Journal of Controlled Release - Stanford...

10
Ultrasound-guided delivery of microRNA loaded nanoparticles into cancer Tzu-Yin Wang a , Jung Woo Choe b , Kanyi Pu a , Rammohan Devulapally a , Sunitha Bachawal a , Steven Machtaler a , Sayan Mullick Chowdhury a , Richard Luong c , Lu Tian d , Butrus Khuri-Yakub b , Jianghong Rao a , Ramasamy Paulmurugan a, , Jürgen K. Willmann a, ⁎⁎ a Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, School of Medicine, Stanford, CA, USA b Department of Electrical Engineering, Stanford University, Stanford, CA, USA c Department of Comparative Medicine, Stanford University, Stanford, CA, USA d Department of Health, Research & Policy, Stanford University, Stanford, CA, USA abstract article info Article history: Received 21 October 2014 Accepted 13 February 2015 Available online 14 February 2015 Keywords: Image guidance Ultrasound Cancer Drug delivery Nanocarriers Therapy Ultrasound induced microbubble cavitation can cause enhanced permeability across natural barriers of tumors such as vessel walls or cellular membranes, allowing for enhanced therapeutic delivery into the target tissues. While enhanced delivery of small (b 1 nm) molecules has been shown at acoustic pressures below 1 MPa both in vitro and in vivo, the delivery efciency of larger (N 100 nm) therapeutic carriers into cancer remains unclear and may require a higher pressure for sufcient delivery. Enhanced delivery of larger therapeutic carriers such as FDA approved pegylated poly(lactic-co-glycolic acid) nanoparticles (PLGA-PEG-NP) has signicant clinical value because these nanoparticles have been shown to protect encapsulated drugs from degradation in the blood circulation and allow for slow and prolonged release of encapsulated drugs at the target location. In this study, various acoustic parameters were investigated to facilitate the successful delivery of two nanocarriers, a uorescent semiconducting polymer model drug nanoparticle as well as PLGA-PEG-NP into human colon cancer xenografts in mice. We rst measured the cavitation dose produced by various acoustic parameters (pressure, pulse length, and pulse repetition frequency) and microbubble concentration in a tissue mimicking phantom. Next, in vivo studies were performed to evaluate the penetration depth of nanocarriers using various acoustic pressures, ranging between 1.7 and 6.9 MPa. Finally, a therapeutic microRNA, miR-122, was loaded into PLGA- PEG-NP and the amount of delivered miR-122 was assessed using quantitative RT-PCR. Our results show that acoustic pressures had the strongest effect on cavitation. An increase of the pressure from 0.8 to 6.9 MPa resulted in a nearly 50-fold increase in cavitation in phantom experiments. In vivo, as the pressures increased from 1.7 to 6.9 MPa, the amount of nanoparticles deposited in cancer xenografts was increased from 4- to 14-fold, and the median penetration depth of extravasated nanoparticles was increased from 1.3-fold to 3-fold, compared to control conditions without ultrasound, as examined on 3D confocal microscopy. When delivering miR-122 loaded PLGA-PEG-NP using optimal acoustic settings with minimum tissue damage, miR-122 delivery into tumors with ultrasound and microbubbles was 7.9-fold higher compared to treatment without ultrasound. This study demonstrates that ultrasound induced microbubble cavitation can be a useful tool for delivery of therapeutic miR loaded nanocarriers into cancer in vivo. © 2015 Elsevier B.V. All rights reserved. 1. Introduction Nanometer scaled particles prepared with natural or synthetic polymers, lipids, or inorganic solids have been widely investigated as therapeutic carriers for anti-cancer drugs [1]. These particles, once injected into the bloodstream, can passively accumulate in tumors due to the leaky tumor vasculature and reduced lymphatic drainage, a phenomenon called enhanced permeability and retention (EPR) effect [2]. However, the delivery efciency via EPR is low: less than 5% of the administered particles accumulate in the tumor [3]. Furthermore, it has been shown in animal models that the EPR effect is highly heterogeneous Journal of Controlled Release 203 (2015) 99108 Correspondence to: R. Paulmurugan, Department of Radiology, Stanford University School of Medicine, Stanford University, 3155 Porter Drive, #2236, Palo Alto, CA 94304, USA. ⁎⁎ Correspondence to: J.K. Willmann, Department of Radiology and Molecular Imaging Program at Stanford, School of Medicine, Stanford University, 300 Pasteur Drive, Room H1307, Stanford, CA 94305-5621, USA. E-mail addresses: [email protected] (R. Paulmurugan), [email protected] (J.K. Willmann). http://dx.doi.org/10.1016/j.jconrel.2015.02.018 0168-3659/© 2015 Elsevier B.V. All rights reserved. Contents lists available at ScienceDirect Journal of Controlled Release journal homepage: www.elsevier.com/locate/jconrel

Transcript of Journal of Controlled Release - Stanford...

Page 1: Journal of Controlled Release - Stanford Universitystanford.edu/group/khuri-yakub/publications/15_Wang_01.pdfloaded PLGA-PEG-NP using optimal acoustic settings with minimum tissue

Journal of Controlled Release 203 (2015) 99–108

Contents lists available at ScienceDirect

Journal of Controlled Release

j ourna l homepage: www.e lsev ie r .com/ locate / jconre l

Ultrasound-guided delivery of microRNA loaded nanoparticlesinto cancer

Tzu-YinWang a, JungWooChoe b, Kanyi Pu a, RammohanDevulapally a, Sunitha Bachawal a, StevenMachtaler a,Sayan Mullick Chowdhury a, Richard Luong c, Lu Tian d, Butrus Khuri-Yakub b, Jianghong Rao a,Ramasamy Paulmurugan a,⁎, Jürgen K. Willmann a,⁎⁎a Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, School of Medicine, Stanford, CA, USAb Department of Electrical Engineering, Stanford University, Stanford, CA, USAc Department of Comparative Medicine, Stanford University, Stanford, CA, USAd Department of Health, Research & Policy, Stanford University, Stanford, CA, USA

⁎ Correspondence to: R. Paulmurugan, Department ofSchool of Medicine, Stanford University, 3155 Porter DriUSA.⁎⁎ Correspondence to: J.K. Willmann, Department of RaProgram at Stanford, School of Medicine, Stanford UniveH1307, Stanford, CA 94305-5621, USA.

E-mail addresses: [email protected] (R. Paulmu(J.K. Willmann).

http://dx.doi.org/10.1016/j.jconrel.2015.02.0180168-3659/© 2015 Elsevier B.V. All rights reserved.

a b s t r a c t

a r t i c l e i n f o

Article history:Received 21 October 2014Accepted 13 February 2015Available online 14 February 2015

Keywords:Image guidanceUltrasoundCancerDrug deliveryNanocarriersTherapy

Ultrasound induced microbubble cavitation can cause enhanced permeability across natural barriers of tumorssuch as vessel walls or cellular membranes, allowing for enhanced therapeutic delivery into the target tissues.While enhanced delivery of small (b1 nm) molecules has been shown at acoustic pressures below 1 MPa bothin vitro and in vivo, the delivery efficiency of larger (N100 nm) therapeutic carriers into cancer remains unclearandmay require a higher pressure for sufficient delivery. Enhanced delivery of larger therapeutic carriers such asFDA approved pegylated poly(lactic-co-glycolic acid) nanoparticles (PLGA-PEG-NP) has significant clinical valuebecause these nanoparticles have been shown to protect encapsulated drugs from degradation in the bloodcirculation and allow for slow and prolonged release of encapsulated drugs at the target location. In thisstudy, various acoustic parameters were investigated to facilitate the successful delivery of two nanocarriers, afluorescent semiconducting polymer model drug nanoparticle as well as PLGA-PEG-NP into human colon cancerxenografts in mice. We first measured the cavitation dose produced by various acoustic parameters (pressure,pulse length, and pulse repetition frequency) and microbubble concentration in a tissue mimicking phantom.Next, in vivo studies were performed to evaluate the penetration depth of nanocarriers using various acousticpressures, ranging between 1.7 and 6.9 MPa. Finally, a therapeutic microRNA, miR-122, was loaded into PLGA-PEG-NP and the amount of delivered miR-122 was assessed using quantitative RT-PCR. Our results show thatacoustic pressures had the strongest effect on cavitation. An increase of the pressure from 0.8 to 6.9MPa resultedin a nearly 50-fold increase in cavitation in phantom experiments. In vivo, as the pressures increased from 1.7 to6.9 MPa, the amount of nanoparticles deposited in cancer xenografts was increased from 4- to 14-fold, and themedian penetration depth of extravasated nanoparticles was increased from 1.3-fold to 3-fold, compared tocontrol conditions without ultrasound, as examined on 3D confocal microscopy. When delivering miR-122loaded PLGA-PEG-NP using optimal acoustic settings with minimum tissue damage, miR-122 delivery intotumors with ultrasound and microbubbles was 7.9-fold higher compared to treatment without ultrasound.This study demonstrates that ultrasound induced microbubble cavitation can be a useful tool for delivery oftherapeutic miR loaded nanocarriers into cancer in vivo.

© 2015 Elsevier B.V. All rights reserved.

Radiology, Stanford Universityve, #2236, Palo Alto, CA 94304,

diology and Molecular Imagingrsity, 300 Pasteur Drive, Room

rugan), [email protected]

1. Introduction

Nanometer scaled particles prepared with natural or syntheticpolymers, lipids, or inorganic solids have been widely investigated astherapeutic carriers for anti-cancer drugs [1]. These particles, onceinjected into the bloodstream, can passively accumulate in tumors dueto the leaky tumor vasculature and reduced lymphatic drainage, aphenomenon called enhanced permeability and retention (EPR) effect[2]. However, the delivery efficiency via EPR is low: less than 5% of theadministered particles accumulate in the tumor [3]. Furthermore, it hasbeen shown in animalmodels that the EPR effect is highly heterogeneous

Page 2: Journal of Controlled Release - Stanford Universitystanford.edu/group/khuri-yakub/publications/15_Wang_01.pdfloaded PLGA-PEG-NP using optimal acoustic settings with minimum tissue

100 T.-Y. Wang et al. / Journal of Controlled Release 203 (2015) 99–108

in different tumormodels, and even within a single tumor [4,5]. Little isknown about the EPR effect in humans. One clinical study found sub-stantial variation of the delivery dose of liposomes via the EPR effectranging between 2.7 and 53% injected dose/kg in patients with varioustumors including breast, lung, and head and neck cancers [6]. Therefore,therapeutic delivery of drugs relying only on the EPR effect likely leadsto unreliable and unpredictable clinical outcomes. An active deliveryapproach augmenting drug delivery into tumors is critically needed.

Drug delivery facilitated by ultrasound (US) and contrastmicrobubbles (MB) is a promising active delivery approach. Thisapproach utilizes the interactions between US and clinically used MBto create openings on a nearby surface, such as the vessel wall and/orcell membranes, allowing for increased particle permeability acrossnatural barriers, a process termed “sonoporation”. Sonoporation ishighly associated with cavitation, which involves expansion, contrac-tion, and/or collapse of gas-filled MB. The expansion and contractionof MB can directly ‘push’ and ‘pull’ a nearby surface and induce fluidjets and microstreaming in the surrounding fluid [7,8]. These MBactivities were estimated to produce shear stress as high as 74 kPa,and circumference stress of 0.43–6 MPa [9,10]. Such high stress issufficient to cause deformation or rupture of the nearby surface (cellularmembranes or vessel walls) observed in several previous studies [11,12]. Sonoporation has been investigated for the treatment of preclinicalbrain [13], liver [14], pancreatic [15], breast [16], and ovarian cancers[17], and recently in a first in man clinical case study for adjuvant treat-ment of locally advanced pancreatic cancer [18].

The feasibility of US and MB facilitated delivery has been demon-strated using model drugs with sizes varying from b1 nm to 10 μm,with the majority being less than 20 nm [19]. Delivery of larger(N100 nm) therapeutics or therapeutic carriers is more challengingbecause higher energies may be needed to create larger openings forthe particles to pass through [20]. However, delivery of large therapeu-tics has great clinical significance. Many therapeutic agents are eitherquickly degraded or rapidly cleared by the reticuloendothelial systemafter free intravenous administration. Encapsulating these therapeuticagents in carriers can increase their circulation time and chance toreach the target tissue. In addition, encapsulation of cytotoxic drugssuch as chemotherapeutics can significantly reduce systemic toxicity.Currently, commercially available nanocarriers are produced withsizes ranging between 90 and 300 nm (for examples, Doxil is 90 nm[21], Epaxel is 150 nm [22], and Verteporfin is 150 to 300 nm [23] indiameter) to ensure sufficient drug loading capacity.

MicroRNAs (MiRs) are regulatorymolecules of cells that regulate theexpression of various cellular genes involved in several pathways relatedto the onset of tumor development, proliferation, drug-resistance andmetastasis. It has been found that the MiRs are abnormally expressedin multiple cancer types. Restoration of the miR levels during cancerpathogenesis can affect cancer development, thereby acting as a potentanti-cancer drug [24]. In addition, certain miR can be downregulated inseveral cancer types [24], thereby potentially having patients withdifferent cancer types benefit from successful delivery of a particularmiR. In this study, we investigated the delivery of miR-122, an endoge-nous miR associated with the pathogenesis of cancers. The down-regulation of miR-122 is responsible for enhanced tumor proliferationand resistance to conventional chemotherapy [25]. Over-expression ofmiR-122 can induce apoptosis and/or cell cycle arrest [26], inhibitionof tumorigenesis and angiogenesis [27], and re-sensitizes varioustypes of cancers, including colon [28] and liver [29] cancers to chemo-therapy. Successful delivery of sufficient amounts of miR-122 intotumors to act as new anti-cancer agent is of paramount importance.However, miRs are quickly degraded by nucleases once injectedintravenously. Here we proposed to encapsulate the miR-122 intonanocarriers to prevent nuclease degradation. Delivery of miR-122loadednanocarriers at the tumor sitewas then facilitated byUS andMB.

To investigate US and MB assisted delivery of nanocarriers, twotypes of nanocarriers were investigated in this study. The first type

was semiconducting polymer nanoparticles (SPN). SPN are fluorescentnanomaterials made with completely organic and biologically nontoxiccomponents [30]. The controllable size, good biocompatibility, andstable fluorescence under physiological conditions make them an idealbiosensor for disease monitoring in vivo [31]. SPN were synthesizedwith a size and shell composition similar to FDA approved therapeuticcarriers, such as pegylated poly(lactic-co-glycolic acid) nanoparticles(PLGA-PEG-NP), and served as model nanocarriers. The high fluores-cence of the SPN allows for investigation of the amount and spatialdistribution of the nanocarriers in tumors. The second type ofnanocarriers was FDA approved PLGA-PEG-NP [32], with a similar sizecompared to SPN model nanocarriers and was loaded with a tumorsuppressor therapeutic microRNA, miR-122 [26,27,29,33].

We hypothesized that successful delivery of nanocarriers can beachieved by onset of inertial cavitation. Experiments were firstperformed on tissue mimicking phantoms to evaluate the acousticparameters for successful cavitation. In vivo experiments were thenconducted to explore a combination of appropriate acoustic parametersresulting in successful delivery of SPN model nanocarriers into humancolon cancer xenografts in mice with minimal tissue damage. Finally,the optimal acoustic parameters were applied for the delivery ofmiR-122 loaded PLGA-PEG-NP into human colon cancer xenografts.

2. Material and methods

2.1. Experiments in tissue mimicking phantoms

2.1.1. Tissue mimicking phantomsAgarose-based hydrogel was prepared as tissuemimicking phantom

by melting 1% w/v agarose in water. Extra fine graphite powder wasadded to the phantom during the melting process at 3% w/v, acting asacoustic scatterers for ultrasound imaging. The melted solution waspoured in a polycarbonate mold with acoustic windows for both thecavitation induction and detection transducers. A plastic rod of 3 mmdiameter was placed in the solution and removed after the gel wascompletely solidified, creating a cavitation chamber.

2.1.2. MicrobubblesClinical grade lipid-shelled perfluorobutane-and-nitrogen-filled MB

[34] (BR38; kindly provided by Bracco Research, Geneva, Switzerland),were used as the cavitation nuclei in this study. MB were lyophilizedand stored in septum-sealed vials with perfluorobutane and nitrogengas. Prior to each use, MB lyophilisates were resuspended in sterile0.9% saline.MBhad amean diameter of 1.4±0.1 μmandwere neutrallycharged (zeta potential −0.3 ± 0.3 mV) as assessed by themanufacturer.

2.1.3. Ultrasound apparatusMB cavitation was induced by US pulses of 1.8 MHz generated by an

array transducer (P4-1, Philips Healthcare, Andover,MA) connected to aresearch platform (V1, Verasonics, Redmond, WA). The pulses werecalibrated in degassed water using a needle hydrophone (HNR-0500,Onda, Sunnyvale, CA). The full width at half maximum (FWHM)beamwidths on the pressure profile were calibrated to be 1.4, 10.1,and 12.6 mm in the transducer's X, Y and Z directions, respectively.

Experimental setup of the phantom study is shown in Fig. 1. Thecavitation initiation transducer was placed on top of the phantomsuch that the lateral axis was in parallel with the cavitation chamberwith a 30 mm standoff distance. MB were then injected into the cham-ber and exposed to US. During the exposure, cavitation was detectedpassively by detecting broad band noise emitted from MB collapseand actively by imaging of the destruction of MB using US. For passivecavitation detection, a 10-MHz single element transducer (V312,Panametrics NDT, Waltham, MA) with a −6 dB bandwidth of 5.3–10.7 MHzwas used to collect the acoustic scattering from the cavitationchamber with a standoff distance of 45 mm. The two transducers were

Page 3: Journal of Controlled Release - Stanford Universitystanford.edu/group/khuri-yakub/publications/15_Wang_01.pdfloaded PLGA-PEG-NP using optimal acoustic settings with minimum tissue

Fig. 1. Experimental setup for phantom studies. The cavitation initiation transducer wasoperated at 1.8 MHz to induce microbubble cavitation. Detection of cavitation wasperformed both passively using a 10-MHz single element transducer as well as activelyby using a 5-MHz ultrasound imaging transducer. The cavitation initiation and detectiontransducers were placed perpendicular to each other and co-focused at the cavitationchamber containing microbubbles.

101T.-Y. Wang et al. / Journal of Controlled Release 203 (2015) 99–108

perpendicular and co-focused at the cavitation chamber. The collectedacoustic scattering was amplified by a pulser/receiver (PR5072,Panametrics, Waltham, MA) and recorded by a digital oscilloscope(DSO8104A, Agilent Technologies, Santa Clara, CA) at a sampling rateof 100 MSamples/s. For active cavitation detection, the 10-MHz passivecavitation detection transducer was replaced with a 5-MHz US imagingtransducer (L7-4, Philips Healthcare, Andover, MA) operated with asecond Verasonics system (V1, Verasonics, Redmond, WA). B-modeimages were acquired before, during, and after the treatment usingthe second Verasonics system at a low power setting (pressure0.3 MPa, frame rate 10 Hz) which did not destroy MB. Uncompressedimage data of the B-mode images were collected for data analysis off-line.

Effects of four parameters on cavitation were studied: peak negativepressure, pulse length, pulse repetition frequency (PRF), and MBconcentration (Table 1). In all series in the phantom studies, the centerfrequency of US was fixed at 1.8 MHz, and a total of 200 pulses weredelivered in each exposure. The duration of 200 pulses was selectedbecause the MB in the focal region are completely destroyed withinthis duration.

2.1.4. Cavitation signal processingCavitation activities in the tissuemimicking phantomwere analyzed

using both passive and active cavitation detection methods (describedin Supplementary information SI.1). In brief, for passive cavitation,inertial cavitation dose (ICD) was quantitatively evaluated by measur-ing the broadband noise emitted during inertial cavitation, as describedpreviously [35]. For active cavitation imaging, the ultrasound imaging

Table 1Experimental series in phantoms.

Peak negative pressure (MPa) Pulse length PRF (Hz)

Series 1 0, 0.8, 1.7, 2.5, 3.3, 3.9, 4.5, 5.4, 6.9 5 cycles (2.8 μs) 100Series 2 2.4 3, 5, 7, 10, 15 cycles

(1.7, 2.8, 3.9, 5.6, 8.3 μs)100

Series 3 3 5 cycles (2.8 μs) 10, 20, 50, 1Series 4 3 5 cycles (2.8 μs) 100

signal intensity changes were analyzed and compared for the differentUS conditions.

2.2. In vivo delivery of model nanocarriers

To explore appropriate acoustic conditions for optimal nanocarrierdelivery and minimal tissue damage in vivo, experiments wereconducted to study the delivery profile of the model nanocarriersunder different acoustic conditions. Here we focused on studying theeffects of varying acoustic pressures on the amount of delivery and thepenetration depth, since it was found in the phantom experimentsthat acoustic pressure had themost significant effects onMB cavitation.

2.2.1. Animal modelAll experimental procedures involving animals were approved by

the Institutional Administrative Panel on Laboratory Animal Care. Invivo experiments were performed on subcutaneous LS174T humancolon cancer xenografts established in nude mice (a total of 79 tumorsin 50 mice; see Supplementary information SI.2).

2.2.2. SPN model nanocarriersThe synthesis of fluorescence SPN is described in the Supplementary

information (see Supplementary information SI.3). The diameter ofthe SPN was 116 ± 5 nm. These highly fluorescent SPN allowed forassessment of the spatial distribution of nanocarriers delivered intoxenografts.

2.2.3. In vivo treatment protocolA total of 41 tumors were treated with SPN. Mice were positioned

on the US scanning table and gas anesthetized with 2% isoflurane in2 L of oxygen per minute. A catheter (MicroMarker Tail Vein AccessCannulation kit; VisualSonics, Toronto, Ontario)wasplaced into a lateraltail vein for administration of MB and nanocarriers. A total of 350 μL(3.5 × 108) MB and 150 μL of SPN (5.6 nM; 5 × 1011) were mixed intoa total solution of 500 μL prior to tumor treatment. The treatmentconsisted of 5 repetitive cycles of 100 μL MB/SPN mixture administra-tions combined with US exposures (Fig. 2). In each cycle, the MB/SPNmixture was first administered for 1 min at a constant rate of100 μL/min using an automated infusion pump (GeniePlus, KentScientific, Torrington, CT) to achieve steady state. This resulted in anestimated concentration of 3.3 × 107 MB/mL in the bloodstream,assuming that the total blood volume of a mouse is 2 mL [36] and thatthe MB are completely destroyed by US in each treatment cycle.Administration was then paused and US pulses were delivered to thetarget tumor sites for 1 min. The US focal beam was electronicallysteered across 6 locations, 1.5 mmapart each (Fig. 2), to allow completeanatomical coverage of the tumors. Each treatment location wasexposed to 1000 pulses of 5 cycles in pulse length delivered at a PRF of100 Hzwith varying pressures described in Table 2. The exposure dura-tion of 1000 pulses was longer than that in the phantom experiments(200 pulses) because in the in vivo condition, the tissue was constantlyreplenished with MB from the bloodstream. The longer exposureduration allowed for sufficient interaction between cavitation and thevessels, thus increasing the likelihood of sonoporation.

The MB administration rate of 100 μL/min was selected based onprevious experience that MB can be intravenously delivered without

MB concentration (counts/mL) Total numbers of pulses

1 × 108 2001 × 108 200

00 1 × 108 2004 × 106, 1 × 107, 2 × 107, 4 × 107, 1 × 108, 2 × 108 200

Page 4: Journal of Controlled Release - Stanford Universitystanford.edu/group/khuri-yakub/publications/15_Wang_01.pdfloaded PLGA-PEG-NP using optimal acoustic settings with minimum tissue

Fig. 2. In vivo experimental setup and treatment timeline. The focal beam of the cavitation initiation transducer was electronically steered across 6 locations to cover the entire tumorvolume. The high frequency imaging transducer was aimed at the target tumors at 45° angle to provide anatomical guidance before the treatment and to monitor the microbubbleperfusion/destruction process during the treatment. Microbubble perfusion and ultrasound insonation were alternated in the 10-minute-length treatment windows.

102 T.-Y. Wang et al. / Journal of Controlled Release 203 (2015) 99–108

being destroyed at that rate [37]. At this administration rate, the risetime of the perfusion curve (defined as the time for the image intensityin the tumor to reach the peak intensity fromwhen the image intensityincrease was first detected) was 61 ± 13 s (n = 4) as measured bycontrast enhanced US imaging using a 21-MHz transducer (MS250,VisualSonics, Toronto, Ontario, Canada) connected to a dedicatedsmall animal US system (Vevo 2100, VisualSonics, lateral and axialresolution of 165 and 75 μm, respectively). Therefore, an administrationperiod of 1min was selected to ensure high concentrations of MB at thetumor treatment site. To minimize the overall injection volume to atotal of 500 μL in each mouse, MB administration was paused duringUS exposure.

To provide image guidance during tumor treatment, real-time USimaging was performed in all animals using the same transducer(MS250, VisualSonics) and small animal US system (Vevo 2100,VisualSonics). Contrast-enhanced US imaging was performed usingthe nonlinear contrast mode [38] at a frame rate of 5 fps. Image datawere collected from 10 s before to 30 s after the treatments. The perfu-sion and destruction of MB as indicated by the increase and decrease ofthe image intensity were visually inspected during the treatments aswell as quantitatively assessed post treatments. The image dataacquired during the treatmentwere processed usingdedicated software(VevoCQ, VisualSonics). The temporal change in the mean imageintensitywithin the tumor regionwas obtained for the entire treatmentprocess. The mean image intensity was further normalized to thebaseline image intensity before the treatment. The maximum imageintensity and the post-treatment image intensity were comparedamong groups treated with different pressures.

2.2.4. Ex vivo analysisAll animals were euthanized 4 h post treatment and all tumors were

excised for ex vivo analysis. Excised tumor tissues were fixed in 4%paraformaldehyde overnight at 4 °C and then cut in halves at the centerof the xenografts. One half was immersed in a 30% sucrose solution for

Table 2In vivo experimental series. MiR-122 was only loaded in the third experimental group(miR-122 loaded PLGA-PEG-NP) and not in the others (SPN and control PLGA-PEG-NP).

Nanoparticle type Total injectednanoparticles

Peak negativepressure (MPa)

Numberof tumors

Numberof animals

SPN 5 × 1011 0 (control) 16 311.7 52.5 53.9 55.4 56.9 5

Control PLGA-PEG-NP 7.3 × 1011 0 (control) 3 35.4 3

miR-122 loadedPLGA-PEG-NP

6.5 × 1011 0 (control) 7 75.4 7

cryoprotection, and then embedded in optimal cutting temperature(OCT) media for immunofluorescence analysis of model nanocarriertumor delivery. The other half was immersed in 70% ethanol, andthen embedded in paraffin for histological analysis of tissue damagefollowing Hematoxylin and Eosin (H&E) staining.

2.2.4.1. Immunofluorescence analysis of model nanocarrier tumor delivery.To study the delivery profile of the model SPN nanocarriers, confirmextravasation of the nanocarriers into the tumor parenchyma, andquantify the extent of parenchymal delivery including the distancebetween nanocarriers and tumor vessels, quantitative immunofluores-cence analysis was performed on tissue sections stained for both thevascular endothelial cell marker CD31 and the cytoskeleton F-actinmarker.

Two slices, 10-μmand 50-μm-thick, were cut fromeach tumor halve.The 10-μm slices were used to quantitatively assess the extent ofnanocarrier delivery across the entire tumor diameter; the 50-μmslices were used to volumetrically quantify the distance of nanocarrierdelivery in relation to tumor vessels. The slices were stained for CD31and F-actin (see Supplementary information SI.4). Tissue slices wereimaged with a wide field fluorescence microscope (AxioImager micro-scope, Carl Zeiss GmbH, Jena, Germany) at 20× magnification. Imagesof the SPN, CD31, and F-actin were captured in separate color channels(excitation/emission wavelengths were 470/525 nm, 550/605 nm, and640/690 nm, for SPN, CD31, and F-actin channels, respectively). Rasterscans were obtained across the tissue using a motorized stagecontrolled by the microscope image acquisition software (AxioVision,Carl Zeiss GmbH, Jena, Germany). This scan allowed for reconstructionof the immunofluorescence image of the entire tumor section.

To evaluate the amount of SPN delivered into the tumor, quantita-tive image analysis was performed using Matlab (MathWorks Inc.,Natick, MA). Images of the SPN, CD31, and F-actin channels wereseparated. In each channel, a pixel intensity threshold was set to bemean + 5 × standard deviation of the pixel intensity in a backgroundarea where no vessel/cellular structures existed. This threshold waschosen to include fluorescent pixels from SPN or positive staining ofCD31/F-actin and exclude pixels with autofluorescence, as describedpreviously [39]. Pixels with intensities below this threshold wereconsidered interference from autofluorescence and were excluded.The pixel area with positive signals was calculated for each channeland integrated over all images captured for each tumor, producing atotal area of SPN, CD31 or F-actin. The total area was assumed propor-tional to the amount of SPN, vessels, or cells in each tumor. Next, tworatios were calculated: The first was the ratio of total area of SPN tothe total area of CD31, and the second was the ratio of total area ofSPN to the total area of F-actin. The first ratio normalized the amountof SPN to the amount of vessels in a tumor, allowing for comparisonbetween tumors regardless of various degrees of vascularization. The

Page 5: Journal of Controlled Release - Stanford Universitystanford.edu/group/khuri-yakub/publications/15_Wang_01.pdfloaded PLGA-PEG-NP using optimal acoustic settings with minimum tissue

103T.-Y. Wang et al. / Journal of Controlled Release 203 (2015) 99–108

second ratio normalized the area of SPN to the area of cells, indicatingthe portion of a tumor that may be exposed to the therapeutic carriers.

To confirm extravasation of nanocarriers into tumor parenchymaand to measure the distance of nanocarriers from the vessels, 3Dconfocal laser microscopy was performed on 50-μm-thick sections(LSM510 Meta confocal microscope, Carl Zeiss GmbH, Jena, Germany).Z-stack imaging was performed through the entire section with a1 μm increment. Measurements of the location of SPN with respect tothe vessels were made on the reconstructed 3D images using a 3Dimage processing software (Imaris, Bitplane, South Windsor, CT, USA).The penetration depth was measured as the distance from the SPNto the closest vessel. To study effects of the acoustic pressures on thepenetration depth, a total of 54 measurements were made for eachacoustic pressure.

2.2.4.2. Histological analysis of tissue damage.Afterfixation in 10%neutralbuffered formalin, tumor tissueswere cut into 4-μmsections, stained forH&E using a routine protocol [40], and analyzed for tissue damage by aboard-certified veterinary pathologist using an Olympus BX-41 micro-scope (Olympus, Center Valley, PA). The degree of tissue damagewithin(intratumoral) and around (peritumoral) each tumor was assessed in ablinded fashion (blinded to the US treatment conditions applied to eachtissue sample) and was defined as the percentage of the total cross-sectional area that was affected by acute hemorrhage (extravasationof red blood cells out of blood vessels) and graded using a 5-grade Likertscore (Table 3). The intratumoral hemorrhage gradewas assessed in theentire tumor region while the peritumoral hemorrhage grade wasobtained from a surrounding region within 0.5 mm from the tumorborder. Representative digital photomicrographs were taken using anAxioscope 2 Plus microscope (Carl Zeiss, Thornwood, NY) with aNikon DS-Ri1 digital microscope camera (Nikon, Melville, NY) andNIS-Elements imaging software (Nikon, Melville, NY).

2.3. Delivery of microRNA-122 encapsulated in PLGA nanoparticles

2.3.1. Synthesis of PLGA-PEG nanoparticlesControl PLGA-PEG-NP and PLGA-PEG-NP loadedwithmiR-122were

synthesized as described in Supplementary information SI.5. Thediameters of control PLGA-PEG-NP and miR-122 loaded PLGA-PEG-NPwere 110.6 ± 23 and 115.3 ± 18 nm, respectively. The surface charge,miR-122 loading percentage, and number of miR molecules loadedper nanoparticle in PLGA-PEG-NP can be found in Supplementaryinformation SI.5.

2.3.2. In vivo tumor delivery of miR-122-loaded PLGA nanoparticlesOptimal acoustic parameters resulting in maximum nanocarrier

delivery at minimal tissue damage were chosen to deliver the miR-122loaded PLGA-PEG-NP in vivo (detailed parameters listed in Table 2).The experimental setup and treatment protocols were the same asthose described for the SPN model nanocarriers (Fig. 2).

Ninemicewith a total of 18 tumors (one on each side) were treated.US treatmentwas performed in one of the tumors and in the samemicethe tumor on the other flank served as an untreated intra-animalcontrol. Three mice received control PLGA-PEG-NP without miR-122,and the remaining 6 mice received miR-122 loaded PLGA-PEG-NP.

Table 3Grading of tissue damage following sonoporation.

Grade Definition

0 No hemorrhage1 0–10% of area with hemorrhage2 11–25% of area with hemorrhage3 26–50% of area with hemorrhage4 N50% of area with hemorrhage

Four hours after treatment, animals were perfused with phosphatebuffered saline for 5 min under anesthesia in order to remove excessPLGA-PEG-NP in the systemic circulation. The tumors were excisedand snap frozen on dry ice for quantitative reverse transcription poly-merase chain reaction (RT-PCR) process for miR-122 quantification.The RT-PCR protocol and miR-122 quantification method are describedin the Supplementary information (SI.6).

2.4. Statistical analysis

Values are expressed as mean ± standard deviation. Linear regres-sion analysis, ANOVA, and theWilcoxon rank sum test were performedfor different experimental series (details are provided in Supplementaryinformation SI. 7). A p-value of b0.05 was considered statisticallysignificant.

3. Results

3.1. Experiments in tissue mimicking phantoms

3.1.1. Passive cavitation detectionEffects of the parameters on the cavitation dosemeasured for theMB

in the tissue phantoms are shown in Fig. 3. The cavitation increasedsignificantly with increasing pressure (p b 0.001), although no signifi-cance was found between the two lowest pressures (p = 0.407).

The pulse length had negligible effects on the ICD within the testedrange (p = 0.409). The PRF had a positive influence on cavitation(p = 0.005). A PRF of 100 Hz resulted in significantly higher ICDcompared to PRFs of 10, 20 or 50 Hz (p b 0.001) while no significantdifference was found among PRFs of 10, 20, and 50 Hz (p = 0.425).

Increasing MB concentration resulted in increased ICD (p b 0.001).However, no significant difference was found between the ICDsproduced at 1 × 108/mL and 2 × 108/mL (p = 0.138), suggesting thatthe increasing trend saturated when MB concentrations exceeded1 × 108/mL.

Acoustic pressure had the most significant effect on cavitation.While increasing PRF and increasing MB concentration caused 3- to4-fold increase in ICD, increasing pressure from 0.7 to 6.9 MPa causednearly 50-fold increase in the ICD. This substantial increase in the ICDresulted from the longer duration of enhanced broadband noise withinthe entire 200-pulse exposure duration. It was observed that forpressures below or equal to 5.4 MPa, the broadband noise level washigher within the first 50 pulses and dropped to the baseline levelwhen more pulses were delivered. This suggested that the MB weredestroyed within the first 50 pulses. At the highest pressure of6.9 MPa, the broadband noise level was higher than the baseline levelthroughout the entire 200-pulse exposure. This prolonged broadbandnoise enhancement indicated a prolonged cavitation post MB destruc-tion, which was likely induced from remnants of destroyed MB. Sincethe ICD was a time integral of the broadband noise, the prolongedbroadband noise enhancement resulted in a substantial increase in theICD.

3.1.2. Active cavitation monitoringSince the most significant effect on cavitation was induced using

varying acoustic pressures, the following in vitro and in vivo experi-ments focused on effects of acoustic pressures. Representative USimages of the MB solution in the tissue phantoms after exposure to USof varying acoustic pressures are shown in Fig. 4a. The image intensityin the focal region decreased with increasing pressures (p b 0.001),suggesting more MB being destructed with increasing pressures. Aninteresting phenomenon was noted during the exposure of US atthe highest pressure, 6.9 MPa. A hyperechoic flickering cluster, likelythe cavitating MB, was seen at the focus of the transducer throughoutthe entire exposure duration (Fig. 4b).

Page 6: Journal of Controlled Release - Stanford Universitystanford.edu/group/khuri-yakub/publications/15_Wang_01.pdfloaded PLGA-PEG-NP using optimal acoustic settings with minimum tissue

3

a) b) c) d)

0 2 4 6 80

100

200

300

400

Pressure (MPa)

ICD

(V-s

)

0 5 10 150

10

20

30

40

Pulse length (cycles)

ICD

(V-s

)

0 20 40 60 80 100 1200

10

20

30

40

50

60

PRF (Hz)

ICD

(V-s

)

*

0 0.5 1 1.5 2

x 108

0

10

20

30

40

50

60

MB concentration(count/mL)

ICD

(V-s

)

Fig. 3. Inertial cavitation dose (ICD) measured in the tissue mimicking phantom by the 10-MHz passive cavitation detection transducer after varying a) pressure, b) pulse length, c) pulserepetition frequency (PRF), and d) microbubble concentration. Error bars = ±standard deviation (n = 6 each). *p b 0.001.

104 T.-Y. Wang et al. / Journal of Controlled Release 203 (2015) 99–108

3.2. In vivo delivery of model SPN nanocarriers

In the total of 41 tumors experimented in this study, no significantdifference existed in the sizes of the tumors among groups treatedwith different parameters (p = 0.66).

3.2.1. Ultrasound image monitoringThe perfusion and destruction ofMB in the tumorwere visualized on

contrast enhanced US images (Fig. 5). During the administration period,the image intensity in the tumor region increased to a peak of approxi-mately 1.6- to 3-fold higher than the baseline intensity before MBperfusion. No significant difference in the peak enhancement wasfoundamonggroups treatedwith different pressures (p=0.28). Duringthe US exposure period, the image intensity decreased and returned tothe baseline intensity at the end of the exposure. No significant differ-ence in the post-treatment image intensity was found among groupstreated with different pressures (p = 0.59). This indicated that mostMB were destroyed within the US exposure period, regardless of thepressure levels.

3.2.2. Ex vivo immunofluorescence analysisImmunofluorescence images of tumors treated with increasing US

pressures are shown in Fig. 6a. In control tumors with no US exposure,small amounts of extravasated SPN could be seen in the vicinity of thevessel area. With increasing acoustic pressures, an increasing amountof extravasated SPN was seen. Quantitative analysis of delivered SPNis summarized in Fig. 6b and c.

Both the ratios of the total area of delivered SPN to the total area ofCD31 stained tumor vessels aswell as to the total area of F-actin stainedtumor tissue significantly increased at the lowest US pressure used

a) bb)

Fig. 4. a) Representative images of microbubble solution in tissue mimicking phantom after exthe red dashed circle), likely caused by cavitating microbubbles, was seen at the focus of the(blue arrows) are artifacts from interferences of the cavitation pulses. c) The signal intensity inthat significant acoustic shadowing was caused by the concentrated microbubble solution bdisappeared, and thephantom imagebelow the cavitation chamber became visible. The focal regdestruction of microbubbles. The spatial extent of microbubble destruction was broadened wdisplayedwith a 40 dBdynamic range. Other treatment parameterswere fixed at pulse length=of the references to color in this figure legend, the reader is referred to the web version of this

(1.7 MPa) compared to no US (p = 0.025) and both ratios furtherincreased with increasing pressures (p b 0.0001; Fig. 6b and c).The ratio of the total area of SPN to the total area of CD31 increased by3.9 fold at 1.7 MPa compared to the no US control, and it furtherincreased up to 12.6-fold at 6.9 MPa. The ratio of total area of SPNto the total area of F-actin was increased by 2.5-fold at 1.7 MPa and13.8-fold at 6.9 MPa compared to no US.

3D confocal microscopy further confirmed extravasation of SPN(Fig. 7a). With increasing US pressures, the penetration depth of SPNsignificantly increased (p b 0.0001; Fig. 7b). The median penetrationdepth produced by 6.9-MPa US was approximately 3-fold higher thanthat observed without US. A maximum penetration depth of nearly45 μm was observed in the tumors treated with the highest pressure,6.9 MPa.

3.2.3. Histological examinationsUS inducedMB cavitation caused onlyminimal histologically detect-

able tissue damage, visualized as intra- and peri-tumoral hemorrhage(Fig. 8a). Hemorrhage was present in both non-treated and treatedtumors. Increasing acoustic pressures had no significant effect on theintratumoral hemorrhage grade (p = 0.48), but caused increasedperitumoral hemorrhage grade (p = 0.03). The overall hemorrhagegrades increased with increasing pressures (p = 0.04), which wasmainly attributable to the peritumoral hemorrhage (Fig. 8b).

3.3. Delivery of miR-122 encapsulated PLGA-PEG nanoparticles

Quantitative RT-PCR of miR-122 delivered into human colon cancerxenografts showed significantly enhanced levels of miR-122 in animalsadministered with miR-122 loaded PLGA-PEG-NP compared to thecontrol settings (treatments with US and MB but without injection of

Imag

ein

ten

sit

(AU

)

c)

5

10

15

Imag

ein

ten

sity

(A

..

00

000

000

000

2PPress

4sure (MP

6a)

8

posure to ultrasound at different acoustic pressures. b) Hyperechoic flickering cluster (intransducer during the exposure to ultrasound at 6.9 MPa. The hyperechoic vertical linesthe focal region, marked as red box in panel (a), was plotted against the pressures. Note

efore ultrasound exposure. As microbubbles were destroyed by ultrasound, the shadowion of the cavitation transducer appeared hypoechoic after ultrasoundexposure, indicatingith increasing pressures. Data are mean ± standard deviation (n = 4 each). Images are5 cycles, PRF=100Hz, andmicrobubble concentration=1× 108/mL. (For interpretationarticle.)

Page 7: Journal of Controlled Release - Stanford Universitystanford.edu/group/khuri-yakub/publications/15_Wang_01.pdfloaded PLGA-PEG-NP using optimal acoustic settings with minimum tissue

Fig. 5.Representative contrast-enhanced ultrasound images of a human colon cancer xenograft during a 2-minute treatment cycle. Image signal increased during the perfusion periodwithmicrobubbles floating into the tumor, and then substantially decreased during sonoporation. Microbubble destruction occurred quickly after ultrasound application. The discrete dropof image signal intensity (see images at 70 s, 80 s, and 90 s) corresponded to each steering of focal beam.

105T.-Y. Wang et al. / Journal of Controlled Release 203 (2015) 99–108

either type of PLGA-PEG-NP, and treatments with US and MB andinjection of control PLGA-PEG-NP), and was further increased whenUS and MB were applied (Fig. 9). The levels of miR-122 in tumorstreated with US were increased by an average of 1310 ± 962 fold(range: 270 to 3100 fold), while the levels of miR-122 in contralateraltumors without US were only increased by an average of 224 ±164 fold (range: 20 to 508 fold) compared to the endogenous levels ofmiR-122 in untreated animals. Treatmentswith US andMB significantlyincreased the levels of miR-122 (p = 0.004). Within an animal,the levels of miR-122 in the tumor exposed to US were on average7.9-fold higher than the contralateral tumor without exposure to US.

4. Discussion

Our study showed that FDA-approved PLGA-PEG-NP loaded withtherapeutic miR-122 can be successfully delivered into the extravascu-lar compartment of human colon cancer xenografts using an optimizedUS-guided drug delivery protocol. To our knowledge, this is the firststudy to demonstrate efficient delivery of miRs into tumors using USand MB mediated sonoporation.

a)

Af

SP

N/

fl

b)

Are

a o

fs/

area

of

vess

el(C

D31

) st

ain

ing

0

0.2

0.4

0.6

0.8

0

Fig. 6. a) Representative immunofluorescence images of human colon cancer xenografts treatethe tumor tissue with increasing pressures compared to no ultrasound (control). Note, tumor varea of SPN to the total area of CD31 vessel staining (b) and to the total area of F-actin cytoskelmean ± standard deviation (n = 5 each).

Once injected intravenously, however, miRs are quickly degradedby nucleases. Therefore, miR-122 was encapsulated into PLGA-PEG-NPto protect them fromnucleasemediated degradation in our study. How-ever, due to their size, passive accumulation of PLGA-PEG-NP throughthe EPR effect only yields in minimal accumulation in tumors [41]. Weshowed that despite their size N100 nm, miR-loaded PLGA-PEG-NPdelivery to the extravascular compartment can be successfullyenhanced in tumors through sonoporation. To quantify successful miRdelivery into the extravascular compartment only, mice were perfusedbefore excising the tumors to ensure that all circulating PLGA-PEG-NPwere cleared from the tumor vessels. Using this approach, miR-122delivery in human colon cancer xenografts was increased by on average7.9 fold compared to passive accumulation as assessed by quantitativeRT-PCR.

Using 3D confocal microscopy, we further explored the penetrationdepth of nanocarriers in relation to tumor vessels in tumors. Fourhours following intravenous administration,we found that nanocarrierspenetrated from 5 μm through passive accumulation to up to 45 μmusing sonoporation. To our knowledge, our study is the first to examinethe volumetric distribution of nanocarriers within tumors using 3Dconfocal microscopy, allowing for a more accurate measurement of

2Pressu

4ure (MPa)

6 8

cc)

Are

a o

f S

PN

s/ar

ea o

f ce

ll(F

-act

in)

stai

nin

g

0

0.1

0.2

0.3

0.4

0P2ress

4ure (MPa

6)

8

d with different ultrasound pressures show increased amount of SPN (red) delivered intoessels are stained in green and tumor cytoskeleton was stained in blue. Ratios of the totaleton staining (c) are also shown. Both ratios increased with increasing pressures. Data are

Page 8: Journal of Controlled Release - Stanford Universitystanford.edu/group/khuri-yakub/publications/15_Wang_01.pdfloaded PLGA-PEG-NP using optimal acoustic settings with minimum tissue

a) b

Pt

tid

th(µµ

m)

b)

0

10

20

30

40

50

Pen

etra

tio

n d

epth

0

0

0

0

0

0

00.0 1.7Pres

2.5ssur

3.9re (M

9Pa)

5.4)

6.9

Fig. 7. a) Representative maximum intensity confocal immunofluorescence image reconstructed from 3D confocal microscopy data sets of human colon cancer xenograft treated withultrasound and microbubbles (6.9 MPa) shows extravasated SPN (red) compared to tumor vessels (green) in the imaged volume. Orthogonal slices indicated by the gray dashed linesfurther confirmed presence of SPN in tumor parenchyma (blue) and away from the vessel lumen (contoured in green). b) Box blot shows changing tumor penetration depth with variousacoustic pressure levels. The central mark, upper edge, lower edge, and whiskers of each box indicate median, 75th, 25th percentile, and the range of 54 measurements made for eachacoustic pressure level.

106 T.-Y. Wang et al. / Journal of Controlled Release 203 (2015) 99–108

the penetration depth of extravasated nanocarriers compared to 2Dmicroscopy. While 3D confocal microscopy captures the volumetricdistribution of nanocarriers among tortuous vascular structures, 2Dmicroscopy only images a thin slice of the tissue, often overestimatingthe penetration depth because the shortest distance betweennanocarriers and the vessels may not fall on the imaging plane. Withthe volumetric information reconstructed from3Dconfocalmicroscopy,the shortest distance between nanocarriers and surrounding vesselscould be assessed in our study by projecting the nanocarriers to thenearest vessel in the 3D volume.

Another objective of our study was to assess whether sonoporationusing different acoustic pressures causes macroscopic tissue damage.For this purpose, we analyzed treated tumor tissues for the presenceof intra- and peri-tumoral hemorrhage as a biomarker of vessel destruc-tion with consecutive blood extravasation. While there was a trendtowards slightly increased hemorrhage at higher acoustic pressures,intra-tumoral hemorrhage and peri-tumoral hemorrhage were alsoobserved in non-treated control tumors; even at highest testedpressures there was overall small hemorrhage on histology. Similartrends have been reported previously [42,43]. Notably, overall increasedhemorrhagewasmainly attributable to peri-tumoral hemorrhage in ourstudy. This is likely because vessel density is often higher at the tumorperiphery than at the tumor center [44]. In addition, cavitation maypreferentially occur at an interface, such as that between the couplinggel and the superficial subcutaneous tumors used in this study [45].This interface may not apply to other tumor models where the tumorsare established in a more contiguous tissue environment, such asorthotopic tumors. Peri-tumoral hemorrhage may increase the risk of

Fig. 8. a) Representative H&E images of human colon cancer xenografts treated with different aarea) was observed in some of the treated tumors. b) Box plot shows a slightly increasing trendata points and the bars label the mean in each group. Note that there was already hemorrhag

thrombosis in the peritumoral region, producing favorable (e.g., byblocking drug dilution due to reduced blood flow) or undesired effectsin the tumor treatment (e.g., by reducing drug delivery following vesselblockage). Future studies are warranted to evaluate the occurrence andextent of hemorrhage in orthotopic tumor models deeper in the body.

Several limitations of our study need to be acknowledged. First,while real-time US imaging was used to document MB destruction, itcannot sufficiently indicate the occurrence of inertial cavitation in vivobecause the decrease of image intensity can result from several mecha-nisms, including deflation, dissolution, and/or inertial cavitation ofMB [46]. Future work will incorporate real-time passive cavitationmonitoring in our treatment platform to evaluate inertial cavitationin vivo and its correlation with the delivery outcomes. Second, therange of acoustic parameters tested for optimal drug delivery wasrestrained by the capabilities of our available US system. Therefore,the optimized parameter set found in our study may not be a universaloptimum. Third, the delivery amount and penetration depth reported inthis study were obtained in a well vascularized cancer model. Inhypovascularized cancer with different morphological characteristics,these measurements may vary. Finally, the amount of free miR-122released from PLGA-PEG-NPwas not quantified because RT-PCR cannotdifferentiate between free or still encapsulated miRs, and future studiesare warranted to assess downstream effects of released miR-122 incolon cancer following successful delivery via sonoporation.

In conclusion, our results suggest that FDA-approved PLGA-PEG-NPencapsulated with miR-122 can be successfully delivered into humancolon cancer xenografts using an optimized US and MB mediatedsonoporation platform. Guided by US imaging, this drug delivery

coustic pressures. Small areas of hemorrhage (as shown in the close-up view of the boxedd towards higher hemorrhage at higher acoustic pressures. Each dot represents individuale in non-treated tumors (n = 5 each).

Page 9: Journal of Controlled Release - Stanford Universitystanford.edu/group/khuri-yakub/publications/15_Wang_01.pdfloaded PLGA-PEG-NP using optimal acoustic settings with minimum tissue

Fig. 9. RT-PCR quantification of the fold change inmiR-122 in tumors treatedwith controlPLGA-PEG-NP (left, n = 3 each) or miR122 loaded PLGA-PEG-NP (right, n = 7 each).The miR-122 levels were expressed as fold changes relative to the endogenous levels ofmiR-122 in control animals with neither nanoparticle administration nor ultrasound andmicrobubble treatments. *p = 0.004. **p= 0.002. ***p = 0.001.

107T.-Y. Wang et al. / Journal of Controlled Release 203 (2015) 99–108

approach is safe and spatially localized to the anatomical region underconsideration. Our results pave the way towards future studiesassessing the downstream effects of delivered drugs such as miR-122in cancer.

Acknowledgments

The work is supported by NIH grants 5T32CA009695 (TYW),R01CA155289-01A1 (JKW), R01DK092509-01A1 (JKW), R01DK099800-06 (KP and JR), 1R01CA161091 (RD and RP). The authors thank P. Yehand K. Butts-Pauly at Stanford University for assistance in US pulsecalibration.

Appendix A. Supplementary data

Supplementary data to this article can be found online at http://dx.doi.org/10.1016/j.jconrel.2015.02.018.

References

[1] S. Ganta, H. Devalapally, A. Shahiwala, M. Amiji, A review of stimuli-responsivenanocarriers for drug and gene delivery, J. Control. Release 126 (2008) 187–204.

[2] H. Maeda, J. Wu, T. Sawa, Y. Matsumura, K. Hori, Tumor vascular permeability andthe EPR effect in macromolecular therapeutics: a review, J. Control. Release 65(2000) 271–284.

[3] Y.H. Bae, K. Park, Targeted drug delivery to tumors: myths, reality and possibility, J.Control. Release 153 (2011) 198–205.

[4] U. Prabhakar, H. Maeda, R.K. Jain, E.M. Sevick-Muraca, W. Zamboni, O.C. Farokhzad,S.T. Barry, A. Gabizon, P. Grodzinski, D.C. Blakey, Challenges and key considerationsof the enhanced permeability and retention effect for nanomedicine drug delivery inoncology, Cancer Res. 73 (2013) 2412–2417.

[5] F. Yuan, H.A. Salehi, Y. Boucher, U.S. Vasthare, R.F. Tuma, R.K. Jain, Vascular perme-ability and microcirculation of gliomas and mammary carcinomas transplanted inrat and mouse cranial windows, Cancer Res. 54 (1994) 4564–4568.

[6] K.J. Harrington, S. Mohammadtaghi, P.S. Uster, D. Glass, A.M. Peters, R.G. Vile, J.S.Stewart, Effective targeting of solid tumors in patients with locally advanced cancersby radiolabeled pegylated liposomes, Clin. Cancer Res. 7 (2001) 243–254.

[7] W.L. Nyborg, Ultrasonicmicrostreaming and related phenomena, Br. J. Cancer Suppl.5 (1982) 156–160.

[8] M. Postema, S. Kotopoulis, A. Delalande, O.H. Gilja, Invited editorial— Sonoporation:why microbubbles create pores, Ultraschall Med. 33 (2012) 97–98.

[9] N. Hosseinkhah, H. Chen, T.J. Matula, P.N. Burns, K. Hynynen, Mechanisms ofmicrobubble–vessel interactions and induced stresses: a numerical study, J. Acoust.Soc. Am. 134 (2013) 1875–1885.

[10] J. Collis, R. Manasseh, P. Liovic, P. Tho, A. Ooi, K. Petkovic-Duran, Y. Zhu, Cavitationmicrostreaming and stress fields created by microbubbles, Ultrasonics 50 (2010)273–279.

[11] P. Marmottant, S. Hilgenfeldt, Controlled vesicle deformation and lysis by singleoscillating bubbles, Nature 423 (2003) 153–156.

[12] P. Prentice, A. Cuschieri, K. Dholakia, M. Prausnitz, P. Campbell, Membrane disrup-tion by optically controlled microbubble cavitation, Nat. Phys. 1 (2005) 107–110.

[13] L.H. Treat, N. McDannold, Y. Zhang, N. Vykhodtseva, K. Hynynen, Improved anti-tumor effect of liposomal doxorubicin after targeted blood–brain barrier disruption

by MRI-guided focused ultrasound in rat glioma, Ultrasound Med. Biol. 38 (2012)1716–1725.

[14] Z.K. Liao, K.C. Tsai, H.T. Wang, S.H. Tseng, W.P. Deng, W.S. Chen, L.H. Hwang,Sonoporation-mediated anti-angiogenic gene transfer into muscle effectivelyregresses distant orthotopic tumors, Cancer Gene Ther. 19 (2011) 171–180.

[15] S. Kotopoulis, A. Delalande, M. Popa, V. Mamaeva, G. Dimcevski, O.H. Gilja, M.Postema, B.T. Gjertsen, E. McCormack, Sonoporation-enhanced chemotherapysignificantly reduces primary tumour burden in an orthotopic pancreatic cancerxenograft, Mol. Imaging Biol. 16 (2013) 53–62.

[16] F. Yan, L. Li, Z. Deng, Q. Jin, J. Chen, W. Yang, C.K. Yeh, J. Wu, R. Shandas, X. Liu, H.Zheng, Paclitaxel–liposome–microbubble complexes as ultrasound-triggered thera-peutic drug delivery carriers, J. Control. Release 166 (2013) 246–255.

[17] N.Y. Rapoport, A.M. Kennedy, J.E. Shea, C.L. Scaife, K.H. Nam, Controlled and targetedtumor chemotherapy by ultrasound-activated nanoemulsions/microbubbles,J. Control. Release 138 (2009) 268–276.

[18] S. Kotopoulis, G. Dimcevski, O.H. Gilja, D. Hoem, M. Postema, Treatment of humanpancreatic cancer using combined ultrasound, microbubbles, and gemcitabine: aclinical case study, Med. Phys. 40 (2013) 072902.

[19] M. Aryal, C.D. Arvanitis, P.M. Alexander, N. McDannold, Ultrasound-mediatedblood–brain barrier disruption for targeted drug delivery in the central nervoussystem, Adv. Drug Deliv. Rev. 72 (2014) 94–109.

[20] H. Chen, E.E. Konofagou, The size of blood–brain barrier opening induced by focusedultrasound is dictated by the acoustic pressure, J. Cereb. Blood Flow Metab. 34(2014) 1197–1204.

[21] A. Bao, B. Goins, R. Klipper, G. Negrete, W.T. Phillips, Direct 99mTc labelingof pegylated liposomal doxorubicin (Doxil) for pharmacokinetic and non-invasiveimaging studies, J. Pharmacol. Exp. Ther. 308 (2004) 419–425.

[22] R. Gluck, E. Walti, Biophysical validation of Epaxal Berna, a hepatitis A vaccineadjuvanted with immunopotentiating reconstituted influenza virosomes (IRIV),Dev. Biol. (Basel) 103 (2000) 189–197.

[23] A. Fahr, P. van Hoogevest, S. May, N. Bergstrand, M.L.S. Leigh, Transfer of lipophilicdrugs between liposomal membranes and biological interfaces: consequences fordrug delivery, Eur. J. Pharm. Sci. 26 (2005) 251–265.

[24] R. Garzon, G.A. Calin, C.M. Croce, MicroRNAs in cancer, Annu. Rev. Med. 60 (2009)167–179.

[25] Y. Saito, H. Suzuki, M. Matsuura, A. Sato, Y. Kasai, K. Yamada, H. Saito, T. Hibi,MicroRNAs in hepatobiliary and pancreatic cancers, Front. Genet. 2 (2011) 66.

[26] L. Ma, J. Liu, J. Shen, L. Liu, J. Wu,W. Li, J. Luo, Q. Chen, C. Qian, Expression ofmiR-122mediated by adenoviral vector induces apoptosis and cell cycle arrest of cancer cells,Cancer Biol. Ther. 9 (2010) 554–561.

[27] W.C. Tsai, P.W. Hsu, T.C. Lai, G.Y. Chau, C.W. Lin, C.M. Chen, C.D. Lin, Y.L. Liao, J.L.Wang, Y.P. Chau, M.T. Hsu, M. Hsiao, H.D. Huang, A.P. Tsou, MicroRNA-122, atumor suppressormicroRNA that regulates intrahepaticmetastasis of hepatocellularcarcinoma, Hepatology 49 (2009) 1571–1582.

[28] J. He, G. Xie, J. Tong, Y. Peng, H. Huang, J. Li, N. Wang, H. Liang, Overexpressionof microRNA-122 re-sensitizes 5-FU-resistant colon cancer cells to 5-FU throughthe inhibition of PKM2 in vitro and in vivo, Cell Biochem. Biophys. 70 (2014)1343–1350.

[29] S. Bai, M.W. Nasser, B. Wang, S.H. Hsu, J. Datta, H. Kutay, A. Yadav, G. Nuovo, P.Kumar, K. Ghoshal, MicroRNA-122 inhibits tumorigenic properties of hepatocellularcarcinoma cells and sensitizes these cells to sorafenib, J. Biol. Chem. 284 (2009)32015–32027.

[30] C. Wu, D.T. Chiu, Highly fluorescent semiconducting polymer dots for biology andmedicine, Angew. Chem. Int. Ed. Engl. 52 (2013) 3086–3109.

[31] K. Pu, A.J. Shuhendler,M.P. Valta, L. Cui,M. Saar,D.M. Peehl, J. Rao, Phosphorylcholine-coated semiconducting polymer nanoparticles as rapid and efficient labeling agentsfor in vivo cell tracking, Adv. Healthc. Mater. 3 (2014) 1292–1298.

[32] R. Devulapally, R. Paulmurugan, Polymer nanoparticles for drug and small silencingRNA delivery to treat cancers of different phenotypes, Wiley Interdiscip. Rev.Nanomed. Nanobiotechnol. 6 (2013) 40–60.

[33] B. Wang, H. Wang, Z. Yang, MiR-122 inhibits cell proliferation and tumorigenesis ofbreast cancer by targeting IGF1R, PLoS One 7 (2012) e47053.

[34] M. Schneider, B. Anantharam, M. Arditi, D. Bokor, A. Broillet, P. Bussat, X. Fouillet, P.Frinking, I. Tardy, J. Terrettaz, R. Senior, F. Tranquart, BR38, a new ultrasound bloodpool agent, Investig. Radiol. 46 (2011) 486–494.

[35] R. Bekeredjian, S. Chen, P.A. Frenkel, P.A. Grayburn, R.V. Shohet, Ultrasound-targetedmicrobubble destruction can repeatedly direct highly specific plasmid expression tothe heart, Circulation 108 (2003) 1022–1026.

[36] A.C. Riches, J.G. Sharp, D.B. Thomas, S.V. Smith, Blood volume determination in themouse, J. Physiol. 228 (1973) 279–284.

[37] C.M. Panje, D.S. Wang, M.A. Pysz, R. Paulmurugan, Y. Ren, F. Tranquart, L. Tian, J.K.Willmann, Ultrasound-mediated gene delivery with cationic versus neutralmicrobubbles: effect of DNA andmicrobubble dose on in vivo transfection efficiency,Theranostics 2 (2012) 1078–1091.

[38] A. Needles, M. Arditi, N.G. Rognin, J. Mehi, T. Coulthard, C. Bilan-Tracey, E. Gaud, P.Frinking, D. Hirson, F.S. Foster, Nonlinear contrast imaging with an array-basedmicro-ultrasound system, Ultrasound Med. Biol. 36 (2010) 2097–2106.

[39] S. Eggen, M. Afadzi, E.A. Nilssen, S.B. Haugstad, B. Angelsen, L. Davies Cde, Ultra-sound improves the uptake and distribution of liposomal Doxorubicin in prostatecancer xenografts, Ultrasound Med. Biol. 39 (2013) 1255–1266.

[40] R.D. Lillie, Histopathologic Technic and Practical Histochemistry, The BlackistonCompany, Inc., New York, 1954.

[41] H. Cabral, Y. Matsumoto, K. Mizuno, Q. Chen, M. Murakami, M. Kimura, Y. Terada,M.R. Kano, K. Miyazono, M. Uesaka, N. Nishiyama, K. Kataoka, Accumulation ofsub-100 nm polymeric micelles in poorly permeable tumours depends on size,Nat. Nanotechnol. 6 (2011) 815–823.

Page 10: Journal of Controlled Release - Stanford Universitystanford.edu/group/khuri-yakub/publications/15_Wang_01.pdfloaded PLGA-PEG-NP using optimal acoustic settings with minimum tissue

108 T.-Y. Wang et al. / Journal of Controlled Release 203 (2015) 99–108

[42] A. Xie, T. Belcik, Y. Qi, T.K. Morgan, S.A. Champaneri, S. Taylor, B.P. Davidson, Y. Zhao,A.L. Klibanov, M.A. Kuliszewski, H. Leong-Poi, A. Ammi, J.R. Lindner, Ultrasound-mediated vascular gene transfection by cavitation of endothelial-targeted cationicmicrobubbles, JACC Cardiovasc Imaging 5 (2012) 1253–1262.

[43] J.H. Hwang, A.A. Brayman, M.A. Reidy, T.J. Matula, M.B. Kimmey, L.A. Crum, Vasculareffects induced by combined 1-MHz ultrasound and microbubble contrast agenttreatments in vivo, Ultrasound Med. Biol. 31 (2005) 553–564.

[44] J. Holash, P.C. Maisonpierre, D. Compton, P. Boland, C.R. Alexander, D. Zagzag, G.D.Yancopoulos, S.J. Wiegand, Vessel cooption, regression, and growth in tumorsmediated by angiopoietins and VEGF, Science 284 (1999) 1994–1998.

[45] L.A. Frizzell, Threshold dosages for damage to mammalian liver by high intensityfocused ultrasound, IEEE Trans. Ultrason. Ferroelectr. Freq. Control 35 (1988)578–581.

[46] M.A. Borden, D.E. Kruse, C.F. Caskey, S. Zhao, P.A. Dayton, K.W. Ferrara, Influenceof lipid shell physicochemical properties on ultrasound-induced microbubbledestruction, IEEE Trans. Ultrason. Ferroelectr. Freq. Control 52 (2005) 1992–2002.