• Aucun résultat trouvé

Hybrid quantum dot−fatty ester stealth nanoparticles: toward clinically relevant in vivo optical imaging of deep tissue

N/A
N/A
Protected

Academic year: 2021

Partager "Hybrid quantum dot−fatty ester stealth nanoparticles: toward clinically relevant in vivo optical imaging of deep tissue"

Copied!
10
0
0

Texte intégral

(1)

Publisher’s version / Version de l'éditeur:

ACS Nano, 5, 3, pp. 1958-1966, 2011-02-21

READ THESE TERMS AND CONDITIONS CAREFULLY BEFORE USING THIS WEBSITE. https://nrc-publications.canada.ca/eng/copyright

Vous avez des questions? Nous pouvons vous aider. Pour communiquer directement avec un auteur, consultez la première page de la revue dans laquelle son article a été publié afin de trouver ses coordonnées. Si vous n’arrivez pas à les repérer, communiquez avec nous à PublicationsArchive-ArchivesPublications@nrc-cnrc.gc.ca.

Questions? Contact the NRC Publications Archive team at

PublicationsArchive-ArchivesPublications@nrc-cnrc.gc.ca. If you wish to email the authors directly, please see the first page of the publication for their contact information.

NRC Publications Archive

Archives des publications du CNRC

This publication could be one of several versions: author’s original, accepted manuscript or the publisher’s version. / La version de cette publication peut être l’une des suivantes : la version prépublication de l’auteur, la version acceptée du manuscrit ou la version de l’éditeur.

For the publisher’s version, please access the DOI link below./ Pour consulter la version de l’éditeur, utilisez le lien DOI ci-dessous.

https://doi.org/10.1021/nn103024b

Access and use of this website and the material on it are subject to the Terms and Conditions set forth at

Hybrid quantum dot−fatty ester stealth nanoparticles: toward clinically

relevant in vivo optical imaging of deep tissue

Shuhendler, Adam J.; Prasad, Preethy; Chan, Ho-Ka Carol; Gordijo, Claudia

R.; Saroushian, Behrouz; Kolios, Michael; Yu, Kui; O’Brien, Peter J.; Rauth,

Andrew Michael; Wu, Xiao Yu

https://publications-cnrc.canada.ca/fra/droits

L’accès à ce site Web et l’utilisation de son contenu sont assujettis aux conditions présentées dans le site LISEZ CES CONDITIONS ATTENTIVEMENT AVANT D’UTILISER CE SITE WEB.

NRC Publications Record / Notice d'Archives des publications de CNRC:

https://nrc-publications.canada.ca/eng/view/object/?id=cffc700c-eb51-4d91-86d0-31cc0a110398

https://publications-cnrc.canada.ca/fra/voir/objet/?id=cffc700c-eb51-4d91-86d0-31cc0a110398

(2)

1

Hybrid Quantum Dot-Fatty Ester

2

Stealth Nanoparticles: Toward

3

Clinically Relevant in Vivo Optical

4

Imaging of Deep Tissue

5

Adam J. Shuhendler,

Preethy Prasad,

Ho-Ka Carol Chan,

Claudia R. Gordijo,

Behrouz Soroushian,

6

Michael Kolios,

Kui Yu,

§

Peter J. O’Brien,

Andrew Michael Rauth,

^,

* and Xiao Yu Wu

†,

*

7

Leslie L. Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, Ontario, Canada, M5S 3M2,

Department of Physics, Ryerson University, 350

8

Victoria Street, Toronto, Ontario, Canada, M5B 2K3,

§

NRC Steacie Institute for Molecular Sciences, National Research Council of Canada, 100 Sussex Drive, Ottawa,

9

Ontario, Canada, K1A 0R6, and

^

Department of Medical Biophysics, University Health Network, Princess Margaret Hospital, 610 University Avenue, Toronto, Ontario,

10

Canada, M5G 2M9

11

12

M

edical imaging has become a

sta-13

ple in a clinician's arsenal for the

14

detection and treatment of

hu-15

man disease, especially for cancers.

1

Current

16

imaging modalities applied to oncology,

17

such as magnetic resonance imaging and

18

positron emission tomography (PET),

re-19

quire expensive instrumentation and long

20

subject exposure times. In the case of PET,

21

potentially genotoxic radioisotope contrast

22

agents are employed.

2

In contrast,

fluores-23

cent imaging methods utilize nonionizing

24

radiation are relatively less expensive and

25

less hazardous, and can be performed

26

quickly with brief exposure times. However,

27

traditional fluorescent imaging in medicine

28

has been severely limited by light scattering

29

and autofluorescence of tissue in the

ultra-30

violet and visible light range.

3,4

Hence images

31

have only been acquired at depths of a few

32

millimeters below the surface of the skin.

3,4

In

33

the near-infrared (NIR) wavelength range of

34

650-900 nm, the light scattering,

absor-35

bance, and autofluorescene of tissue are at

36

a minimum, providing an opportunity for

37

deeper optical interrogations in vivo with a

38

predicted tissue penetration depth of 1-2

39

cm (Figure 1

F1

).

4,5

Whether or not fluorescent

40

medical imaging can become of broad use

41

in clinical practice now depends on the

42

development of a safe, biocompatible

fluoro-43

phore with excitation and emission

wave-44

lengths in the NIR.

45

Ideally, an NIR fluorophore for medical

46

applications should have a large absorption

47

exctinction coefficient, high NIR quantum

48

yield, low photobleaching, and no toxicity.

6 49

Organic fluorophores are poor candidates for

50

deep fluorescent medical imaging because of

51 52 53 54 55 56 57 58 59 60 61 62 63 64 65 66 67 68 69 70 71

72

their poor circulation half-lives, poor NIR

73

quantum yields, rapid photobleaching, and

74

spectral shifting due to rapid plasma protein

75

binding, metabolism, and/or chemical

76

degradation.

6-8

For this reason, NIR-tuned

77

semiconductor quantum dots (QD) have

78

been developed and applied to medical

imag-79

ing for sentinal lymph node mapping

9-11 80

and tumor imaging.

12,13

The properties of

81

QDs that render them effective

fluoropho-82

res for medical fluorescence imaging stem

83

*Address correspondence to xywu@phm.utoronto.ca.

Received for review November 8, 2010 and accepted December 27, 2010. Published online

10.1021/nn103024b

CXXXX American Chemical Society

ABSTRACT

Despite broad applications of quantum dots (QDs) in vitro, severe toxicity and

dominant liver uptake have limited their clinical application. QDs that excite and emit in the

ultraviolet and visible regions have limited in vivo applicability due to significant optical interference

exerted by biological fluids and tissues. Hence we devised a new biocompatible hybrid fluorophore

composed of near-infrared-emitting PbSe quantum dots encapsulated in solid fatty ester

nanoparticles (QD-FEN) for in vivo imaging. The quantum yield and tissue penetration depth of

the QD-FEN were characterized, and their biological fate was examined in a breast tumor-bearing

animal model. It was found for the first time that chemical modification of the headgroup of

QD-encapsulating organic fatty acids was a must as these groups quenched the photoluminescence of

PbSe nanocrystals. The use of fatty esters enhanced aqueous quantum yields of PbSe QDs up to

∼45%, which was 50% higher than that of water-soluble PbSe nanocrystals in an aqueous medium.

As a result, a greater than previously reported tissue penetration depth of fluorescence was recorded

at 710 nm/860 nm excitation/emission wavelengths. The QD-FEN had much lower short-term

cytotoxicity compared to nonencapsulated water-soluble QDs. More importantly, reduced liver

uptake, tumor retention, lack of toxic response, and nearly complete clearance of QD-FEN from the

tested animals was demonstrated. With a combination of near-infrared spectral properties,

enhanced optical properties,and significantly improved biosafety profile, this novel hybrid

nanoparticulate fluorophore system demonstrably provides real-time, deep-tissue fluorescent

imaging of live animals, laying a foundation for further development toward clinical application.

KEYWORDS: hybrid quantum dot-fatty ester nanoparticles• high biocompatibility •

deep tissue penetration • near-infrared whole-body imaging • breast tumor

model

ARTICLE

ACS Nano | 3b2 | ver.9 | 5/1/011 | 10:53 | Msc: nn-2010-03024b | TEID: tes00 | BATID: 00000 | Pages: 8.16

(3)

84

from their high quantum yields with NIR excitation and

85

their large effective Stoke's shifts, resulting in the

86

potential for optical image acquisition at greater tissue

87

depths.

3,14,15

Currently, however, the major hurdle in

88

the clinical application of QDs as medical-grade

fluor-89

ophores is their toxicity,

16

attributable to the heavy

90

metal precursors required for their synthesis,

17

the

91

ligands employed for their surface functionalization,

18

92

and their extensive liver uptake and poor clearance

93

rates that result in unsafe pharmacokinetic profiles.

19,20 94

By mitigating the toxicity issues of QDs through

im-95

provement of their pharmacokinetic profiles while

96

maintaining their superior spectral properties in the

97

NIR region, an inorganic QD-organic fatty ester

nano-98

particle hybrid fluorophore has been formulated.

99

These particles may be well suited for medical imaging

100

applications, advancing the field of fluorescence

med-101

ical imaging.

102

RESULTS AND DISCUSSION

103

This paper describes the formulation and

character-104

ization of quantum dots encapsulated in fatty ester

105

nanoparticles. The novel nanocarrier system can

effec-106

tively encapsulate NIR PbSe QDs resulting in lower in

107

vitro

cytotoxicity, improved in vivo biodistribution, and

108

clearance of the semiconductor nanocrystals, and

en-109

hanced aqueous QD optical properties resulting in

110

deep-tissue imaging capabilities. While QDs emitting

111

in the visible range have previously been encapsulated

112

in lipid-based nanoparticles,

21

this paper presents the

113

first encapsulation of QDs with clinically relevant

op-114

tical properties in stealth, fatty ester-based

nanoparti-115

cles, the first demonstration of improved aqueous

116

quantum yield of QDs, and a vast improvement of

117

imaging depth, resulting in significant progress toward

118

enhanced fluorophore biocompatibility and utility.

119

Formulation of QD-FEN.

Lead-selenide (PbSe)

nano-120

crystals were employed for the formulation of QD-FENs

121

due to their relative ease of synthesis, the tunability of

122

their emisson within the NIR range, their high resultant

123

quantum yields with NIR excitation, and the demonstrated

124

photostability of this semiconductor system.

14,15,22

The

125

synthesis of PbSe QDs was based on a room

tempera-126

ture scheme under ambient atmospheric conditions

127

previously published, resulting in hydrophobic, oleic

128

acid-capped nanocrystals, ∼7 nm in diameter that

129

were suspended in hexanes.

15

The formulated

QD-130

loaded fatty acid or fatty ester nanoparticles were

131

110 ( 15 nm in hydrodynamic diameter, as measured

132

by dynamic light scattering, with the PbSe QD being

133

trapped within the now solid matrix (Figure 2

A). The

F2 134

measured zeta potential (ζ = -20 ( 1.3 mV) was

135

favorable to both a stable emulsion and to good

136

biocompatibility. The Myrj block copolymers used in

137

the formulation are essential to both stabilize the

138

molten nanoemulsion droplets and coat the surface

139

of the solidified nanoparticles with poly(ethylene

140

glycol) chains, imparting enhanced biocompatilibity

141

to the QD-FENs (Figure 2A). The coating of

nanoparti-142

cles with a poly(ethylene glycol) corona has previously

143

been shown to impart a stealth property to liposomal

144

formulations, evading the recognition and uptake by

145

the reticuloendothelial system thus extending the

146

circulation time of the particles in the body.

23

Under

147

electron microscopic imaging, the PbSe QDs were

148

clearly seen as black spots within the matrix of the

149

QD-FENs (Figure 2B). The electron density

character-150

istic of metal particles was absent in electron

micro-151

graphs of QD-free fatty ester nanoparticles (Figure 2C).

152

Spectral Characteristics of QD-FEN.

The PbSe QDs in our

153

hands showed an emission maximum at 860 nm

154

(Figure 3

A) and a quantum yield in hexanes of ∼15%

F3 155

at an excitation wavelength (λ

ex

) of 710 nm, as

deter-156

mined using 1,1

0

-diethyl-4,4

0

-carbocyanine iodide as

157

the quantum yield standard (λ

ex

= 710 nm, λ

em

= 735

158

nm, quantum yield =3.6%

24

). Suprisingly, when PbSe

159

QDs were encapsulated in myristic acid-based

nano-160

particles, near complete quenching of the QD emission

161

was observed (Figure 3A). Initially, this novel QD

162

quenching phenomenon was attributed to either the

163

physical transition of the QD environment from a liquid

164

to a solid phase as the nanoemulsion was cooled and

165

the fatty droplets were solidified, or to the presence of

166

the carboxylic acid headgroups of the fatty acid chains.

167

To investigate each potential cause separately, PbSe

168

QDs were synthesized and suspended in n-octanol or

169

1-octanoic acid, and the fluorescence emission spectra

170

were recorded (Figure 3B). Since both dispersing

me-171

dia were liquid at room temperature, any fluorescence

172

quenching would only be due to the presence of the

173

carboxylate of the octanoic acid and not the physical

174

phase of the medium itself. The photoluminescence

175

curves obtained provided strong evidence that the

176

fluorescence quenching of the QDs by the myristic acid

177

nanoparticles was due to the carboxylate groups of the

178

fatty acid matrix. To mask the carboxylate groups of the

179

fatty acid matrix, fatty acid ethyl esters of eicosanoic acid

Figure 1. Schematic representation of the utility of fluoro-phores in the visible (green, yellow) versus the near-infrared (dark red) for deep fluorescence imaging. In the visible region, due to light scattering and absorption by living tissue, poor penetration of the excitation photon into the skin (green) or the penetration of the emission photon out of the skin (yellow) prevents the detection of the fluoro-phore signal in vivo. Photons in the near-infrared are cap-able of deep tissue penetration.

ARTICLE

(4)

180

were synthesized according to previously published

181

methods

25

and QD-FENs were formulated. The return

182

of the strong photoluminescence at 860 nm was

183

observed (Figure 3A), providing further evidence that

184

the previously observed QD fluorescence quenching

185

was due to the interaction of the carboxylate groups of

186

the fatty acid with the surface of the PbSe QDs and not

187

due to the liquid-to-solid phase transition of the

188

nanoparticulate carrier matrix.

189

Unexpectedly with the use of ethyl eicosanoate as

190

the carrier matrix, the quantum yield of QD-FENs

191

suspended in water increased 3-fold over free PbSe

192

QDs suspended in hexanes, from ∼15% to ∼45%,

193

determined at λ

ex

= 710 nm with 1,1

0

-diethyl-4,4

0

-194

carbocyanine iodide as the quantum yield standard.

195

The previously published quantum yield for

water-196

soluble PbSe QDs stabilized by

(1-mercaptoundec-11-197

yl)tetra(ethylene glycol) (MTPEG) was 30%,

15

making

198

our QD-FEN formulation 50% more efficient, spectrally,

199

than the free water-soluble QDs. The enhanced

fluor-200

escence efficiency of QD-FENs can be ascribed to the

201

protection of QDs in a hydrophobic nanoenvironment

202

of fatty ester matrix. The preparation of water-soluble

203

QDs involves exchange of hydrophobic ligand with

204

hydrophilic ones and direct dispersion of QDs in

aqu-205

eous medium, which have previously been shown to

206

decrease the quantum yield of PbSe QDs.

15

In our

207

formulation the PbSe QDs are maintained in their oleic

208

acid cap and encapsulated in the hydrophobic matrix

209

of QD-FENs. Thus they are never in direct contact with

210

the aqueous medium, leading to high quantum yields.

211

Imaging Depth of Penetration of QD-FEN.

One limitation

212

of medical fluorescence imaging is the shallow

dis-213

tance of image acquisition, often to below 10 mm from

214

the surface of the skin.

6

To quantitatively measure the

Figure 2. PbSe QDs are dispersed throughout the matrix of QD-FENs, which bear a corona of poly(ethylene glycol), as shown in the schematic A. Transmission electron micro-graphs of QD-FENs (B) and fatty ester nanoparticles (C) clearly demonstrate the encapsulation of the metal QDs within the fatty matrix. Transmission electron micrograph of PbSe QDs on their own is shown in the top right inset of panel B. Scale bars in panels B and C are 100 nm, scales bars in insets of panel B are 10 nm.

Figure 3. (A) The photoluminescence quenching of PbSe QDs encapsulated in solid fatty acid nanoparticles is clear by comparing the emission spectra of free PbSe QDs (red) to the emission of QD-loaded myristate nanoparticles (blue), as is the reversal of this quenching effect with QD-FENs (green). (B) The appearance of this quenching effect when QDs were dispersed in 1-octanoic acid (blue) but not in n-octanol (red) suggests that the hydrophobic fatty acid moiety of the fatty carrier matrix as the cause of the QD signal attenuation.

ARTICLE

(5)

215

depth of image acquisition possible using QD-FENs, a

216

novel system for the continuous measurement of

217

biologically relevant imaging depth was developed

218

and implemented (Figure 4

F4

). Gelatin tissue phantoms

219

were created, mimicking the absorbance and

scatter-220

ing characterisitcs of living tissue as previously

report-221

ed.

26

Gelatin, hemoglobin, sodium azide, and

Intrali-222

pid were mixed at elevated temperatures and were

223

poured into a prechilled plastic mold tilted at an ∼45

224

angle (Figure 4A). A 1-mm thick gelatin slab was cast

225

containing the desired dilution of QD-FENs, and was

226

laid on top of the bottom portion of the set

gela-227

tin-hemoglobin-Intralipid phantom. The mold

con-228

taining the diagonal phantom bottom and the

229

encapsulated QD slab was set flat on a surface and

230

was slowly filled with more

gelatin-hemoglobin-Int-231

ralipid mixture heated to 40 C. The addition of warm

232

phantom mixture slightly melted the hardened

phan-233

tom, preventing the formation of a phantom-phantom

234

interface to yield a single solid phantom mass (Figure 4A).

235

Measures of the distance a photon could travel in the

236

phantom before the photon was absorbed (μ

a

) or

237

scattered (μ

s

) were performed on 1-mm thick samples

238

of the tissue phantom using two integrating spheres in

239

series to prevent cross talk between absorption and

240

scattering measurements (Figure 4B), and were found

241

to be in agreement with the same parameters of real

242

tissue.

26

The 1-mm thick slab containing the QD-FENs

243

continuously and nearly linearly decreased in depth

244

from one side of the phantom to the other, permitting

245

the quantification of fluorescence signal strength as a

246

function of phantom depth from a single fluorescent

247

image acquired with a Xenogen IVIS Spectrum whole

248

animal imager (Figure 4C). Quantification of the signal

249

intensity versus depth was performed with ImageJ,

27

250

yielding the plot of signal intensity versus penetration

251

depth in Figure 4D. From this plot, it can be seen that the

252

QD-FENs showed strong signal penetration beyond 20

Figure 4. Quantitative, continuous measurement of fluorophore signal tissue depth penetration in a tissue phantom model. (A) Schematic diagram of the tissue phantom constructed for depth penetration measurements, showing the 1-mm thick gel (red) containing QD-FENs encased at an angle within the tissue-like phantom (brown). The depth measurements account for excitation photon (λex, orange) and emission photon (λem, red) penetration. (B) The light scattering and light absorption

characteristics of the tissue phantom characterized by the scattering (μs, dotted) and absorption (μa, solid) coefficients,

respectively, as measured by two integrating spheres in series. (C) Fluorescent images taken from the top of a 1.5 cm (1 mg QD-FEN/mL, top) and 2.7 cm (5 mg QD-FEN/mL, bottom) maximal QD-FEN gel depth phantom, illustrating

disappearance of detectable signal as a function of gel depth. (D) A plot of the pixel intensity versus tissue phantom depth of the phantoms depicted in Figure 4C, quantifying the maximal penetration of the 1 mg QD-FEN/mL gel to ∼1.3 cm (solid) and of the 5 mg QD-FEN/mL gel to ∼2.4 cm (dotted). Backgound fluorescence levels are indicated by a solid red line for the 1 mg QD-FEN/mL phantom and a dotted red line for the 5 mg QD-FEN/mL phantom.

ARTICLE

(6)

253

mm, reaching nearly 25 mm in depth with 5 mg

QD-254

FENs/mL phantom containing a total of 1.6 mg of PbSe

255

QDs. Previous quantitative investigations of the

pene-256

tration depth of fluorescence of NIR fluorophores have

257

reported limits up to 9.7 mm,

26

less than half of the

258

penetration depth of the QD-FENs. By increasing the

259

quantum yield of the encapsulated QDs, it is

reason-260

able to postulate that this depth of penetration may be

261

significantly extended.

262

Toxicity Assessment of QD-FEN.

The main limitation

263

preventing the application of semiconductor

nano-264

crystals to medical fluorescent imaging is the toxicity

265

arising from their heavy metal constituents,

17

their

266

surface capping and functionalizing agents,

18

and their

267

poor pharmacokinetic performance,

19

for example,

268

accumulating mainly in the liver without being

269

cleared.

20

In the case of PbSe QDs, the well-known

270

toxicity of Pb

28

and the potential toxicity of Se at high

271

doses

29

limit the biomedical applications of this

nano-272

crystal in its free form. As a preliminary assessment of

273

the safety of FEN for in vivo use, the toxicity of

QD-274

FENs was assessed against water-soluble

MTPEG-275

capped PbSe QDs using isolated rat hepatocytes. This

276

model has been used for rapid toxicity screening and

277

has demonstrated in vitro-in vivo toxicity

extrapola-278

tion.

30

Water-soluble, MTPEG-PbSe QDs showed

sig-279

nificant hepatocyte toxicity, resulting in less than 40%

280

hepatocyte survival upon exposure to 6 μg/mL of PbSe

281

QDs after 120 min (Figure 5

F5

). Exposure of hepatocytes

282

to the same dose of PbSe QDs in QD-FENs for 120 min

283

resulted in the survival of 80% of hepatocytes. Even

284

after exposure to 30 μg/mL of QD-FENs for 120 min,

285

approximately 70% of hepatocytes still survived. The

286

fatty ester nanoparticle itself has been shown to be

287

biocompatible in vitro

31

and in vivo,

32

a result that was

288

confirmed with the hepatocyte toxicity screening assay

289

presented herein. The reduced toxicity of QD-FENs

290

may arise from the mitigation of two processes

in-291

volved in QD toxicity: the loss of free heavy metal ions

292

from QD degradation

17

and the aggregation of the

293

QDs in tissue due to loss of the water-soluble capping

294

ligand from the nanocrystal surface.

6,18

The

degrada-295

tion of the nanocrystal into its free metal ions is

296

thought to occur in the endolysosomal compartment

297

of the cells following QD uptake, a compartment that is

298

acidic and promotes metal ion loss. Since PbSe QDs are

299

trapped within the solid matrix core of the fatty ester

300

carrier (Figure 2), the endolysosomal acidification is

301

unlikely to reach the fatty ester encased QDs, resulting

302

in maintenance of structural integrity and prevention

303

of free Pb

leaching. Since the PbSe QDs in their

304

hydrophobic, oleic acid capped state favorably interact

305

with the hydrophobic core of the fatty ester

nanopar-306

ticles that are stabilized by PEG and surfactant, their

307

aggregation becomes moot. As a result, the

encapsula-308

tion of PbSe QDs in the unique hydrophobic,

biocom-309

patible matrix of fatty ester nanoparticles significantly

310

reduces the toxicity of PbSe QDs in vitro. The low acute

311

toxicity of QD-FEN observed in this preliminary test is

312

encouraging suggesting the enhanced

biocompatibil-313

ity of PbSe QDs. However further assessment of in vivo

314

end points of PbSe QD toxicity in more detail is

315

required to establish the safety profile of QD-FEN.

316

In Vivo

Whole Animal Imaging With QD-FEN.

To both

317

demonstrate the utility of QD-FENs for deep tissue,

318

whole animal fluorescent imaging, and preliminarily

319

assess the in vivo fate and tolerability of this

formula-320

tion, 200 μL of QD-FENs in saline suspension (64 μg of

321

PbSe QD total) was injected intravenously in the tail

322

vein of nude mice (Figure 6

A). The mice were inocu-

F6 323

lated in the inguinal mammary fat pad with a human

324

breast tumor cell line (MDA435) stably expressing

325

green fluorescent protein. The subiliac lymph node

326

was used as a landmark for tumor inoculation in the

327

mammary fat pad. The tumor can be seen in the

328

leftmost panel of Figure 6A after 3 weeks of growth

329

using whole animal fluorescent imaging. At this point

330

the QD-FENs were injected intravenously. Mice were

331

imaged intact for 7 days to determine both the

dis-332

tribution of the fluorescent hybrid nanoparticle, as well

333

as the clearance of the fluorophore over time.

Back-334

ground fluorescence can be seen in the “background”

335

panel of Figure 6A, with mostly intestinal background

336

due to porphyrins in the mouse chow. Following the

337

intravenous injection of QD-FENs, clear, bright

fluores-338

cence can be seen. Within the first 2 h of

administra-339

tion, broad fluorescence was observed throughout the

340

animal with bright spots in the abdominal region.

Figure 5. The toxicity of QD-FENs or PbSe QDs in their free, water-soluble form was assessed in vitro in a rat hepatocyte assay for accelerated assessment of cytotoxicity. Saline, fat-ty ester nanoparticles, QD-FENS, and water-soluble QDs (M-TPEG-PbSe QD) were incubated at a low (60μg) and high (300μg) dose in 10 mL of cell suspension, and cytotoxicity was assessed at 30 min (black), 60 min (dark gray), and 120 min (light gray). Statistical comparisons of survival data of QD-FEN and MTPEG-PbSe QD against fatty ester nanopar-ticles for each dosing level were performed using a Nested ANOVA and posthoc Tukey's Test. (†) Significant differences (p < 0.05) in survival rates between low dose fatty ester nano-particles, and QD-FEN or MTPEG-PbSe QD. (††) Significant differences (p < 0.05) in survival rates between low dose QD-FEN and MTPEG-PbSe QD. (/) Significant differences (p < 0.05) in survival rates between high dose fatty ester nano-particles and QD-FEN or MTPEG-PbSe QD. (//) Significant differences (p < 0.05) in survival rates between high dose QD-FEN and MTPEG-PbSe QD. Data represent the mean and standard deviation of three trials.

ARTICLE

(7)

341

Suprisingly, none of the abdominal fluorescence arose

342

from the liver, which is the main site of uptake of free,

343

water-soluble QDs and the main site of QD retention

344

that results in the poor biocompatibility of the

flu-345

orophore.

20

From 4 to 8 h after injection, the QD-FENs

346

were seen to accumulate in the spleen, and this

acc-347

umulation could be seen to decrease to nearly

pre-348

injection levels by 96 h after injection. Within 7 days

349

(168 h), the whole animal fluorescence decreased to

350

nearly background levels, providing preliminary

evi-351

dence that the encapsulation of PbSe QDs in fatty ester

352

nanoparticles can enhance the biocompatibility of QDs

353

by enabling near complete clearance of the

fluoro-354

phore from the body. This is in contrast to the

applica-355

tion of free QDs in vivo, as long-term retention and

356

poor clearance have been noted to be the major cause

357

of concern regarding the application of semiconductor

358

nanocrystals in a clinical setting.

19,20,33

By zooming in

359

on the mammary fat pad area of the animal, tumor

360

fluorescence above tissue background could be noted

361

by 1 h (60 min) after QD-FEN injection, indicating the

362

passive accumulation of QD-FENs within the tumor

363

tissue (Figure 6B). A post-mortem examination of nude

364

mice 3 h after injection with saline, 3 h after QD-FEN

in-365

jection, and 7 days following QD-FEN injection

re-366

vealed accumulation of the QD-FENs primarily in the

367

spleen and gall bladder at 3 h, with some accumulation

368

in the kidneys (Figure 6C). The renal accumulation

de-369

creased to background and the QD-FENs were almost

370

completely cleared from the spleen and gall bladder by

371

7 days. No hepatic, heart, or lung accumulation was

372

noted even at 3 h after injection, further demonstrating

373

that the danger of hepatic uptake of the PbSe QDs was

374

mitigated by encapsulation in the solid fatty ester

375

nanoparticles. Of note is confirmation of the passive

376

accumulation of the QD-FENs in the tumor by 3 h after

377

injection and the retention of some of the QD-FENs

378

even at 7 days after administration. This preliminary

379

assessment of the novel nanocomposite QD-FEN

de-380

monstrates the synergy of enhanced biocompatibility

381

through optimized tissue distribution and clearance

382

with enhanced aqueous optical efficiency, resulting in

383

an advancement in the applicability of QDs to deep

384

tissue, macroscopic, fluorescent imaging. A

shortcom-385

ing of nontomographic fluorescence imaging

em-386

ployed in this work is the inability to distinguish the

387

precise depth profile of the QD-FEN in vivo. To precisely

388

locate the QD-FEN in three-dimensional space in an

389

animal, further studies are in progress using

fluores-390

cence molecular tomography. In addition, an in depth

391

assessment of the retention of Pb

in body tissues and

392

longer term tissue toxicity following intravenous

ad-393

ministration of QD-FENs will need to follow. The results

394

of this study will have a large impact on the possible

395

application of NIR QDs to medical fluorescent imaging

396

applications.

397

CONCLUSIONS

398

Noninvasive, nonionizing, and low cost deep

fluor-399

escent imaging can provide rapid, high spatial and

400

temporal resolution images and may present a new

Figure 6. The formulation of QD-FENs provides for deep, real-time, in vivo fluorescence imaging following intravenous administration, providing favorable biodistribution and clearance of the nanocrystal cargo. (A) The biodistribution of QD-FENs in a green fluorescent protein-expressing MDA435 model of human breast cancer was followed over 7 days with whole animal fluorescent imaging: (right) fluorescent image of the breast tumor; (left) longitudinal biodistribution and elimination of the QD-FENs. (B) Tumor accumulation of the QD-FENs was noted by 30 min after intravenous administration. (C) Necropsy analysis of the tissue distribution showed significant accumulation of QD-FENs in the spleen and gall bladder, without accumulation in the liver after 3 h. Near complete elimination of the QD-FENs was observed 7 days after

administration: GB, gall bladder; H, heart, K, kidney; Li, liver; Lu, lung; S, spleen; T, tumor.

ARTICLE

(8)

401

frontier in medical imaging modalities. While the work

402

presented here uses single photon excitation for whole

403

animal fluorescence imaging, innovation in

multipho-404

ton imaging instrumentation continues to advance the

405

possible applicability of this modality. Similar

innova-406

tive strides need to be made in the development of

407

biocompatible NIR fluorophores for deep tissue

ima-408

ging so that, in combination with instrumentation

409

innovation, fluorescent medical imaging may become

410

a reality. Here we present the formulation of a novel

411

nanocomposite fluorophore of PbSe QDs

encapsu-412

lated in a stealth fatty ester nanoparticle carrier. Along

413

with demonstrated enhanced spectral efficiency and

414

deeper tissue imaging capability, this system boasts an

415

improved safety profile that contributes to the

mitiga-416

tion of concerns regarding the in vivo use of

water-417

soluble semiconductor nanocrystals. While the

liver-418

sparing biodistribution and more complete

elimina-419

tion results are preliminary, work is in progress to

420

quantify these important characteristics of this NIR

421

nanocomposite fluorophore.

422

423

424

METHODS

425 Myristic acid, eicosanoic acid, poly(ethylene

oxide)-40-stea-426 rate (Myrj 52), poly(ethylene glycol)-100-stearate (Myrj 56), lead

427 oxide, selenium, trioctylphosphine, oleic acid, octadecene,

hex-428 anes, 1,10

-diethyl-4,40

-carbocyanine iodide, methanol, ethanol,

429 chloroform, octanoic acid, octanol,

(1-mercaptoundec-11-yl)tet-430 ra(ethylene glycol), hemoglobin, Intralipid, Trizma base, sodium

431 chloride, cupric acetate, butanol, and acetyl chloride were

432 purchased from Sigma-Aldrich and used without further

pur-433 ification. Gelatin was purchased from J. T. Baker, and sodium

434 azide was purchased from Fisher Scientific. Pluronic F-68 was

435 purchased from BASF. Type II Collagenase was purchased from

436 Worthington. CDCl3was purchased from CIL. Human breast

437 cancer MDA435 cells were purchased from ATCC. Nude mice

438 (NCR-NU-F) were purchased from Taconic, Inc.

439 Synthesis of Fatty Esters. Fatty esters were synthesized as

440 previously reported.25Briefly, eicosanoic acid was dissolved in

441 chloroform, to which an ethanolic solution of cupric acetate, as

442 well as ethanolic HCl, was added. This solution was stirred for 1

443 h, after which time the reaction was quenched with distilled

444 deionized water, and the fatty ester was extracted with

chloro-445 form. The chloroform was evaporated under nitrogen gas,

446 yielding a white solid. The solid was dissolved in CDCl3, and

447 the structure was confirmed by standard1H NMR and13C NMR

448 performed on a Varian Mercury 300 MHz instrument. The proton

449 and carbon NMR spectra of the fatty ester and the fatty acid are

450 given in the Supporting Information. No residual eicosanoic

451 acid can be seen in either NMR spectrum, indicating its

com-452 plete conversion to ester.

453 Synthesis of PbSe Quantum Dots. PbSe quantum dots were

454 synthesized according to previously published methods.15

455 Briefly, lead oxide was dissolved in octadecene and oleic acid

456 under a nitrogen atmosphere at 150 C. Once dissolved, the

457 solution was cooled, opened to ambient air, and a solution of

458 selenium in trioctylphosphine was added. This solution was

459 stirred for 4 h, after which time the quantum dots were isolated

460 through the addition of butanol and methanol. For preparation

461 of water-soluble QD for toxicity assessment, the exchange of

462 oleic acid for MTPEG on the PbSe QD surface was accomplished

463 as previously described.15The fluorescence spectra of the PbSe

464 QDs were recorded on an Ocean Optics spectrofluorimeter.

465 Formulation of QD-FENs. The fatty esters prepared in this work,

466 Myrj 52, and Myrj 56, were heated to 80 C until melted. The

467 molten mixture was stirred and PbSe QD suspended in hexanes

468 was added. The mixture was stirred for 20 min to allow for the

469 evaporation of hexanes and dispersion of the QDs in the molten

470 fatty ester. An aqueous solution of Pluronic F68 was then added

471 to the molten mixture, followed by distilled deionized water

472 preheated to 80 C. This emulsion was ultrasonicated using a

473 Hielscher UP-100S probe sonicator (100 W, 30 kHz) for 5 min.

474 Immediately following sonication, the entire emulsion was

475 dispersed in cold 0.9% saline. Particle size and zeta potential

476 were measured on a Nicomp 380 Zetasizer by dynamic light

477 scattering and electrophoretic light scattering, respectively.

Parti-478 cle morphology and QD loading were assessed by transmission

479

electron microscopy of unstained samples using a Hitachi

480

H-7000 microscope.

481

Preparation of Tissue Phantom.Tissue phantoms were prepared

482

based on the compositions reported previously.26Tris-buffered

483

saline was poured on top of a known amount of sodium azide

484

and gelatin to make a final concentration of 15 mM and 10w/v%

485

respectively. The mixture was heated to 50 C using a water

486

bath (Haake D8 Immersion heater) with constant stirring by a

487

Caframo BDC 1850 overhead stirrer at 650-800 rpm. After the

488

gelatin had dissolved, the mixture was cooled to 37 C with

489

constant stirring. Hemoglobin and intralipid were added to

490

reach concentrations of 170 μM and 1 v/v%, respectively.

491

A gel cassette intended for electrophoresis was used as a

492

mold to form a 1 mm thick slab of tissue phantom containing

493

QD-FEN. QD-FEN in saline was mixed with a warm phantom

494

mixture at 1 mg of QD-FE/mL or 5 mg of QD-FEN/mL

concen-495

trations. For the 5 mg QD-FEN/mL slab, extra gelatin was added

496

to make a final gelatin solution of 10 wt % in order to ensure

497

proper slab solidification. All glassware used was warmed to 37 C

498

to prevent premature solidification of the phantom mixture.

499

The warm QD-FEN mixture was then pipetted into the space

500

between the two glass plates almost to the brim. The entire set

501

up was then placed in refrigeration (4 C) until it solidified.

502

The construction of the tissue phantom is outlined in

503

Supporting Information, Figure S2. Before phantom

construc-504

tion, the mold was kept under refrigeration to ensure rapid

505

solidification of the phantom and prevent intralipid separation.

506

To construct the bottom layer of the phantom, the mold was

507

tilted such that the bottom was inclined to 45. Warm tissue

508

phantom mixture was poured to a desired depth and tapped to

509

remove any air bubbles. The bottom layer was refrigerated until

510

solid. The remainder of the tissue phantom mixture was kept

511

warm at 37 C with constant stirring. The QD-FEN slab was

512

removed from the electrophoresis gel cassette and trimmed to

513

fit the mold. The slab was then laid midline across the length of

514

the bottom tissue phantom layer, and any air bubbles were

515

smoothed out. Warm tissue phantom mixture was poured over

516

top to fill the mold, and the mold was refrigerated until

517

solidified. For depths approximately 3 cm or deeper, warm

518

tissue phantom was poured and solidified layer-by-layer to

519

prevent dissolution of the QD-FEN slab layer. Solidified tissue

520

phantom was removed from the mold by running a thin spatula

521

around the edge. Once complete, the tissue phantom was

522

stored at 4 C until needed. Excess tissue phantom mixture or

523

QD-FEN slab mixtures were also stored at 4 C, to be melted at

524

37 C for reuse.

525

Optical Characterization of Tissue Phantom. To ensure that the

526

composition of the tissue phantom produced a gel with optical

527

properties comparable to that of human tissue, the scattering

528

coefficient (μs) and absorption coefficient (μa) were quantified

529

using integrating spheres. The sample was mounted between 2

530

BK7, 1 mm thick optical windows (Esco Products), which were

531

separated by a 1 mm thick spacer. The spacer had a cutout at the

532

top so that the phantom mixture could be poured in before it

533

solidified. A halogen white light source (HL-2000, Ocean Optics)

534

was used, via focusing lenses, to illuminate the sample when it

ARTICLE

(9)

535 was mounted in a reflectance-transmittance (RT) integrating

536 sphere system. The 6 in. diameter integrating spheres

(Sphere-537 Optic) were mounted one at a time to eliminate cross talk. The R

538 sphere was mounted in front of the sample position and

refle-539 ctance, background, and reference signal spectra were

re-540 corded. The R sphere was then removed and the T sphere

541 was mounted behind the sample. The transmittance,

back-542 ground, and reference spectra were recorded. A Monte Carlo

543 simulation using 106photons and an asymmetry parameter =

544 0.9 generated a lookup table which matched the R and T values

545 with μaand μsvalues.

546 Tissue Phantom Image Acquisition and Data Analysis. Tissue

phan-547 toms were imaged using a Xenogen IVIS Spectrum instrument

548 in epifluorescence mode. The phantom was placed on the stage

549 and images were taken with automatic exposure and aperture

550 settings in field of view C. One image was taken from the top of the

551 gel phantom, and a second image was taken of the cross section of

552 the gel. Intensity scales were set to a monochromatic (red) type

553 with units of radiant efficiency and the range was set to full.

554 ImageJ27was used for image analysis, which comprised four

555 steps:

556 (1) Using the monochromatic scale, a calibration of pixel

557 intensity versus the intensity value in radiant efficiency

558 was performed. The dimensions of the intensity scale

559 were determined in pixels, allowing the dimensions of

560 one pixel to be determined in terms of the intensity

561 scale dimensions. ImageJ was then used to determine

562 the pixel-by-pixel intensity of the intensity scale bar,

563 allowing for a plot of pixel intensity versus radiant

564 efficiency to be generated.

565 (2) Using the top view image of the gel phantom, the pixel

566 intensity versus the distance in millimeters from the

567 origin (selected to be the topmost point of the QD-FEN

568 slab in the gel phantom) was determined. The

determi-569 nation of the dimensions of a pixel allowed for a plot of

570 pixel intensity versus distance in millimeters from the

571 origin to be generated.

572 (3) Using the cross-sectional image, the depth of the

QD-573 FEN gel slab was determined from the top of the gel

574 phantom, resulting in a plot of distance from the origin

575 versus QD-FEN slab depth.

576 (4) Combining the plot of pixel intensity versus distance

577 from the origin (Step 2) with the plot of distance from

578 the origin versus QD-FEN slab depth (Step 3), a plot of

579 pixel intensity in radiant efficiency versus QD-FEN slab

580 depth was generated, as shown in Figure 4D.

581 Determination of Cytotoxicity of QDs and QD-FENs. Hepatocytes

582 were isolated from rats by perfusion of the liver with

collage-583 nase as described previously.34Isolated hepatocytes (106cells/

584 mL) (10 mL) were suspended in Krebs-Henseleit buffer (pH 7.4)

585 containing 12.5 mM HEPES in continually rotating 50 mL

round-586 bottomed flasks, under an atmosphere of 95% O2and 5% CO2in

587 a water bath of 37 C for 30 min. Hepatocyte viability was

588 assessed microscopically by plasma membrane disruption as

589 determined by the trypan blue (0.1% w/v) exclusion test.35

590 Hepatocyte viability was determined at 30, 60, and 120 min,

591 and the cells were at least 80-90% viable before use.

592 Transfection of Tumor Cell.Human MDA435 breast

adenocar-593 cinoma cells were stably transfected to express enhanced green

594 fluorescent protein (EGFP). The EGFP plasmid (pEGFP-N1,

595 Clonetech) was linearized by digestion with the restriction

596 endonuclease Dra III (New England Biolabs), and was

trans-597 fected into the cells by incubation with CaPO4. After 24 h of

598 incubation, cells were grown in selection media containing

599 minimal essential medium supplemented with 10% fetal calf

600 serum and 600 μg/mL of the selection antibiotic G418. Cell

601 colonies that formed and that were fluorescent were lifted and

602 plated in selection medium in individual wells of a 24-well

603 microplate. The three best colony expansions, determined by

604 fluorescent intensity and number of fluorescent cells, were

605 selected for sorting by fluorescence activated cell sorting

606 (FACS). The brightest 20% of each of the three expanded

607 colonies were selected by FACS and were further expanded in

608 regular growth medium.

609

In VivoWhole Animal Imaging in Tumor-Bearing Mice. To establish

610

orthotopic xenograft breast tumors, 1 million human MDA435

611

cells expressing EGFP were injected into the inguinal mammary

612

fat pads of nude mice using the subiliac lymph node as an

613

injection landmark. This ensured greater mouse-to-mouse

re-614

producibility of the tumor model. Tumors were monitored for

615

growth with fluorescent imaging during the course of the

three-616

week growth period. Mice were injected with QD-FEN

formula-617

tion into the lateral tail veins and anaesthetized with isoflurane.

618

Images were acquired using a Xenogen IVIS Spectrum whole

619

animal imager with λex= 710 nm and λem= 840 nm. All animal

620

work was approved by the animal care committee at the

621

University Health Network, and all experiments were performed

622

in accordance with all guidelines and regulations put forth by

623

the Canadian Council on Animal Care.

624

Acknowledgment. The authors gratefully thank the

Cana-625

dian Breast Cancer Foundation-Ontario Region for funding the

626

work herein, the National Science and Engineering Council of

627

Canada, the University of Toronto and the Ben Cohen Fund for

628

providing scholarships to A. Shuhendler. The authors also

629

acknowledge A. Worthington for performing the

measure-630

ments of the optical properties of the tissue phantoms, J. T.

631

Henderson for providing the enhanced green fluorescent

pro-632

tein plasmid used to label the tumor cells, M. Monroy for

633

technical assistance with the mammary fat pad mouse model,

634

and STTARR for technical assistance with fluorescence imaging.

635

Supporting Information Available:Further experimental

de-636

tails regarding the NMR characterization of fatty acid ester

637

structure and the construction of the tissue phantom for

638

quantification of optical probe fluorescent depth penetration.

639

This material is available free of charge via the Internet at http://

640

pubs.acs.org.

641

REFERENCES AND NOTES

642

1. Holm, B. A.; Bergey, E. J.; De, T.; Rodman, D. J.; Kapoor, R.;

643

Levy, L.; Friend, C. S.; Prasad, P. N. Nanotechnology in

644

Biomedical Applications. Mol. Cryst. Liq. Cryst. Sci. Technol.,

645

Sect. A 2002, 374, 589.

646

2. Aswathy, R. G.; Yoshida, Y.; Maekawa, T.; Kumar, D. S.

Near-647

Infrared Quantum Dots for Deep Tissue Imaging. Anal.

648

Bioanal. Chem. 2010, 397, 1417.

649

3. Altinoglu, E. I.; Adair, J. H. Near Infrared Imaging With

650

Nanoparticles. Wiley Interdiscip. Rev. Nanomed.

Nanobio-651

technol. 2010, 2, 461.

652

4. Smith, A. M.; Mancini, M. C.; Nie, S. Bioimaging: Second

653

Window for in Vivo Imaging. Nat. Nanotechnol. 2009, 4,

654

710.

655

5. Ntziachristos, V.; Bremer, C.; Weissleder, R. Fluorescence

656

Imaging With Near-Infrared Light: New Technological

657

Advances That Enable in Vivo Molecular Imaging. Eur.

658

Radiol. 2003, 13, 195.

659

6. Ghoroghchian, P. P.; Therien, M. J.; Hammer, D. A. In Vivo

660

Fluorescence Imaging: A Personal Perspective. Wiley

Inter-661

discip. Rev. Nanomed. Nanobiotechnol. 2009, 1, 156.

662

7. Prasad, P. N. Introduction to Biophotonics; John Wiley and

663

Sons: Hoboken, NJ, 2003.

664

8. Santra, S.; Dutta, D. Nanoparticles for Optical Imaging of

665

Cancer. In Nanomaterials for Cancer Diagnosis; Challa, S. S. R. K.,

666

Ed.; Wiley-VCH Verlag GmbH: Weinheim, Germany, 2007; 44.

667

9. Ballou, B.; Ernst, L. A.; Andreko, S.; Harper, T.; Fitzpatrick,

668

J. A.; Waggoner, A. S.; Bruchez, M. P. Sentinel Lymph Node

669

Imaging Using Quantum Dots In Mouse Tumor Models.

670

Bioconjugate Chem. 2007, 18, 389.

671

10. Kim, S.; Lim, Y. T.; Soltesz, E. G.; De Grand, A. M.; Lee, J.;

672

Nakayama, A.; Parker, J. A.; Mihaljevic, T.; Laurence, R. G.;

673

Dor, D. M.; et al. Near-Infrared Fluorescent Type II

Quan-674

tum Dots for Sentinel Lymph Node Mapping. Nat.

Bio-675

technol. 2004, 22, 93.

676

11. Zimmer, J. P.; Kim, S. W.; Ohnishi, S.; Tanaka, E.; Frangioni,

677

J. V.; Bawendi, M. G. Size Series of Small Indium Arsenide

678

-Zinc Selenide Core-Shell Nanocrystals and Their

Appli-679

cation to In Vivo Imaging. J. Am. Chem. Soc. 2006, 128,

680

2526.

ARTICLE

(10)

681 12. Cai, W.; Shin, D. W.; Chen, K.; Gheysens, O.; Cao, Q.; Wang,

682 S. X.; Gambhir, S. S.; Chen, X. Peptide-Labeled

Near-Infra-683 red Quantum Dots for Imaging Tumor Vasculature in

684 Living Subjects. Nano Lett. 2006, 6, 669.

685 13. Diagaradjane, P.; Orenstein-Cardona, J. M.;

Colon-Casas-686 novas, N. E.; Deorukhkar, A.; Shentu, S.; Kuno, N.; Schwartz,

687 D. L.; Gelovani, J. G.; Krishnan, S. Imaging Epidermal

688 Growth Factor Receptor Expression In Vivo:

Pharmacoki-689 netic and Biodistribution Characterization of a

Bioconju-690 gated Quantum Dot Nanoprobe. Clin. Cancer Res. 2008,

691 14, 731.

692 14. Du, H.; Chen, C.; Krishnan, R.; Krauss, T. D.; Harbold, J. M.;

693 Wise, F. W.; Thomas, M. G.; Silcox, J. Optical Properties of

694 Colloidal PbSe Nanocrystals. Nano Lett. 2002, 2, 1321.

695 15. Evans, C. M.; Guo, L.; Peterson, J. J.; Maccagnano-Zacher, S.;

696 Krauss, T. D. Ultrabright PbSe Magic-Sized Clusters. Nano

697 Lett. 2008, 8, 2896.

698 16. Azzazy, H. M.; Mansour, M. M.; Kazmierczak, S. C. From

699 Diagnostics to Therapy: Prospects of Quantum Dots. Clin.

700 Biochem. 2007, 40, 917.

701 17. Cho, S. J.; Maysinger, D.; Jain, M.; Roder, B.; Hackbarth, S.;

702 Winnik, F. M. Long-Term Exposure to CdTe Quantum Dots

703 Causes Functional Impairments in Live Cells. Langmuir

704 2007,23, 1974.

705 18. Hardman, R. A Toxicologic Review of Quantum Dots:

706 Toxicity Depends on Physicochemical and Environmental

707 Factors. Environ. Health Perspect. 2006, 114, 165.

708 19. Bentolila, L. A.; Ebenstein, Y.; Weiss, S. Quantum Dots for in

709 VivoSmall-Animal Imaging. J. Nucl. Med. 2009, 50, 493.

710 20. Yang, R. S.; Chang, L. W.; Wu, J. P.; Tsai, M. H.; Wang, H. J.;

711 Kuo, Y. C.; Yeh, T. K.; Yang, C. S.; Lin, P. Persistent Tissue

712 Kinetics and Redistribution of Nanoparticles, Quantum

713 Dot 705, in Mice: ICP-MS Quantitative Assessment.

En-714 viron. Health Perspect. 2007, 115, 1339.

715 21. Liu, W.; He, Z.; Liang, J.; Zhu, Y.; Xu, H.; Yang, X. Preparation

716 and Characterization of Novel Fluorescent

Nanocompo-717 site Particles: CdSe/ZnS Core-Shell Quantum Dots

718 Loaded Solid Lipid Nanoparticles. J. Biomed. Mater. Res.

719 A 2008, 84, 1018.

720 22. Ouyang J.; Schuurmans C.; Zhang Y.; Nagelkerke R.; Wu X.;

721 Kingston D.; Wang Z. Y.; Wilkinson D.; Li C.; Leek M. D.; et al.

722 Low-Temperature Approach to High-Yield and

Reprodu-723 cible Syntheses of High-Quality Small-Sized PbSe

Colloi-724 dal Nanocrystals for Photovoltaic Applications. ACS Appl.

725 Mater. Interfaces2010, DOI: am-2010-01129m.R1,

726 23. Gabizon, A. A. Stealth Liposomes and Tumor Targeting:

727 One Step Further in the Quest for the Magic Bullet. Clin.

728 Cancer Res. 2001, 7, 223.

729 24. Benson, R. C.; Kues, H. A. Absorption and Fluorescence

730 Propertes of Cyanine Dyes. J. Chem. Eng. Data 1977, 22, 379.

731 25. Hoshi, M.; Williams, M.; Kishimoto, Y. Esterification of Fatty

732 Acids at Room Temperature by Chloroform-Methanolic

733 HCl-Cupric Acetate. J. Lipid Res. 1973, 14, 599.

734 26. De Grand, A. M.; Lomnes, S. J.; Lee, D. S.; Pietrzykowski, M.;

735 Ohnishi, S.; Morgan, T. G.; Gogbashian, A.; Laurence, R. G.;

736 Frangioni, J. V. Tissue-like Phantoms for Near-Infrared

737 Fluorescence Imaging System Assessment and the

Train-738 ing of Surgeons. J. Biomed. Opt. 2006, 11, 014007.

739 27. Abramoff, M. D.; Magelheas, P. L.; Ram, S. J. Image

Proces-740 sing with ImageJ. Biophoton. Int. 2004, 11, 36.

741 28. Patocka, J.; Cerny, K. Inorganic Lead Toxicology. Acta Med.

742 (Hradec Kralove) 2003, 46, 65.

743 29. MacFarquhar, J. K.; Broussard, D. L.; Melstrom, P.;

744 Hutchinson, R.; Wolkin, A.; Martin, C.; Burk, R. F.; Dunn,

745 J. R.; Green, A. L.; Hammond, R.; et al. Acute Selenium

746 Toxicity Associated with a Dietary Supplement. Arch.

747 Intern. Med. 2010, 170, 256.

748 30. O'Brien, P. J.; Chan, K.; Silber, P. M. Human and Animal

749 Hepatocytes in Vitro with Extrapolation in Vivo. Chem. Biol.

750 Interact. 2004, 150, 97.

751 31. Shuhendler, A. J.; Cheung, R. Y.; Manias, J.; Connor, A.;

752 Rauth, A. M.; Wu, X. Y. A Novel Doxorubicin-Mitomycin C

753 Co-encapsulated Nanoparticle Formulation Exhibits

Anti-754 cancer Synergy in Multidrug Resistant Human Breast

755 Cancer Cells. Breast Cancer Res. Treat. 2010, 119, 255.

756

32. Wong, H. L.; Rauth, A. M.; Bendayan, R.; Wu, X. Y. In Vivo

757

Evaluation of a New Polymer-Lipid Hybrid Nanoparticle

758

(PLN) Formulation of Doxorubicin in a Murine Solid Tumor

759

Model. Eur. J. Pharm. Biopharm. 2007, 65, 300.

760

33. Mansur, H. S. Quantum Dots and Nanocomposites. Wiley

761

Interdiscip. Rev. Nanomed. Nanobiotechnol. 2010, 2, 113.

762

34. Moldeus, P.; Hogberg, J.; Orrenius, S. Isolation and Use of

763

Liver Cells. Methods Enzymol. 1978, 52, 60.

764

35. Moridani, M. Y.; Pourahmad, J.; Bui, H.; Siraki, A.; O'Brien,

765

P. J. Dietary Flavonoid Iron Complexes as Cytoprotective

766

Superoxide Radical Scavengers. Free Radical Biol. Med.

767

2003,34, 243.

ARTICLE

Références

Documents relatifs

In this general setting, our main results extend several properties of the replicator dynamics such as the elimination of dominated strategies, the asymptotic stability of strict

3 University of Ottawa Brain and Mind Research Institute, Ottawa, Ontario, Canada 4 McGovern Institute for Brain Research, Cambridge, Massachusetts, USA 5 Department of Brain

(years); fully adjusted models were adjusted for age (years), sex, ethnicity, mean arterial blood pressure and BMI of the child at the moment of the follow-up visit, the season in

Cela pose nécessairement un certain nombre de questions, comme celle des échanges, des réseaux, des har- monisations, la question des partenariats, de la mobilité, mais aussi

Recently a few patients have been described with mild MMAuria associated with a mutation of the MMCoA epimerase gene (MCEE), but these patients had no consistent clinical phenotype,

We used the sub field OI and the temporal modulation ratio (F1/F0) derived from the drifting grating stimulus to identify simple and complex cells in tree shrew V1.. The laminar

Developmental transitions in body color in chacma baboon infants: Implications to estimate age and developmental pace... 2 Department of Anthropology, Stony Brook University,

Nessaram di taggara n tezrewt-a inexdem, ad yili nessaweḍ ɣer kra n yigemmaḍ, ideg neɛreḍ ad nessekfel, ismawen iqburen icudden ɣer umawal n uxxam aqdim, seg