Sei sulla pagina 1di 14

Published on Web 10/04/2010

Engineered Biocompatible Nanoparticles for in Vivo Imaging


Applications
Shu Chen,†,‡ Lijun Wang,§ Suzanne L. Duce,| Stuart Brown,§ Stephen Lee,†
Andreas Melzer,§ Sir Alfred Cuschieri,*,§ and Pascal André*,†
School of Physics and Astronomy (SUPA), UniVersity of St Andrews,
St Andrews KY16 9SS, U.K., School of Chemistry (EaSt CHEM), UniVersity of St Andrews,
St Andrews KY16 9ST, U.K., Institute for Medical Science and Technology, UniVersity of
Dundee, Dundee DD2 1FD, U.K., and DiVision of Biological Chemistry and Drug DiscoVery,
College of Life Sciences, UniVersity of Dundee, Dundee DD1 5EH, U.K.
Received July 23, 2010; E-mail: Pascal.Andre@st-andrews.ac.uk; a.cuschieri@dundee.ac.uk

Abstract: Iron-platinum alloy nanoparticles (FePt NPs) are extremely promising candidates for the next
generation of contrast agents for magnetic resonance (MR) diagnostic imaging and MR-guided interventions,
including hyperthermic ablation of solid cancers. FePt has high Curie temperature, saturation magnetic
moment, magneto-crystalline anisotropy, and chemical stability. We describe the synthesis and characteriza-
tion of a family of biocompatible FePt NPs suitable for biomedical applications, showing and discussing
that FePt NPs can exhibit low cytotoxicity. The importance of engineering the interface of strongly magnetic
NPs using a coating allowing free aqueous permeation is demonstrated to be an essential parameter in
the design of new generations of diagnostic and therapeutic MRI contrast agents. We report effective cell
internalization of FePt NPs and demonstrate that they can be used for cellular imaging and in vivo MRI
applications. This opens the way for several future applications of FePt NPs, including regenerative medicine
and stem cell therapy in addition to enhanced MR diagnostic imaging.

1. Introduction development of exogenous MRI contrast agents.12 Common T2-


agents include superparamagnetic nanoparticles (Fe3O4, γ-Fe2O3,
Nanoparticles (NPs) have attracted considerable interest as MnFe2O4, FeCo), the clinical MRI applications of which range
diagnostic and therapeutic tools for biomedicine,1-3 with from imaging lymph-node to liver and spleen, as well as bone
magnetic NPs offering unique opportunities for magnetic marrow and gastrointestinal tract.4,13,14 Despite its widespread
separation, targeted drug delivery, and hyperthermic ablation usage, superparamagnetic iron oxide (SPIO) has several disad-
of cancer and as contrast agents in magnetic resonance imaging vantages, including rapid clearance by phagocytic cells and
(MRI).4-11 MRI is a powerful, noninvasive medical imaging hence limited trans-endothelia passage and tissue penetration.4,15
technique where the signal originates predominately from water There is therefore a need for the design of biocompatible MRI
and lipids. Medical diagnosis requires enhanced contrast contrast agents with prolonged intravascular retention, improved
between normal and pathological tissues, resulting in the tissue delineation, high chemical stability, and improved
selectivity when targeting a tissue of interest.

SUPA, University of St Andrews. Small NPs avoid fast clearance by the reticulo-endothelial

EaSt CHEM, University of St Andrews. system and therefore should enable long blood circulation half-
§
Institute for Medical Science and Technology, University of Dundee.
|
Division of Biological Chemistry and Drug Discovery, University of life, thereby improving their ability to reach specific targets.4,16
Dundee. Silica is often considered as the most attractive coating for
(1) Shubayev, V. I.; Pisanic, T. R.; Jin, S. H. AdV. Drug DeliV. ReV. 2009, engineered NPs, forming an inert and biocompatible outer shell.
61, 467–477.
(2) Daniel, M. C.; Astruc, D. Chem. ReV. 2004, 104, 293–346.
Not only does it create a protective shell against chemical
(3) Medintz, I. L.; Uyeda, H. T.; Goldman, E. R.; Mattoussi, H. Nat. Mater. degradation, but it also allows surface functionalization with
2005, 4, 435–446. functional alkoxysilanes.7 An outer coating with functional
(4) Na, H. B.; Song, I. C.; Hyeon, T. AdV. Mater. 2009, 21, 2133–2148.
(5) Pankhurst, Q. A.; Connolly, J.; Jones, S. K.; Dobson, J. J. Phys. D:
organic molecules has many advantages, including steric and/
Appl. Phys. 2003, 36, R167–R181. or electrostatic repulsion between the NPs, which can increase
(6) Gao, J. H.; Gu, H. W.; Xu, B. Acc. Chem. Res. 2009, 42, 1097–1107.
(7) Sun, C.; Lee, J. S. H.; Zhang, M. Q. AdV. Drug DeliV. ReV. 2008, 60,
1252–1265. (12) Brown, M. A.; Semelka, R. C. MRI: Basic Principles and Applications,
(8) Veiseh, O.; Gunn, J. W.; Zhang, M. Q. AdV. Drug DeliV. ReV. 2010, 3rd ed.; Wiley: Hoboken, NJ, 2005.
62, 284–304. (13) Yan, G.; Robinson, L.; Hogg, P. Radiography 2007, 13, e5-e19.
(9) Jeong, U.; Teng, X. W.; Wang, Y.; Yang, H.; Xia, Y. N. AdV. Mater. (14) Kim, J.; Piao, Y.; Hyeon, T. Chem. Soc. ReV. 2009, 38, 372–390.
2007, 19, 33–60. (15) Bulte, J. W. M.; Kraitchman, D. L. NMR Biomed. 2004, 17, 484–
(10) Lu, A. H.; Salabas, E. L.; Schuth, F. Angew. Chem., Int. Ed. 2007, 499.
46, 1222–1244. (16) Modo, M. M. J.; Bulte, J. W. M. Molecular and Cellular MR Imaging,
(11) Gao, J. H.; Xu, B. Nano Today 2009, 4, 37–51. 1st ed.; CRC Press: Boca Raton, FL, 2007.

15022 9 J. AM. CHEM. SOC. 2010, 132, 15022–15029 10.1021/ja106543j  2010 American Chemical Society
Engineered Biocompatible Nanoparticles ARTICLES

water solubility and stability in biological environment.17 In this article, we describe the fabrication of FePt NPs
Positively charged NPs demonstrate increased cell internalization specifically designed to enhance their magnetic properties for
efficiency and biomedical molecules binding capability for biomedical applications. We demonstrate that, in contrast with
multimodal imaging applications and drug delivery.4,18,19 previous reports, FePt NPs can be made nontoxic, and we
Characterized by high Curie temperature and magneto- provide the first data on their cellular uptake mechanisms. We
crystalline anisotropy, iron-platinum metallic alloy NPs have report a 6-fold increase in the FePt-based T2 contrast properties
emerged as promising candidates for a new generation of compared to those of clinical iron oxide NPs. The relationship
magnetic nanomaterials.20 Over the past decade, several chemi- between the MRI contrast properties and the NPs’ architecture
cal pathways have been developed to synthesize FePt NPs with is explored and rationalized as the basis for the design of NPs
controlled stoichiometry, size, and shape.5 Such synthetic routes as enhanced MRI contrast agents. Finally, we report the first
include co-reduction of Fe and Pt precursors through both low- observations of cellular and in ViVo MR imaging with FePt NPs.
temperature emulsion protocols and the high-temperature polyol Our results opens the way for several applications of FePt NPs,
pathway,21-25 along with the thermal decomposition route.26-31 such as regenerative medicine and stem cell therapy, in addition
The latter approach often relies on Pt(acac)2, Fe(CO)5, or to enhanced MR diagnostic imaging, thus providing a platform
Collman’s regeant precursors mixed with alkyl amines and acids to develop novel diagnostic and therapeutic agents.
solubilized in high-boiling-point solvents heated at temperatures
of ∼300 °C and above to control the growth, composition, and 2. Results
morphology of the NPs. Figure 1 displays the schematic structure of a clinical iron
Studies have demonstrated the biomedical potential of FePt oxide NP coated with dextran (Feridex, Figure 1A1) and two
NPs,6 such as in magnetic separation32,33 and hyperthermic FePt NPs: the face-centered-cubic (fcc) FePt NP surface
ablation34,35 and as T2 MRI contrast agents.36-38 However, functionalized with cysteamine (fcc-FePt-A, Figure 1B1) and
toxicity remains a major concern, preventing their use as a the fcc FePt NP coated with silica and surface-functionalized
bioplatform for diagnosis and therapy.37,39,40 with (3-aminopropyl)triethoxysilane (fcc-FePt-silica-A, Figure
1C1). The preparation of the family of FePt NPs is described
(17) Tanaka, Y.; Maenosono, S. J. Magn. Magn. Mater. 2008, 320, L121– in the Supporting Information.27,30,17,41
L124. In agreement with published literature,42 transmission electron
(18) Josephson, L.; Tung, C. H.; Moore, A.; Weissleder, R. Bioconjugate
Chem. 1999, 10, 186–191. microscopy (TEM) revealed that the commercial iron oxide NPs
(19) Kang, H. W.; Josephson, L.; Petrovsky, A.; Weissleder, R.; Bogdanov, samples (Figure 1A2) are composed of aggregated crystals
A. Bioconjugate Chem. 2002, 13, 122–127. approximately 5 nm in diameter, embedded in an organic
(20) Sun, S. H. AdV. Mater. 2006, 18, 393–403. coating. These aggregates form large particles of a size
(21) Nakaya, M.; Kanehara, M.; Teranishi, T. Langmuir 2006, 22, 3485–
3487. distribution ranging from 20 to >50 nm. In contrast, the fcc-
(22) Jeyadevan, B.; Hobo, A.; Urakawa, K.; Chinnasamy, C. N.; Shinoda, FePt-A NPs are well dispersed, with diameters of ∼5 nm (Figure
K.; Tohji, K. J. Appl. Phys. 2003, 93, 7574–7576. 1B2). The fcc-FePt-silica-A NPs are coated with a homogeneous
(23) Takahashi, M.; Ogawa, T.; Hasegawa, D.; Jeyadevan, B. J. Appl. Phys.
2005, 97, 10J307. silica shell of ∼17 nm in thickness, leading to a total diameter
(24) Yan, Q. Y.; Purkayastha, A.; Kim, T.; Kroger, R.; Bose, A.; Ramanath, of ∼40 nm (Figure 1C2).
G. AdV. Mater. 2006, 18, 2569–2573. The powder X-ray diffraction (XRD) pattern of the Feridex
(25) Saita, S.; Maenosono, S. Chem. Mater. 2005, 17, 6624–6634.
(26) Sun, S. H.; Murray, C. B.; Weller, D.; Folks, L.; Moser, A. Science iron oxide confirms the presence of magnetite (Fe3O4), the broad
2000, 287, 1989–1992. peaks at 38°, 45°, and 55° corresponding to the (220), (311),
(27) Chen, M.; Liu, J. P.; Sun, S. J. Am. Chem. Soc. 2004, 126, 8394– and (400) peaks, respectively (Figure 1A3). Figure 1B3,C3
8395.
(28) Nandwana, V.; Elkins, K. E.; Poudyal, N.; Chaubey, G. S.; Yano, K.; shows peaks around 51° and 60°, characteristic of fcc-FePt (111)
Liu, J. P. J. Phys. Chem. C 2007, 111, 4185–4189. and (200) peaks, respectively. The (111) peak position suggests
(29) Howard, L. E. M.; Nguyen, H. L.; Giblin, S. R.; Tanner, B. K.; Terry, that the NPs’ composition includes between 40 and 45% of Fe
I.; Hughes, A. K.; Evans, J. S. O. J. Am. Chem. Soc. 2005, 127, 10140– (Table 1). For the silica-coated NPs, the SiO2 XRD characteristic
10141.
(30) Nguyen, H. L.; Howard, L. E. M.; Stinton, G. W.; Giblin, S. R.; Tanner, peak is observed at ∼28° (Figure 1C3). The crystal size is ∼3.6
B. K.; Terry, I.; Hughes, A. K.; Ross, I. M.; Serres, A.; Evans, J. S. O. nm for both fcc-FePt-A and fcc-FePt-silica-A NPs (Table 1).
Chem. Mater. 2006, 18, 6414–6424. Zero-field-cooling and field-cooling (ZFC-FC) of Feridex
(31) Delalande, M.; Marcoux, P. R.; Reiss, P.; Samson, Y. J. Mater. Chem.
2007, 17, 1579–1588. samples highlight a 45 K blocking temperature (Tb, Figure 1A4).
(32) Gu, H. W.; Ho, P. L.; Tsang, K. W. T.; Yu, C. W.; Xu, B. Chem. At 2 K, i.e., lower than Tb, Feridex hysteresis loop measurements
Commun. 2003, 1966–1967. display ferrimagnetic behavior with a small coercivity value of
(33) Gu, H. W.; Ho, P. L.; Tsang, K. W. T.; Wang, L.; Xu, B. J. Am.
Chem. Soc. 2003, 125, 15702–15703. 300 Oe and a saturation moment (Ms) of 101 emu/g of Fe. At
(34) Maenosono, S.; Saita, S. IEEE Trans. Magn. 2006, 42, 1638–1642. 300 K, Feridex is superparamagnetic with no coercivity, as
(35) Seehra, M. S.; Singh, V.; Dutta, P.; Neeleshwar, S.; Chen, Y. Y.; Chen, illustrated in Figure 1A5. The Ms value is 82 emu/g of Fe, in
C. L.; Chou, S. W.; Chen, C. C. J. Phys. D: Appl. Phys. 2010, 43,
145001–145007.
agreement with the literature.16,42 In comparison, the fcc-FePt-A
(36) Maenosono, S.; Suzuki, T.; Saita, S. J. Magn. Magn. Mater. 2008, NPs have a higher Tb of ∼90 K (Figure 1B4). More importantly,
320, L79–L83. when compared to Feridex, the Ms of fcc-FePt-A NPs is ∼2
(37) Gao, J. H.; Liang, G. L.; Cheung, J. S.; Pan, Y.; Kuang, Y.; Zhao, F.; times larger at 2 K and 35% stronger at 300 K (Figure 1B5,
Zhang, B.; Zhang, X. X.; Wu, E. X.; Xu, B. J. Am. Chem. Soc. 2008,
130, 11828–11833. Table 1). After silica coating, the blocking temperature is
(38) Chou, S. W.; Shau, Y. H.; Wu, P. C.; Yang, Y. S.; Shieh, D. B.; reduced to 50 K, and the ZFC slope below Tb is steeper (Figure
Chen, C. C. J. Am. Chem. Soc. DOI: 10.1021/ja1035013, Publication
Date (Web) June 24, 2010.
(39) Gao, J.; Liang, G.; Zhang, B.; Kuang, Y.; Zhang, X.; Xu, B. J. Am. (41) Koole, R.; van Schooneveld, M. M.; Hilhorst, J.; Donega, C. D.; Hart,
Chem. Soc. 2007, 129, 1428–1433. D. C.; van Blaaderen, A.; Vanmaekelbergh, D.; Meijerink, A. Chem.
(40) Xu, C. J.; Yuan, Z. L.; Kohler, N.; Kim, J. M.; Chung, M. A.; Sun, Mater. 2008, 20, 2503–2512.
S. H. J. Am. Chem. Soc. 2009, 131, 15346–15351. (42) Jung, C. W.; Jacobs, P. Magn. Reson. Imaging 1995, 13, 661–674.
J. AM. CHEM. SOC. 9 VOL. 132, NO. 42, 2010 15023
ARTICLES Chen et al.

Figure 1. Schematic representation (1), TEM images (2), XRD patterns (3), ZFC-FC (4), and hysteresis (5) curves of Feridex (A1-A5), fcc-FePt-A NPs
(B1-B5), and fcc-FePt-silica-A NPs (C1-C5).

Table 1. XRD, SQUID, and MRI Dataa

Ms (emu/g Fe)
sample DXRD (nm) FexPt1-x (%) Tb (K) at 2 K at 300 K Hc at 2 K (kOe) r1b (s-1 mM-1) r2b (s-1 mM-1)
fcc-FePt-A 3.6 ( 0.1 42.7 ( 0.7 90 201 112 0.7 2.5 ( 1.0 887 ( 32
fcc-FePt-silica-A 3.6 ( 0.1 43.8 ( 1.4 50 191 83 1.3 0.3 ( 0.1 210 ( 3
Feridex I.V.c (Endorem) 5.3 ( 0.1 - 45 101 82 0.3 0.9 ( 0.1 148 ( 2
a
DXRD is the crystalline grain size of the NPs; in FexPt1-x, x is the composition based on the FePt lattice constant (SI-Figure 1);43 Tb is the blocking
temperature; Ms is the saturation moment; Hc is the magnetic coercivity of the NPs; and r1 and r2 are the 1H relaxivity values for 1% agarose gel
containing Feridex, fcc-FePt-A, and fcc-FePt-silica-A NPs. b A range of other commercial MRI contrast agent relaxivity values are presented in the
Supporting Information (SI-Table 2). c Trademark of AMAG Pharmaceuticals, Lexington, MA.

15024 J. AM. CHEM. SOC. 9 VOL. 132, NO. 42, 2010


Engineered Biocompatible Nanoparticles ARTICLES

Figure 2. Viability of A375M cells incubated with fcc-FePt-A NPs (A). Fe released from fcc-FePt-A NPs (orange [) in pH 4.8 PBS (B1). Fe released from
fcc-FePt-A NPs (orange [) and from Feridex (4)49 in solutions at pH ∼4.8 of RPMI-1640 cell media containing 20 mM sodium citrate. The incubation
concentrations for FePt and Feridex49 were 120 and 25 µg/mL, respectively (B2). Photomicrographs (magnification ×40) of Prussian blue-stained A375M
cells cultured on coverslips in cell media (C1), media containing 30 µg/mL of fcc-FePt-A NPs (C2), and media containing 30 µg/mL of Feridex (C3) after
overnight incubation. TEM images of A375M cells after 16 h incubation in media containing 30 µg/mL of fcc-FePt-A NPs (D1-D3).

1C4). At 2 K and 5 T, the magnetization of fcc-FePt-silica-A results were obtained with fcc-FePt-A NPs and both MCF7 and
NPs is ∼5% lower than that of fcc-FePt-A and 2 times larger U2OS tumor cell lines (SI-Figure 5A,B).43
than that of Feridex. At 300 K and 5 T, the magnetization of To confirm these results, the chemical stability of fcc-FePt-A
fcc-FePt-silica-A NPs is ∼36% lower than that of fcc-FePt-A NPs in an acidic environment was investigated by incubation
NPs and similar to that of Feridex (Figure 1C5, Table 1). of fcc-FePt-A in pH 4.8 buffer solutions. The concentration of
FePt NPs incubated in buffer solution is ∼120 µg/mL, to match
Cytotoxicity of the NPs was evaluated in an A375M cell line
the highest NPs concentration used in the cytotoxicity study
using fcc-FePt-A NPs and MTS assay with various concentra- (Figure 2A). In a pH 4.8 phosphate buffered saline (PBS),
tions and incubation times.43 Both dose-response and time cysteamine-coated FePt NPs show excellent chemical stability,
course studies were performed (Figure 2A). As expected,44,45 with only ∼0.6% Fe, i.e. <0.1 µg/mL, of iron released after 7
and in contrast with bare iron oxide,46,47 Feridex’s dextran days of incubation (Figure 2B1). As suggested by the recent
organic coating does not lead to any detectable cytotoxicity. literature,48,49 an in Vitro lysosomal model, i.e. RPMI-1640 cell
At 120 µg/mL, fcc-FePt-A NPs exhibited cytotoxicity that media containing 20 mM sodium citrate (pH 4.8), was also used
increases with incubation time. At a concentration of 60 µg/ to match the metabolic conditions. In these more extreme
mL, fcc-FePt-A NPs incubated with cells for 7 days showed a conditions (Figure 2B2), more Fe was releaseds0.3 µg/mL after
∼15% loss of cell viability. Significantly, there is no loss of 6 h and up to ∼3.3 µg/mL after 7 dayssbut importantly it
cell viability at concentrations of 30 µg/mL and below, even remained at a level comparable to the release observed with
after prolonged incubation periods of up to 7 days. Similar Feridex.
The high chemical stability of fcc-FePt-A was investigated
further. Prussian blue staining experiments were carried out and
(43) See Supporting Information for details. showed that FePt NPs have greater stability compared to Feridex
(44) Wang, L. J.; Wang, Z. G.; Frank, T. G.; Brown, S. I.; Chudek, S. A.;
Cuschieri, A. Nanomedicine-UK 2009, 4, 305–315. (Figure 2C1-C3).43 Indeed, photomicrographs obtained after
(45) Dobrovolskaia, M. A.; McNeil, S. E. Nat. Nanotechnol. 2007, 2, 469– overnight incubation show no stain for both blank control sample
478. (Figure 2C1) and cells labeled with fcc-FePt-A NPs (Figure
(46) Lewinski, N.; Colvin, V.; Drezek, R. Small 2008, 4, 26–49.
(47) Gupta, A. K.; Gupta, M. Biomaterials 2005, 26, 1565–1573.
(48) Skotland, T.; Sontum, P. C.; Oulie, I. J. Pharmaceut. Biomed. 2002, (49) Arbab, A. S.; Wilson, L. B.; Ashari, P.; Jordan, E. K.; Lewis, B. K.;
28, 323–329. Frank, J. A. NMR Biomed. 2005, 18, 383–389.
J. AM. CHEM. SOC. 9 VOL. 132, NO. 42, 2010 15025
ARTICLES Chen et al.

Figure 3. T2-1 (s-1) vs [Fe] (mM) of the water in 1% w/v agarose gel containing (blue b) fcc-FePt-A, (green 2) fcc-FePt-silica-A, and (orange [) Feridex
(A1). 1H longitudinal relaxation time T2-weighted MRI images of 1% agarose gel/water solution containing fcc-FePt-A NPs with [Fe] ) 0.00 mM (a), 0.02
mM (CFePt ) 6 µg/mL) (b), and 0.04 mM (CFePt ) 12 µg/mL) (c) (A2). T2-weighted cellular imaging by MRI (3D spin echo, TE ) 30 ms, TR ) 1 s) of
A375M cells loaded with fcc-FePt-A NPs with a cell density of 10 × 103 cells/mL (a), 100 × 103 cells/mL (b), and 1000 × 103 cells/mL (c) (B). Embryo
injected in oVo with cell culture media (1 µL) containing no NPs (C) and 20 µg/mL fcc-FePt-A (D). 3D surface reconstruction of embryo eyes (blue) and
blood vessels (red) showing the position in yellow of the transverse and coronal images (C1,D1); dorsal view of transverse image of the embryo’s head
aligned through the eyes (C2,D2); and anterior view of coronal image of the embryo’s head aligned through the eyes (C3,D3). 2D slices from 128 × 128
× 128 3D Rare-8 MRI data set of day 4 quail embryo egg (TR/TE ) 250/25 ms), field of view of 30 mm and pixel dimensions of 0.234 mm/pixel. Labels:
y, yolk; a, albumen; b, brain; in-e, injected eye; un-e, un-injected eye; h, heart. Scale bar indicates 1 mm. All the MRI measurements were completed at 7.1
T.

2C2). In contrast, Feridex-labeled cells show significantly commonly observed in other NPs systems, the FePt NPs appear
staining (Figure 2C3) characteristic of the release of Fe in the typically “entrapped” inside these subcellular vesicles (Figure
cells. These results confirm that the cysteamine coating provides 2D3).
fcc-FePt-A NPs with high chemical stability. The effectiveness of fcc-FePt-A and fcc-FePt-silica-A as MRI
The cysteamine coating is positively charged, resulting in contrast agents was investigated by measuring the dependence
higher cellular uptake by human non-phagocytic A375M tumor of the longitudinal relaxation rate (T1-1) and transverse relax-
cells (Figure 2D1-D3) compared to Feridex. FePt NPs appear ation rate (T2-1) of 1% agarose gels.43 Relaxivity (ri) quantifies
to be internalized by an endocytotic pathway. Indeed, TEM the enhancement of either the longitudinal or the transverse
studies suggest a macropinocytosis cellular uptake mechanism relaxation rates of the protons in water. As expected at high
by the large protrusions of the plasma membrane around NP magnetic field, the FePt NPs and Feridex have a weak influence
clusters (Figure 2D2).50,51 The intracellular localization route on the water 1H longitudinal relaxation rate (Table 1 and
is endosome, early lysosome, and late lysosome, which are basic SI-Figure 7A).16,43 In contrast, FePt NPs have a significant effect
cell biology phenomena. Both endosomes and lysosomes are upon the aqueous transverse relaxation rate (Figure 3A1). The
confined subcellular organelles, and, as both expected and relaxivity r2 of the fcc-FePt-silica-A NPs is 40% larger than
that of Feridex, which is attributed to the stronger magnetic
moment of the fcc-FePt-silica-A NPs (SI-Figure 7B) at the MRI
(50) Conner, S. D.; Schmid, S. L. Nature 2003, 422, 37–44.
(51) Kirkham, M.; Parton, R. G. Biochim. Biophys. Acta 2005, 1745, 273– magnetic field (7 T).43 Importantly, the relaxivity r2 of the fcc-
286. FePt-A NPs is more than 6 times larger than that of Feridex.
15026 J. AM. CHEM. SOC. 9 VOL. 132, NO. 42, 2010
Engineered Biocompatible Nanoparticles ARTICLES

Table 2. Summary of FePt Nanoparticles Cytotoxicity Studies Reported in the Literaturea

cytotoxicity
b
material Fe precursor coating cells [NPs] (µg/mL) [Fe] (µg/mL) tinc (h) cell damage (%) ref
FePt-CoS2 Fe(CO)5 CoS2 shell HeLa 1.5 ∼0.007 24 50 39
FePt-Cys Fe(CO)5 cysteine HeLa 5 ∼0.7 24 ∼10 39
15.5 ∼2.5 24 50
FePt-Fe2O3 Fe(CO)5 Fe2O3 shell, L-DOPA coating HeLa ∼8.3 ∼0.045c 24 50 37
∼3.8d
FePt-Fe3O4 Fe(CO)5 Fe3O4 shell HeLa 40 e 24 ∼10 37
10 e 72 ∼10
20 e 72 ∼30
FePt-COOH Fe(CO)5 bilayer of phospholipid HeLa ∼10.7 1.72 24 ∼40 52
(DSPE-PEG(2000) carboxylic acid lipid) A431 ∼10.7 1.72 24 ∼40
and oleic acid/oleylamine SK-BR3 ∼10.7 1.72 24 ∼20
HEK-293 ∼10.7 1.72 24 ∼30
A2780 ∼7.8 1.25 24 50
FePt-SiO2-A Na2Fe(CO)4 SiO2 shell A375M, MCF7, U2OS 200 ∼2 168 0 f
FePt-A Na2Fe(CO)4 cysteamine A375M, MCF7, U2OS 30 ∼5.3 168 0 f
60 ∼10.5 72 ∼10
a
Listed are NPs’ structure, precursors, and coating; “cells” stands for the cell line; [NPs] is the nanoparticle concentration; tinc is the incubation time;
cell damage is that after incubation. b Pt precursor always Pt(acac)2. c Fe contribution from Fe2O3 shell not included. d Fe contribution from Fe2O3 shell
included. e [Fe] cannot be calculated. f Data presented in this work.

Figure 3A2 displays T2-weighted MRI images of various reported to induce significant cell viability damage of over 50%
concentrations of fcc-FePt-A NPs in agarose gels. The signal even at concentrations of NPs as low as 1.5 and ∼8.3 µg/mL,
intensity reduces and the image appears darker as the NPs respectively.37,39 A recent toxicity study with phospholipids-
concentration increases. The FePt NPs produce an observable coated fcc-FePt NPs showed 50% cell viability damage above
change in image contrast at concentrations as low as 6 µg/mL, 1.25 µg/mL Fe concentration (∼7.8 µg/mL Fe40Pt60 NPs).52 In
which is well within the safety range indicated by the cytotox- this latter study, the high toxicity was attributed to fast Fe release
icity data (Figure 2A). Similarly, signal intensity loss was from phospholipids-coated fcc-FePt NPs. It was reported that
observed in cells labeled with fcc-FePt-A NPs as the cell density ∼6% Fe was already released after 6 h of incubation, and the
increases from 10 × 103 to 1000 × 103 cells/mL (Figure 3B). percentage of Fe released increased to ∼20% after 24 h of
The in ViVo potential of FePt NPs as MRI contrast agents incubation in pH 4.8 PBS.52 While iron is essential for almost
was demonstrated by injecting NPs in cell culture media into all living organisms, it is toxic. It can act as a catalyst in the
the eyes of day 4 quail embryos in oVo. Figure 3C,D displays Fenton reaction, which generates a wide range of free radical
3D MRI image data sets after injection. The transverse and species, including hydroxyl radicals. These are among the most
coronal images through the embryo’s head are presented. The reactive free radical species known and have the ability to react
uninjected eye (un-e) acts as an internal control. There is no with a wide range of cellular constituents, including pyrimidine
difference in the contrast between the uninjected and the bases of DNA, and also to trigger lipid peroxidation in cellular
uppermost eye (in-e) which had been injected with a 1 µL of membranes, leading to cell death.53
culture medium only (Figure 3C2,C3). However, when the In this study, the fcc-FePt-silica-A NPs have good biocom-
injection medium contained 20 µg/mL of fcc-FePt-A NPs, a patibility and, as expected, the silica coating forms an inert shell
very distinct hypointensity appears in the injected eye (Figure (SI-Figure 5C).43 In contrast to previously published results,
3D2-D3). This was consistently observed when varying the our study demonstrates that cysteamine-coated fcc-FePt NPs
NPs concentration. also have very high chemical stability in pH 4.8 PBS. Fe release
This thus demonstrates the capability of nontoxic FePt NPs after 24 h of incubation is only ∼0.3%, with only a slight
as potent MRI probes for cellular imaging, even at very low increased to 0.6% after 7 days of incubation (Figure 2B1). In
NP concentrations. Labeling the cells and these magnetic NPs an in Vitro model (i.e., cell medium containing sodium
will allow, for instance, cell migration to be studied, with their citrate),48,49 fcc-FePt-A NPs were found to release as little Fe
biodistribution and behavior monitored in ViVo, especially as as Feridex (Figure 2B2), with ∼5% Fe found to be released
we have demonstrated that the effect of contrast agents can be after 24 h and 20% after 7 days. This confirms that the low pH
observed at non-cytotoxic concentrations lower than 30 µg/mL. in the endosome/lysosome environment is not sufficient to
dissolve the NPs and that the presence of endogenous citrate
3. Discussion
or similar metallic chelates is instrumental to the NPs degrada-
tion by binding to solubilized Fe atoms.48,49 The results
The major hurdle preventing the wider use of FePt NPs in presented herein are significant as they suggest that, when
biomedicine is their potential toxicity. In the literature, there considering the Fe release, the safety of fcc-FePt-A NPs is
are many inconsistencies in FePt cytotoxicity, leaving the subject comparable to that of Feridex under similar experimental
rather confused. A summary of the cytotoxicity data of FePt conditions. The chemical stability of the fcc-FePt-A NPs is also
NPs is presented in Table 2. While it was reported that 24 h supported by the Prussian blue staining experiments (Figure
incubation of FePt-cysteine NPs had no significant toxicity at 2C1-C3).
concentrations below 5 µg/mL, these compounds resulted in
50% cell damage at 15.5 µg/mL;39 FePt-Fe3O4 NPs showed no (52) Xu, C.; Yuan, Z.; Kohler, N.; Kim, J.; Chung, M. A.; Sun, S. J. Am.
Chem. Soc. 2009, 131, 15346–15351.
significant cell viability damage up to 3 days of incubation at (53) Crichton, R. R.; Wilmet, S.; Legssyer, R.; Ward, R. J. J. Inorg.
10 µg/mL.37 In contrast, FePt-Fe2O3 and FePt-CoS2 were Biochem. 2002, 91, 9–18.
J. AM. CHEM. SOC. 9 VOL. 132, NO. 42, 2010 15027
ARTICLES Chen et al.

The high chemical stability of cysteamine-coated FePt NPs being toxic. It then appears that, to be able to compare
certainly contributes to the NPs’ noteworthy low cytotoxicity. cytotoxicity data in a meaningful way, one must to develop and
Of course, iron release is only one parameter that contributes use standardized approaches including the correlation between
to toxicity. It is quite possible that differences in the NPs (i) the evolution of the nanocolloids’ integrity, relying on both
preparation, surface coating, ligand exchange, and extraction the inorganic NPs and the organic stabilizers, and (ii) the cell
protocols could account for the spread of the cytotoxicity data viability. It should be noticed that, in the context of apparently
reported in the literature. The toxicity may not necessarily be toxic NPs, the nanocolloids’ solution stability could easily be
due to the material itself through the release of its constituent. measured by, for instance, time-resolved transmission monitored
In the present study, we used Collman’s reagent, Na2Fe(CO)4, sedimentation experiments, as while the ligands come off the
to control the FePt NPs stoichiometry and to ensure the NPs’ surface, the NPs tend to aggregate and fall out of solution.
simultaneous presence of Fe and Pt species during the formation The cellular internalization of Feridex is poor due to the weak
of FePt alloy NPs; the use of Fe(CO)5 has been shown to form binding of the neutral dextran coating to the plasma membrane,
NPs with a core and a shell rich in platinum and iron, which limits the capability of cell internalization by the fluid-
respectively.31 After ligand exchange, the extraction process was phase endocytosis pathway.57,58 Enhancing cellular uptake of
repeated up to six times to ensure removal of hazardous dextran-coated NPs is possible with transfection agents and
chemicals such as chloroform, oleic acid, and oleylamine, while further surface functionalization but was not relevant to the
the thiol group provides a strong bond between the cysteamine present study.44 In contrast, the amine-coated FePt NPs, fcc-
and the NPs’ surface, preventing the ligands from becoming FePt-A and fcc-FePt-Silica-A, are efficiently internalized directly
lethal. During the review of this publication, cysteamine-coated without requiring the use of any external mediation such as
FePt NPs were reported to induce, after 24 h, very low cell transfection agents or electroporation, illustrated with non-
damage on Veron cells;38 however, it is noticeable that the phagocytic human cells in Figure 2D1. The uptake appears to
strong bonding to the surface of the NPs, protecting them from be mediated via macropinocytosis, a nonspecific endocytic
the cells’ environment, can be contrasted with, for instance, a process, which involves the internalization of large areas of
coating made with a bilayer structure of phospholipids and oleic plasma membrane together with significant amounts of fluid
acid/oleylamine.52 Indeed, even though this phospholipid ap- (Figure 2D2). Irregular macropinosome vesicles larger than 1
proach provides aqueous solubility, it is also characterized by µm are usually generated when membrane protrusions fuse back
a weaker bonding strength and a more dynamic behavior, to the plasma membrane (SI-Figure 6A).43,50,51 Other pathways
allowing the coating to slowly disperse in the cell. By doing could be involved, e.g., lipid raft-dependent mechanisms or the
so, the “free” ligands can alter the cell viability as well as the clathrin-dependent pathway;50,51,59,60 however, we did not
release of the NP constituents as the surface of the particles is observe any evidence supporting their involvement. Further
exposed to the cell, and the NPs’ chemical integrity can be studies are needed to clarify the exact cellular internalization
compromised further. Such outcomes have been reported by mechanism. However, and as expected once inside the cells,
Xu et al., although they elegantly focused on the opportunity the FePt NPs that are stabilized by Coulombic interactions
this provided to use the NPs as metal reservoirs to kill cancer appear to be confined in specific subcellular organelles, most
cells.52 likely endosomes and lysosomes (Figure 2D3). This is of
While the very high chemical stability of fcc-FePt-A NPs significant importance when developing FePt NPs-based bio-
was demonstrated in this report, cytotoxicity is observed at high medical platforms and can be attributed to the interplay between
concentrations above 30 µg/mL FePt and long incubation times. the cationic surface of the NPs and the negatively charged cell
This could be explained by contributions from (i) the very small membrane.
release of Fe, which has been found to be <1% after 168 h and By considering a family of FePt NPs, we established that,
(ii) the high concentration of positive surface charges, which despite the attractiveness of the silica coating, it weakens the
could also interact with the more negatively charged cell NPs’ magnetic moment and their applicability in clinical MRI.
membranes and induce membrane disruption.54 In addition, This may be the result of a reduction of the magnetic effective
ligands with strong anchoring groups can reduce the toxicity, volume and/or an alteration of the material. In support of this
as illustrated in this publication, and it is noticeable that the interpretation, the strong base environment associated with the
present results were obtained with commercially available silica coating can partially oxidize the FePt surface, resulting
ligands (i.e. cysteamine), so one should expect enhanced in a thin layer of softer magnetic material like iron oxide or
viability from, for instance, custom-made dithiolate ligands, such iron silicide, which because of its thinness is not visible by XRD
as those developed recently for quantum dots.55,56 These could or TEM.61,62
provide cell viability at NP concentrations higher than 30 µg/ The fcc-FePt-A NPs demonstrated a very high MRI relaxivity
mL. r2, which can be attributed to the combination of (i) an
In this context, the little consistency observed when consider- intrinsically high magnetic moment, (ii) a thin organic coating,
ing FePt NPs’ cytotoxicity data available in the literature stresses
that the measured toxicity should always be associated with the (57) Arbab, A. S.; Bashaw, L. A.; Miller, B. R.; Jordan, E. K.; Lewis,
nanocolloids, i.e. inorganic NPs + organic stabilizer. The B. K.; Kalish, H.; Frank, J. A. Radiology 2003, 229, 838–846.
(58) Arbab, A. S.; Yocum, G. T.; Wilson, L. B.; Parwana, A.; Jordan, E. K.;
interplay between the components will indeed contribute to Kalish, H.; Frank, J. A. Mol. Imaging 2004, 3, 24–32.
the cytotoxicity results, with, for instance, a weak bonding to (59) Dausend, J.; Musyanovych, A.; Dass, M.; Walther, P.; Schrezenmeier,
the NPs’ surface increasing the probability of the nanocolloids H.; Landfester, K.; Mailander, V. Macromol. Biosci. 2008, 8, 1135–
1143.
(60) Raffa, V.; Ciofani, G.; Vittorio, O.; Riggio, C.; Cuschieri, A.
(54) Bilensoy, E. Exp. Opin. Drug DeliV. 2010, 7, 795–809. Nanomedicine 2010, 5, 89–97.
(55) Han, G.; Mokari, T.; Ajo-Franklin, C.; Cohen, B. E. J. Am. Chem. (61) Lee, D. C.; Mikulec, F. V.; Pelaez, J. M.; Koo, B.; Korgel, B. A. J.
Soc. 2008, 130, 15811–15813. Phys. Chem. B 2006, 110, 11160–11166.
(56) Mei, B. C.; Susumu, K.; Medintz, I. L.; Mattoussi, H. Nat. Protoc. (62) Thomson, T.; Terris, B. D.; Toney, M. F.; Raoux, S.; Baglin, J. E. E.;
2009, 4, 412–423. Lee, S. L.; Sun, S. J. Appl. Phys. 2004, 95, 6738–6740.
15028 J. AM. CHEM. SOC. 9 VOL. 132, NO. 42, 2010
Engineered Biocompatible Nanoparticles ARTICLES

and (iii) their small size, resulting in effective dispersion across pinocytosis mechanism. The 7.1 T MRI studies, in Vitro and in
a sample. While this combination created very effective T2 MRI ViVo, have confirmed that both fcc-FePt-A and fcc-FePt-silica-A
contrast agents even at µg/mL concentrations, these results also NPs are stronger T2 contrast agents than commercial Feridex,
underline the importance of engineering the interface of NPs with fcc-FePt-A T2 relaxivity (r2) being more than 6 times larger.
and demonstrate that a strong magnetic moment such as that of The superior capability of MRI contrast enhancement associated
FePt, combined with a thin coating layer having an open with the FePt NPs makes them an ideal platform for the design
molecular structure allowing very close aqueous proximity, are of diagnostic and therapeutic agents for image-guided magnetic
essential parameters for the design of new generations of drug delivery and hyperthermic tumor ablation. The findings
diagnostic and therapeutic MRI contrast agents. of this study underline the importance of engineering magnetic
Finally, we demonstrate that FePt NPs can be successfully
NP interfaces combining intrinsically strong magnetic moment
used as a potent cellular and in ViVo MRI probes. This is
with coating layers. Finally, our studies have also demonstrated
illustrated by both cell MRI (Figure 3B) and distinct hypointen-
sity of ∼1 mm in size observed in the in ViVo environment into that FePt NPs can be used successfully for cellular and in ViVo
which the FePt NPs were injected (Figure 3D and SI-Figure 8).43 imaging, thus confirming the potential of FePt NPs in regenera-
Their more than 6 times higher relaxivity r2 allows using less tive medicine and stem cell therapy.
NPs than with iron oxide contrast agents such as Feridex, which Acknowledgment. The authors gratefully acknowledge the
makes FePt NPs very promising contrast agents to identify small Scottish Universities Physics Alliance for funding of a SUPA
lesions such as lymph node metastases. Advanced Research Fellowship (P.A.) and the James and Enid Nicol
Trust for funding a studentship (S.C.). The authors thank Dr. Lorna
4. Conclusion Eades (EaSt CHEM, University of Edinburg) for the ICP-OES
In conclusion, we have presented the synthesis and charac- measurements, Prof. Kate Storey (University of Dundee) for use
terization of a family of FePt NPs: fcc-FePt-A NPs and fcc- of the microinjection equipment, Dr. Steve Francis (University of
FePt-silica-A NPs. The reduction of magnetic properties ob- St Andrews) for the FTIR measurements, and Profs. David Cole-
served with both SQUID magnetometry and MRI relaxivity of Hamilton and Peter Bruce (University of St Andrews) for discussion
silica-coated NPs is attributed to the formation of very thin layers and access to instruments. S.L.D. acknowledges the Wellcome Trust
of oxides and silicides. In sharp contrast with some previous (WT081039) for funding.
reports, our biocompatibility studies clearly demonstrate and
explain the absence of cytotoxicity at concentrations below 30 Supporting Information Available: Experimental section
µg/mL, even after 7 days of incubation. The biocompatibility including the NPs synthesis and characterization, protocols,
is mainly attributed to the strong surface coating and the cytotoxicity, cellular uptake, and MRI application data. This
extraction protocols. Another striking feature for cellular imag- material is available free of charge via the Internet at http://
ing is the ability of the FePt NPs to efficiently enter tumor cells pubs.acs.org.
without requiring any external mediation. The cellular uptake
by non-phagocytic tumor cells occurs via an apparent macro- JA106543J

J. AM. CHEM. SOC. 9 VOL. 132, NO. 42, 2010 15029


Engineered Biocompatible nanoParticles for in vivo Imaging Applications

Shu Chen,a,b Lijun Wang,c Suzanne Duce,d Stuart Brown,c Stephen Lee,a Andreas Melzer,c Sir Alfred Cuschieri,c Pascal André a*
a
School of Physics and Astronomy (SUPA), University of St Andrews, St Andrews KY16 9SS (UK)
b
School of Chemistry (EaStChem), University of St Andrews, St Andrews KY16 9ST (UK)
c
Institute for Medical Science and Technology, University of Dundee, Dundee DD2 1FT (UK)
d
College of Life Sciences, University of Dundee, Dundee DD1 5EH (UK)

* Pascal.Andre@st-andrews.ac.uk, a.cuschieri@dundee.ac.uk

Supplementary Information

SI-I. Experimental Section surface was further functionalized with (3-aminopropyl)


SI-I.1. FePt nPs preparation triethoxysilane.4 Reverse microemulsions were prepared by
Materials: All chemical reagents, unless otherwise stated, mixing under vigorous stirring 10 mL cyclohexane, 1.3 mL NP-5
were purchased from Sigma, used without further purification but and 50 µL DI H2O. 2 mg (~ 3-4 nmol) of FePt nPs were then
degassed before use: Disodium tetracarbonylferrate-dioxane dispersed in 1 mL cyclohexane and added dropwise into the
complex (Na2Fe(CO)4•1.5 C4H8O2), platinum(II) acetylacetonate reverse microemulsion. After 15 min, 80 µL TEOS was added
(Pt(acac)2, 97 %), oleylamine (70 %), oleic acid (90 %), dropwise. After another 15 min, 150 µL NH4.H2O (28-30 %) was
cysteamine (CA, HSCH2CH2NH2, BioChemika, ≥ 98.0 %), added dropwise. The solution was kept under constant stirring at
tetraethylorthosilicate (TEOS, Si(OC2H5)4, GC, ≥ 99.0 %), (3- for 72 h. To form amine functionalized FePt-silica nPs, 100 µL of
Aminopropyl)triethoxysilane (APTES, H2N(CH2)3Si(OC2H5)3, ≥ APTES was added after 48 h and kept stirring for another 24 h.
98 %), IGEPAL® CO-520 (NP-5, (C2H4O)n•C15H24O, n ~ 5). The nPs were precipitated by centrifugation after addition of 3
When available ACS grade was chosen to select the solvents: mL of ethanol and 2 mL of methanol. The nPs were redispersed
dibenzyl ether (≥ 98.0 %), hexane (99.0 %), ethanol (≥ 99.5 %), in 5 mL of ethanol and precipitated by centrifugation after
chloroform (≥ 99 %, anhydrous), methanol (≥ 99.8 %), cyclo- addition of 10 mL of hexane. This step was repeated up to 6 times
hexane (GC, ≥ 98.0 %), ammonium hydroxide aqueous solution to completely remove the surfactant. FePt-silica nPs were stable
(NH4OH, 28.0 - 30.0 %). both in ethanol and DI water, while FePt-silica-A nPs were stable
in deionized water.
FePt nPs synthesis: all the syntheses were carried out inside a
glove box. A mixture of Pt(acac)2 (1 mmol), oleyl amine (8 SI-I.2. Experimental Techniques
mmol) and oleic acid (4 mmol) in 10 mL of dibenzyl ether was TEM: Transmission electron microscopy (TEM) images were
placed in a 50 mL round bottom flask connected to a condenser. recorded using a Gatan CCD camera on a JEOL JEM-2011
Under stirring, the mixture was heated up to 100 °C for 1 h to electron microscope operating at 200 kV. The chemical
remove oxygen and moisture, then Na2Fe(CO)4 (1 mmol) in 10 composition of FePt nPs was examined with energy-dispersive X-
mL dibenzyl ether mixture was added and heated up to 150 °C for ray spectroscopy (EDX) using an Oxford Link system installed in
1h. The mixture was further heated up to reflux ~ 300 °C to for 3 the JEM-2011 microscope.
h. The dark solution was cooled down to room temperature and
after washing with hexane and ethanol, the nPs were collected by XRD: Wide-angle powder X-ray diffraction (XRD) data were
centrifugation.1-2 collected on a Stoe STADI/P powder diffractometer operating in
transmission mode and with a small angle position sensitive
fcc-FePt-A: Ligand exchange experiments were carried out in detector. Incident radiation was generated using a FeKα1 source
a glove box according to a modified protocol based on the (λ=1.936 Å). The strongest (111) peak was fitted with Lorentzian-
literature.3 For a typical experiment, 30 mL hexane was added shaped peaks using STOEwinXpow and Kaleida-Graph softwares
into 3 mL (13.5 mg/mL) of as-synthesized oleic acid/ oleylamine to precisely determine the diffraction peak positions. The
coated fcc-FePt hexane solution. To remove the excess of ligands, crystalline grain size, DXRD, of the FePt nPs was calculated
the nPs were precipitated by addition of 60 mL ethanol and according to Scherrer’s formula:5
collected by centrifugation. The nPs were redispersed in 15 mL of 0.9λ
D XRD = (eq. 1)
chloroform assisted with sonication. 15 mL of cysteamine in B cos θ
methanol solution (0.5 M) was added dropwise to the FePt where DXRD is the “ average” dimension of the crystallites, λ is
NPs/CHCl3 mixture under vigorous stirring. The final solution the wavelength of the X-ray source (for Fe source is equal to
was left under stirring for 48 h to complete the ligand exchange. 0.193604 nm), B is the full width at half maximum of the peak
To extract the nPs, they were redispersed in 10 mL ethanol and intensity, θ is the glancing angle.
sonicated, 20 mL hexane was added and the solution was The atomic composition of FePt NPs was calculated based on
centrifuged. This process was repeated twice to remove CHCl3, the linear relations between lattice constant, a, and Fe percentage,
methanol and the excess of ligands. The nPs were then Fe %, reported by Bonakdarkpour et al.6
redispersed in 10 mL ethanol and 20 mL deionized water mixture, From SI-Figure 1, the following equations can be obtained:
sonicated and collected by centrifugation. This step was repeated x%Fe < 40 % a = - 0.0014 x%Fe + 3.929 SI-eq. (1)
6 times to obtain positively charged nPs stable in deionized water x%Fe > 40 % a = - 0.0041 x%Fe + 4.039 SI-eq. (2)
for more than 8 months (SI-Figure 1).

fcc-FePt-silica-A: The fcc-FePt-silica-A nPs were prepared by


hydrolysis of tetraethylorthosilicate (TEOS) and the silica shell

S. Chen et al. March-July 2010 S 1/6


3.95 5, the weight concentration of FePt-silica-A nPs at 100 µg/mL
corresponds then to ~ 0.02 mM of Fe in molar concentration.
3.90

a (Angstrom)
SI-I.3. Cell culture
3.85
Materials: Human breast adenocarcinoma cell line MCF7
3.80 (ATCC; Cat# HTB-22), human osteosarcoma cell line U2OS
(ATCC; Cat# HTB-96) were grown in Dulbecco’s Modified
3.75 Eagle Medium (DMEM; GIBCO, Invitrogen, Paisley, UK).
Fe Pt (%) Human malignant melanoma cell line A375M was kindly
x 100-x
3.70
0 20 40 60 80 100 provided by Dr Daniele Bergamaschi (Institute of Cell and
Molecular Science, Barts and The London, Queen Mary’s School
SI-Figure 1. FePt lattice constant (a) analysed by XRD vs. composition
curve reported by Bonakdarkpour et al. 6 of Medicine and Dentistry) and was grown in RPMI-1640
medium (GIBCO, Invitrogen). All media were supplemented with
SQUID: A 5.0 Tesla Superconducting Quantum Interference 10 % fetal calf serum, 2 mM glutamine, 100 IU/mL penicillin,
Device (SQUID) from Quantum Design (MPMS XLTM) was used and 100 µg/mL streptomycin. Cells were grown under standard
to characterize the nPs magnetic properties. The nPs were cell culture conditions in 5 % CO2 at 37 °C to reach confluence of
dispersed in polyvinylpyrrolidone matrix (VPolymer/VnPs = 20) to 60-70 % before subjected to any further treatments.
prevent interaction between the nPs while the resulting sample
was loaded into a low background gelatin capsule. Zero-Field Cellular labelling/uptake of FePt nPs and TEM: Cell culture
Cooled and Field Cooled (ZFC/FC) measurements were media were removed and replaced with the media containing
completed as follow: the sample was first cooled from room FePt-A (30 µg/mL). For control experiments, media without FePt-
temperature to 2 K without any external field, next a small field A was used. After 12-16 h of culture, cells were washed and fixed
100 Oe was applied and the nPs magnetization was recorded as with 4 % (w/v) paraformaldehyde/2.5 % (v/v) glutaraldehyde
the temperature was increased up 275 K. The FC curve was buffered in 0.2 mol/L PIPES as a standard buffer solution. After
obtained by cooling the sample back to 2 K under a 100 Oe 30 min in fixative the cells were scraped off the flasks in1 mL of
magnetic field. The magnetization was then measured while the fixative, transferred to Eppendorf tubes and cetrifuged at 14000 g
temperature was increased up to 275 K. Hysteresis measurements for 20 min. Pellets were washed in 0.2M PIPES and post fixed in
were completed at temperatures of 2 K and 300 K. The 1 % aqueous osmium tetraoxide for 1 h, washed in water, and
magnetization of the gelatine capsules and the PVP matrix was dehydrated in graded ethanol and propylene oxide before
subsequently subtracted. embedding in Durcupan resin (Sigma) and polymerisation at 60
°C for 24 h. 70 nm sections were cut on a Leica UCT
ICP-OES: FePt nPs were dissolved by addition of ultramicrotome, mounted on Pioloform coated 100 mesh copper
hydrochloric acid (Trace SELECT®, ≥ 37 %) and nitric acid grids, stained with uranyl acetate and lead citrate before being
(Trace SELECT®, ≥ 69.0 %) mixed in 3:1 as volume ratio and left examined in a Tecnai 12 electron microscope. Images were
overnight at room temperature. After full dissolution of the nPs, recorded in Digital Imaging Plates and scanned in a Ditabis
the solution was further diluted with deionised water to prepare Micron scanner (Pforzheim, Germany).
inductively coupled plasma-atomic emission spectrometry
measurement (ICP-OES, Perkin Elmer Optima 5300 DV). Cell viability study by MTS assay: The MTS CellTitre 96
The conversion of the weight concentration of FePt metal core AQueous One Solution Cell Proliferation Assay (Promega,
into Fe molar concentration was completed according to the Southampton, UK) was used to determine cell viability in control
following equation: and FePt nPs treated cells. This colorimetric assay is based on the
C R conversion of the 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxy
[Fe] = FePt - A Fe (eq. 2) methoxy-phenyl)-2-(4-sulfophenyl)-2H tetrazolium (MTS)
M w- Fe
compound to a colored, soluble formazan product in
where [Fe] (mM) is the molar concentration of iron, CFePt (µg/mL)
metabolically active cells. Cells were seeded at densities 1,250-
is the FePt core concentration, Mw-Fe the molecular weight of the
10,000 cells/well in 96-well plates depending on the duration of
iron atom, RFe is the mass ratio of Fe in FePt metal core. The mass
incubation time and cultured overnight. Cell culture media were
of the ligand was neglected. The FexPt1-x nPs composition was
then removed and replaced with the media containing the FePt
found to be x ~ 0.43 by XRD and ICP-OES. The mass ratio of Fe
nPs at various concentrations with the iron content being
in the nP core (RFe ~ 0.18) was calculated with the equation below:
determined by XRD and ICP-OES. For control experiments,
xM w- Fe media without FePt nPs was used. MTS assay was performed at
RFe = (eq. 3)
xM w- Fe + (1 − x )M w - Pt specific time intervals by addition of CellTiter 96® AQueous One
A similar approach as described by equation 4 was used for Solution reagent to each well, in triplicate, and further incubation
fcc-FePt-silica-A nPs: of cells with the reagent for 3 h at 37 °C in 5 % CO2. The colour
C R' development, i.e. absorbance, was read at 490 nm using a plate-
[Fe] = FePt -silica - A Fe (eq. 4) reader (E-Max; Molecular Devices, CA, USA) and absorbance
M w - Fe
values of both labelled and control cells were subtracted by that
where [Fe] (mM) is the molar concentration of iron, CFePt-silica-A is
of blank wells. Cell viability in labelled cells was expressed as
the concentration of FePt-silica-A in µg/mL, R’Fe is the mass ratio
percentage of that from the corresponding control cells.
of Fe in FePt-silica-A nPs which is ~ 0.01 according to eq. 5,
′ = yRFe
RFe (eq. 5)
Fe release experiment: 3.6 mg (~ 0.6 mg Fe) of FePt fcc-
where y is the mass ratio of FePt in FePt-silica-A nPs, i.e. [FexPt1- FePt-A nPs in aqueous solution was dispersed in a dialysis tubing
x]y[silica-A]1-y, which was measured experimentally by ICP-OES (MWCO 1000, Spectrum Laboratories, Inc.) which was further
and found to be ~ 0.052. As an illustration, according to eq. 4 and immersed into a 30 mL PBS (pH 4.8) or RPMI-1640 (20 mM
S. Chen et al. March-July 2010 S 2/6
sodium citrate, pH 4.8) bath held at a constant temperature of 37 (TI) that ranged from 100 to 15000 ms were applied and the echo
°C.7-8 Aliquot buffer solution was then sampled at 6, 12, 24, 48, time (TE) was 4 ms.
72, 168 h, and it composition was subsequently analyzed with T2 relaxation times were measured using a CPMG (Carr Purcell
ICP-OES. Meiboom Gill) spin echo imaging pulse sequence; a train of 16
echoes was acquired and the delay (τ) between 180º pulses ranged
Prussian blue staining and light microscopy analysis: To from 8 to 15 ms depending on the samples. Single exponential
gain insight in intracellular iron ions distribution, cells grown on relaxation times were calculated from experimental data. Each
glass coverslips or cytospun on glass slides were washed with sample was studied three times and average Longitudinal and
PBS to remove the excess of ferumoxides and then fixed with 4 % transverse relaxation rate calculated.
para-formaldehyde for 30 min. They were then washed with ri is defined as longitudinal relaxivity (i=1) or transverse
double distilled water (ddH2O) and incubated for 30 min with 2.5 relaxivity (i=2), and calculated with the following equation:9
% potassium ferrocyanide in 2.5 % HCl to produce Fe(III) 1 1
ri [CA ] = − (eq. 2)
ferrocyanide (Prussian blue). Samples were then counterstained Ti Tio
with nuclear fast red for cell nuclei. Cells were examined using a
where [CA] is the concentration of contrast agent, 1/Ti (i=1 or 2)
Zeiss microscope Axiovert 200 (Zeiss, Oberkochen, Germany) at
is the longitudinal or transverse relaxation rate with the presence
x20, x40 and x63 magnification and Axiovision 4.6 software
of contrast agent, 1/Tio is the relaxation rate of the medium in the
(Zeiss). Cells were considered Prussian blue positive if
absence of contrast agent.
intracytoplasmic blue granules could be identified.
The 128 x 128 x 128 three-dimensional RARE-8 (rapid
acquisition with relaxation enhancement) experiments were
Preparation of Agarose Gel Phantoms for MRI of FePt nPs
acquired with TR of 250 ms and TE between 180º pulses of 25 ms.
and FePt nP-labeled Cells: In the gel MRI study, 1 % (w/v) low
The field of view was 30 mm and in-plane resolution was 0.234
melting point agarose was dissolved in deionized water at 60-70
mm/pixel. The scanning time was 34 minutes.
°C. The desired amount of nPs, i.e. Fe concentration as
3D representation of the embryo eye and major blood vessels
determined by XRD and ICP-OES was then added to the solution
were produced using Amira® software (Visage Imaging, Inc. San
to obtain the targeted concentrations (up to 0.25 mM) and mixed
Diego, CA USA).
gently. For cellular MRI, FePt labelled melanoma A375M cells
were washed three times with PBS and harvested, suspended in
low melting point agarose gel (1 % in PBS at densities of 104,
SI-II. FePt nanoParticles Characterization
105, 106 cell/mL, respectively. Samples were then transferred into
SI-II.1. Ligand exchange
200 µL thin wall tubes. In both cases, samples were rapidly
Hexane and water are not miscible, and water which is denser
cooled on ice to form homogenous gel suspensions and kept at 4
(1.0 g/mL vs 0.7 g/mL) lies beneath the hexane phase. This
°C until further experiments.
provides a straightforward illustration of the effectiveness of the
ligand exchange protocol described in the Section SI-I.1.
Quail Eggs Preparation: Fertilized Japanese quail (Coturnix
The oleic acid and oleylamine capped FePt nPs are only
japonica) eggs were obtained from Rosedean, Huntingdon,
soluble in hexane giving a black color to the upper phase while
Cambs. The eggs were then placed vertically with air sac
the DI water remains colorless (SI-Figure 2A). In contrast, SI-
uppermost in a humidified incubator with a temperature of 38 °C,
Figure 2B presents the cysteamine capped FePt nPs which are
this was referred to as Day 0. On Day 4, the shell above the air
soluble in deionized water dispersion leading the lower phase to
sac was pierced and cut away to remove the inner shell membrane
and expose the embryo. The Day 4 embryos typically lay on their
sides, the upper eye is visible allowing the solution containing the
nPs to be injected into it using syringe with thin glass capillary
needle. The top of the eggs were re-sealed with adhesive tape and
then imaged. fcc-FePt-A nPs coated with either cysteamine or B
silica were added to cell culture media at concentrations ranging
A
from 1-100 µg/mL. A control medium without nPs was also used.
O

SI-I.4. Micro-MRI
H2N

Ligand
HO

Micro-MRI data were acquired on a Bruker Avance FT NMR exchange


NH3
spectrometer with a wide bore 7.1 Tesla magnet resonating at FePt hexane
300.15 MHz for 1H, fitted with Bruker micro-imaging magnetic
hexane HS

field gradients. A birdcage radio frequency resonator with an


internal diameter of 30 mm was used. MRI pulse sequences were water water FePt
taken from the Bruker Paravision® library. All acquisitions were
made at 19 ºC. Two acquisition sequences were collected and
averaged to improve the signal-to-noise ratio and reduce artefacts. SI-Figure 2. Chemical structure of the ligands and photographs of FePt
Relaxation measurements were determined from 128 x 128 nPs dispersions in hexane or water phase: oleic acid and oleylamine
capped FePt nPs (A), and cysteamine capped FePt (B).
axial planes across the samples with field of view of 30 mm, in-
plane resolution of 0.234 mm/pixel and image slice thickness of 1 be black whilst the upper hexane phase is remains transparent.
mm. In a typical experiment, 9 tubes were studied After having replaced oleic acid and oleylamine with cysteamine,
simultaneously, gels with no contrast agent acted as a standard. A the nPs are soluble in water solution and stable for more than 8
recycle time (TR) of 12 s was used to avoid saturation. T1 months.
relaxation times were measured using an inversion recovery The efficiency of ligand exchange was then confirmed by
(180º-TI-90º) imaging pulse sequence; 8 different inversion times Fourier transform infrared spectroscopy (FTIR). The FTIR
S. Chen et al. March-July 2010 S 3/6
spectra of nPs before and after ligand exchange are shown in SI- iron oxide or iron silicide, which because of its thinness is not
Figure 3, with all characteristic transmission peaks labelled and visible by XRD or TEM.13-14
matching the literature values.10 The upper spectra in SI-Figure 3
is characteristic of the nPs coated with oleic acid and oleylamine. SI-Table 1. SQUID data of the FePt nPs. Tb is blocking temperature, Ms
The peaks at 1552 and 1713 cm-1 can be assigned to the bidendate is the saturation moment and Hc is the magnetic coercivity of the nPs.
Sample Tb Ms Ms Hc
(-COO-) and monodendate -CO-) modes of oleic acid binding.
(2 K) (300 K) (2K)
The shoulder at 1595 cm-1 is due to the scissoring mode of the
(K) emu per g of Fe kOe
molecularly bonded oleylamine. The two peaks at 2854 and 2924 fcc-FePt-A 90 201 112 0.7
cm-1 correspond to the symmetric and asymmetric -CH2- fcc-FePt-silica-A 50 191 83 1.3
stretching modes of the oleyl group. The peak at 3005 cm-1 is
characteristic of the stretching of =C-H.
The original oleic acid and oleylamine coating of the FePt nPs SI-III. Cytotoxicity
surface was replaced by cysteamine through the ligand exchange SI-Figure 5A and B demonstrate that fcc-FePt-A nPs induce no
protocol described in Section SI-I.1. It efficiency is demonstrated cell viability damage at concentrations up to 30 µg/mL even after
by the bottom FTIR spectra in SI-Figure 3 characterised by i) the 7 days of incubation. Whilst, the same period of incubation with
FePt-oleic acid/oleylamine 60 µg/mL and 120 µg/mL nPs lead to a decrease of the cell
viability of only about 80 % and 70 % cell viability, respectively.
-CO- The results obtain with MCF7 and U2OS cells are consistent
Transmittance

=C-H
-NH
2 with A375M cells data presented in the core of this publication.
-COO- As expected the silica layer preserved the cell viability, with SI-
Figure 5C showing that fcc-FePt-silica-A nPs did not decrease
-CH -
human cell proliferative activity under culture conditions at
2
FePt-cysteamine concentration up to 200 µg/mL.

-CH - -SH A B
2 15 (µg/mL) 60 (µg/mL) 15 (µg/mL) 60 (µg/mL)
-NH 30 (µg/mL) 120 (µg/mL) 30 (µg/mL) 120 (µg/mL)
2 120
MCF7 cell viability (%) 120

U2OS cell viability (%)


4000 3000 2000 1000 100 100
-1
Wavenumber (cm )
80 80
SI-Figure 3. FTIR spectra of as-synthesized nPs coated with oleic acid
60 60
and oleylamine (top) and cysteamine (bottom) after ligand exchange.
40 40
absence of -COO-M and -CO-M, =C-H peaks along with ii) the 20 20
dramatic decrease of the -CH2- stretching transmission mode. The 0 0
shoulder at 2560 cm-1 is associated with –SH,11 while the peaks at
1 3 5 7 1 3 5 7
Time (Day) Time (Day)
3350 and 3448 cm-1 are consistent with the –NH2 end group of the C 25 (µg/mL) 100 (µg/mL)
cysteamine.12 120
50 (µg/mL) 200 (µg/mL)
A375M cell viability (%)

100
SI-II.2. Silica Coating
80
The protocol associated with the silica coating was tuned to
provide homogeneous SiO2 shell as illustrated by the large scale 60

TEM image in SI-Figure 4A. In addition, careful control of the 40

experimental condition prevented the formation of silica nPs 20

without any FePt as illustrated in SI-Figure 4B. 0


1 3 5 7
Time (Day)
A B SI-Figure 5. Viability of MCF7 cells (A) and U2OS cells (B) incubated
with different fcc-FePt-A nPs concentrations (15, 30, 60, 120 µg/mL),
A375M cells (C) incubated with different fcc-FePt-silica-A nPs
concentrations (25, 50, 100, 200 µg/mL).

SI-IV. Cellular uptake


SI-Figure 6A illustrates macropinocytosis, one of the main
non-specific endocytic pathways which involves the
200 nm 50 nm internalization of large areas of plasma membrane together with
significant amounts of fluid. Large irregular vesicles called
SI-Figure 4. TEM images of FePt coated with a silica shell: low macropinosomes with size larger than 1 µm are generated when
magnification (A), higher magnification (B). membrane protrusions fuse back with the plasma membrane as
indicated by the arrows on SI-Figure 6A.
The alteration of the magnetic properties of the nPs after the
SI-Figure 6B-C present TEM images of A375M cells incubated
silica coating (SI-Table 1) is attributed to the formation of thin
with fcc-FePt-A nPs at FePt metal core concentration of 30
layers of iron oxide or iron silicide resulting in the reduction of
µg/mL and fcc-FePt-silica-A nPs at nPs concentration of 50
the magnetic effective volume and/or an alteration of the material.
µg/mL after 16 h. SI-Figure 6B shows macropinocytosis uptake
In support of this interpretation, the strong base environment
mechanism. SI-Figure 6C shows intercellular presence of fcc-
associated with the silica coating can partially oxidize the FePt
FePt-silica-A nPs. Both fcc-FePt-A and fcc-FePt-silica-A nPs
surface resulting in a thin layer of softer magnetic material like
demonstrated efficient non-phagocytic human cells internalisation
S. Chen et al. March-July 2010 S 4/6
A SI-Table 2. 1H relaxivity values (r1 and r2) for agarose gel solution
containing Feridex I.V.® , fcc-FePt-A and fcc-FePt-silica-A nPs compared
to values of some commercial contrast agent base on literature (7.1 T, 300
MHz MRI, 37 ºC, in water).9
r1 r2
Sample
(s-1mM-1) (s-1mM-1)
fcc-FePt-A 2.5 ± 1.0 887 ± 32
fcc-FePt-silica-A 0.3 ± 0.1 210 ± 3
Feridex I.V.® (Ferumoxides) 0.9 ± 0.1 148 ± 2
Feridex I.V.® as reported 1.8* 132*
B C Ferucarbotran. A® 1.6* 205*
Ferumoxtran-10® 1.4* 71*
Ferumoxytol® 2* 95*
*
Experimental uncertainty was not reported.

published. The relaxivities values obtained with Feridex are


consistent with literature’s values. In addition and as presented in
the core of the publication, it is noticeable that higher r2 values
were obtained with fcc-FePt based nPs compared with Feridex’.
500 nm 50 nm
SI-Figure 7B shows that at a magnetic field of 5 T and a
SI-Figure 6. Schematic of cellular uptake through macropinocytosis (A). temperature of 300 K, fcc-FePt-A nPs have the highest
TEM image of A375M cells after 16 h incubation with DMEM cell media magnetization of 112 emu/g of Fe; however, fcc-FePt-silica-A
solution containing 30 µg /mL of fcc-FePt-A nPs (B), 50 µg/mL fcc-FePt- nPs and Feridex appear to have similar nPs magnetizations of
silica-A nPs (C). about 80 emu/g of Fe. By design, the commercial SQUID
magnetometer highest external magnetic field is 5 T, whilst MRI
by straightforward incubation, without any external allied force
experiments were performed at 7.1 T. However, a higher nPs
such as transfection agents or electroporation.
magnetization of fcc-FePt-silica-A nPs compared to Feridex can
be expected by extrapolating the magnetization curves on SI-
Figure 7 from 5 T to 7.1 T. It is indeed noticeable that Feridex has
SI-V. MRI
already saturated at 5 T, while the silica coated FePt nPs had yet
As expected for high magnetic fields such as 7.1 T used in this
to reach saturation magnetization.
work,9 all samples (i.e. Feridex, fcc-FePt-A, fcc-FePt-silica-A)
show weak effect on T1 relaxation time of 1H in 1 % agarose gel
SI-Figure 8 illustrates the effect of injecting embryo with fcc-
water solution (SI-Figure 7A).
FePt-A nPs (1 mg/mL, SI-Figure 8, top row) and fcc-FePt-silica-
SI-Table 2 summarizes the relaxivity values r1 and r2 obtained
A nPs (27 µg/mL FePt core concentration, i.e. 560 µg/mL FePt-
in this work and compare them with iron oxide data previously

A1 B1
A

0.8
1/T (s )
-1

0.7
1

0.6

0.5

0.4
[Fe] (mM)
0.3 A2 B2
0 0.05 0.1 0.15 0.2

150
B
100
M (emu per g Fe)

50

-50

-100

-150
SI-Figure 8. Embryo injected in ovo with cell culture media (1 µL)
-40 0 40 H (kOe) containing 1 mg/mL fcc-FePt-A nPs (top row) and fcc-FePt-silica-A nPs at
FePt core concentration 27 µg/mL (bottom row). 3D surface
SI-Figure 7. T1-1 (s-1) vs. [Fe] (mM) curve which illustrates 7.1 T MRI reconstruction of embryo eyes (blue) and blood vessels (red) showing the
studies of the effect of the presence of (blue ●) fcc-FePt-A, (green ▲) fcc- position in yellow of the transverse images (A); Dorsal view of transverse
FePt-silica-A and (orange ♦) Feridex I.V.® samples on 1H longitudinal image of the embryo’s head aligned through the eyes (B). 2D slices from
(T1) relaxation time of the water in 1 % w/v agarose gel (A). Hysteresis 128x128x128 3D Rare-8 MRI data set of Day 4 quail embryo egg (TR ⁄ TE
curves of fcc-FePt-A nPs (blue ●), fcc-FePt-silica-A nPs (green ▲) and = 250 ⁄ 25 ms), field of view of 30 mm and pixel dimensions of
Feridex (orange ♦) at 300 K. The empty symbols are magnetisation values 0.234mm/pixel. Label: y = yolk, b = brain, in-e = injected eye, un-e = un-
extrapolated at 7 T (B). injected eye; scale bar indicates 1mm.

S. Chen et al. March-July 2010 S 5/6


silica-A concentration, SI-Figure 8, bottom row). SI-Figure 8A
are 3D surface reconstruction which have been processed to
highlight embryo eyes in blue, the blood vessels in red, and the
nPs in green. In both cases, the presence of the nPs solution
produced a strong hypointensity in the images clearly visible
because of the darker contrast when comparing with the eye
which has not been injected (SI-Figure 8B).

References

(1) Chen, M.; Liu, J. P.; Sun, S. J Am Chem Soc. 2004, 126, 8394-8395.
(2) Nguyen, H. L.; Howard, L. E. M.; Stinton, G. W.; Giblin, S. R.;
Tanner, B. K.; Terry, I.; Hughes, A. K.; Ross, I. M.; Serres, A.;
Evans, J. S. O. Chem Mater. 2006, 18, 6414-6424.
(3) Tanaka, Y.; Maenosono, S. J Magn Magn Mater. 2008, 320, L121-
L124.
(4) Koole, R.; van Schooneveld, M. M.; Hilhorst, J.; Donega, C. D.; 't
Hart, D. C.; van Blaaderen, A.; Vanmaekelbergh, D.; Meijerink, A.
Chem Mater. 2008, 20, 2503-2512.
(5) Azaroff, L. V., Elements of X-ray Crystallgraphy. 1st ed.; McGraw-
Hill, Inc.: New York, 1968.
(6) Bonakdarpour, A.; Wenzel, J.; Stevens, D. A.; Sheng, S.;
Monchesky, T. L.; Lo¨bel, R.; Atanasoski, R. T.; Schmoeckel, A. K.;
Vernstrom, G. D.; Debe, M. K.; Dahna, J. R. J. Electrochem. Soc.
2005, 152, A61-72.
(7) Xu, C. J.; Yuan, Z. L.; Kohler, N.; Kim, J. M.; Chung, M. A.; Sun, S.
H. J Am Chem Soc. 2009, 131, 15346-15351.
(8) Arbab, A. S.; Wilson, L. B.; Ashari, P.; Jordan, E. K.; Lewis, B. K.;
Frank, J. A. Nmr Biomed. 2005, 18, 383-389.
(9) Modo, M. M. J.; Bulte, J. W. M., Molecular and Cellular MR
Imaging. 1st ed.; CRC Press: London, 2007.
(10) Bagaria, H. G.; Ada, E. T.; Shamsuzzoha, M.; Nikles, D. E.;
Johnson, D. T. Langmuir. 2006, 22, 7732-7737.
(11) Torimoto, T.; Tsumura, N.; Nakamura, H.; Kuwabata, S.; Sakata, T.;
Mori, H.; Yoneyama, H. Electrochim Acta. 2000, 45, 3269-3276.
(12) Koji, N., Infrared Absorption Spectroscopy. 1 st ed.; Holden-Day,
Inc., San Francisco and Nankodo Company Limited, Tokyo: 1962.
(13) Lee, D. C.; Mikulec, F. V.; Pelaez, J. M.; Koo, B.; Korgel, B. A. J
Phys Chem B. 2006, 110, 11160-11166.
(14) Thomson, T.; Terris, B. D.; Toney, M. F.; Raoux, S.; Baglin, J. E. E.;
Lee, S. L.; Sun, S. J. Appl. Phys. 2004, 95, 6738-6740.

S. Chen et al. March-July 2010 S 6/6

Potrebbero piacerti anche