendegenLE. Selec-tivedelievery of liposome-associatedcis-diclorodiammineplatium(II) by heatandits influenceon tumor uptakeand growth.CancerRes 1981;41(5):16027.

    [101] Ponce AM, Vujaskovic Z, Yuan F, Needham D, Dewhirst MW. Hyperthermiamediated liposomal drug delivery. Int J Hyperthermia 20 06;22(3):20513.

    [102] Viglianti BL, Ponce AM, Michelich CR, et al. Chemodosimetry of invivo tumor liposomal drug concentration using MRI. Magn Reson Med2006;56(5):10118.

    [103] Needham D, Dewhirst MW. The development and testing of a newtemperature-sensitive drugdelivery systemfor thetreatment of solidtumors.Adv Drug Deliv Rev 2001;53(3):285305.

    [104] MillsJK,NeedhamD. Thematerialsengineering oftemperature-sensitivelipo-somes. Methods Enzymol 2004;387:82113.

    [105] Anyarambhatla GR, Needham D. Enhancement of the phase transitionpermeability of DPPC liposomes by incorporation of MPPC: a newtemperature-sensitive liposome for use with mild hyperthermia. J LiposomeRes 1999;9(4):491506.

    [106] Celsion. www.celsioncom.[107] Kleiter MM,YuDH, MohammadianLA, etal. A tracerdose oftechnetium-99m-

    labeled liposomes can estimate the effect of hyperthermia on intratumoralDoxil extravasation. Clin Cancer Res 2006;12(22):68007.

    [108] Ponce AM, Viglianti BL, Yu D, et al. Magnetic resonance imaging oftemperature-sensitive liposome release: drug dose painting and antitumoreffects. J Natl Cancer Inst 20 07;99(1):5363.

    [109] Viglianti BL, Abraham SA, MacFall JR, Bally MB, Dewhirst MW. In vivo mon-itoring of real time tissue pharmacokinetics of liposome/drug concentrationusing MRI. Radiology 2002;225:6478.

    [110] VigliantiBL, Abraham SA,Michelich CR,et al.In vivo monitoring oftissuephar-macokinetics of liposome/drug using MRI: illustration of targeted delivery.Magn Reson Med 2004;51(6):115362.

    [111] Kono K, Yoshino K, Takagishi T. Effect of poly(ethylene glycol) grafts ontemperature-sensitivity of thermosensitive polymer-modified liposomes. JControl Release 2002;80(13):32132.

    [112] Kono K. Thermosensitive polymer-modified liposomes. Adv Drug Deliv Rev2001;53(3):30719.

    [113] Chen Q, Tong S, Dewhirst MW, Yuan F. Targeting tumor microvessels usingdoxorubicin encapsulated in a novel thermosensitive liposome. Mol CancerTher 2004;3( 10):13117.

    [114] Amyot R, Morales MA, Rovai D. Contrast echocardiography for myocardialperfusion imaging using intravenous agents: progress and promises. Eur J

    Echocardiogr 2000;1(4):23343.[115] Mayer S, Grayburn PA. Myocardialcontrast agents:recentadvancesand future

    directions. Prog Cardiovasc Dis 2001;44(1):3344.[116] Shohet RV, Chen S, Zhou YT, et al. Echocardiographic destruction of albu-

    min microbubbles directs gene delivery to the myocardium. Circulation2000;101(22):25546.

    [117] Bekeredjian R, Chen S, Frenkel PA, Grayburn PA, Shohet RV. Ultrasound-targeted microbubble destruction can repeatedly direct highly specificplasmid expression to the heart. Circulation 2003;108(8):10226.

    [118] Bekeredjian R, Grayburn PA, Shohet RV. Use of ultrasound contrast agentsfor gene or drug delivery in cardiovascular medicine. J Am Coll Cardiol2005;45(3):32935.

    [119] Korpanty G,Chen S,ShohetRV, etal. Targetingof VEGF-mediatedangiogenesisto rat myocardium using ultrasonic destruction of microbubbles. Gene Ther2005;12(17):130512.

    [120] Korpanty G, Grayburn PA, Shohet RV, Brekken RA. Targeting vascularendothelium with avidin microbubbles. Ultrasound Med Biol 2005;31(9):127983.

    http://www.celsioncom/http://www.celsioncom/
  • 8/7/2019 Bhmer et al 2009

    12/12

    M.R. Bhmer et al. / European Journal of Radiology 70 (2009) 242253 253

    [121] Thuroff S, Chaussy C, Vallancien G, et al. High-intensity focused ultrasoundandlocalizedprostate cancer: efficacy resultsfrom theEuropean multicentricstudy. J Endourol 2003;17(8):6737.

    [122] Haussecker D. The business of RNAi therapeutics. Hum Gene Ther2008;19(5):45162.

    [123] ReischlD, ZimmerA. Drugdeliveryof siRNA therapeutics: potentialsand limitsof nanosystems. Nanomedicine 2008.

    [124] MinchintonAI, Tannock IF. Drugpenetration in solidtumours. NatRev Cancer2006;6(8):58392.

    [125] Lake RA, Robinson BW. Immunotherapy and chemotherapya practical part-nership. Nat Rev Cancer 2005;5(5):397405.

    [126] Witz IP. Tumor-microenvironmentinteractions: dangerous liaisons.Adv Can-cer Res 2008;100:20329.

    [127] Galmarini CM, Galmarini FC. Multidrug resistance in cancer therapy: role ofthe microenvironment. Curr Opin Investig Drugs 2003;4(12):141621.

    [128] McDonaldDM, ChoykePL. Imagingof angiogenesis: frommicroscopeto clinic.Nat Med 2003;9(6):71325.

    [129] Jain RK. Antiangiogenic therapy for cancer: current and emerging concepts.Oncology (Williston Park) 2005;19(4 Suppl 3):716.

    [130] Boucher Y, Baxter LT, Jain RK. Interstitial pressure gradients in tissue-isolated and subcutaneous tumors: implications for therapy. Cancer Res1990;50(15):447884.

    [131] Di Paolo A, Bocci G. Drug distribution in tumors: mechanisms, role in drugresistance, and methods for modification. Curr Oncol Rep 2007;9(2):10914.

    [132] Greenberger S, Shaish A, Varda-Bloom N, et al. Transcription-controlled

    gene therapy against tumor angiogenesis. J Clin Invest 2004;113(7):101724.

    [133] Russ V, Wagner E. Cell and tissue targeting of nucleic acids for cancer genetherapy. Pharm Res 2007;24(6):104757.