ArticlePDF Available

Establishment of a Trimodality Analytical Platform for Tracing, Imaging and Quantification of Gold Nanoparticles in Animals by Radiotracer Techniques

Authors:

Abstract and Figures

This study aims to establish a (198)Au-radiotracer technique for in vivo tracing, rapid quantification, and ex vivo visualization of PEGylated gold nanoparticles (GNPs) in animals, organs and tissue dissections. The advantages of GNPs lie in its superior optical property, biocompatibility and versatile conjugation chemistry, which are promising to develop diagnostic probes and drug delivery systems. (198)Au is used as a radiotracer because it simultaneously emits beta and gamma radiations with proper energy and half-life; therefore, (198)Au can be used for bioanalytical purposes. The (198)Au-tagged radioactive gold nanoparticles ((198)Au-GNPs) were prepared simply by irradiating the GNPs in a nuclear reactor through the (197)Au(n,γ)(198)Au reaction and subsequently the (198)Au-GNPs were subjected to surface modification with polyethylene glycol to form PEGylated (198)Au-GNPs. The (198)Au-GNPs retained physicochemical properties that were the same as those of GNP before neutron irradiation. Pharmacokinetic and biodisposition studies were performed by intravenously injecting three types of (198)Au-GNPs with or without PEGylation into mice; the γ radiation in blood specimens and dissected organs was then measured. The (198)Au-radiotracer technique enables rapid quantification freed from tedious sample preparation and shows more than 95% recovery of injected GNPs. Clinical gamma scintigraphy was proved feasible to explore spatial- and temporal-resolved biodisposition of (198)Au-GNPs in living animals. Moreover, autoradiography, which recorded beta particles from (198)Au, enabled visualizing the heterogeneous biodisposition of (198)Au-GNPs in different microenvironments and tissues. In this study, the (198)Au-radiotracer technique facilitated creating a trimodality analytical platform for tracing, quantifying and imaging GNPs in animals.
Content may be subject to copyright.
Establishment of a Trimodality Analytical Platform for Tracing,
Imaging and Quantication of Gold Nanoparticles in Animals by
Radiotracer Techniques
Chien-Hung Chen,
Fong-Sian Lin,
Wei-Neng Liao,
Sanching L. Liang,
Min-Hua Chen,
Yo-Wen Chen,
Wan-Yu Lin,
Ming-Hua Hsu,
§
Mei-Ya Wang,
§
Jinn-Jer Peir,
§
Fong-In Chou,
§
Ching-Ya Chen,
Sih-Yu Chen,
Su-Chin Huang,
Mo-Hsiung Yang,
Dueng-Yuan Hueng,
Yeukuang Hwu,
#
Chung-Shi Yang,*
,
and Jen-Kun Chen*
,
Institute of Biomedical Engineering & Nanomedicine, National Health Research Institutes, Miaoli 35053, Taiwan
Department of Nuclear Medicine, Taichung Veterans General Hospital, Taichung 40705, Taiwan
§
Nuclear Science & Technology Development Center and
Department of Biomedical Engineering and Environmental Sciences,
National Tsing Hua University, Hsinchu 30013, Taiwan
Department of Neurological Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei 11490, Taiwan
#
Institute of Physics, Academia Sinica, Taipei 11529, Taiwan
ABSTRACT: This study aims to establish a 198Au-radiotracer
technique for in vivo tracing, rapid quantication, and ex vivo
visualization of PEGylated gold nanoparticles (GNPs) in
animals, organs and tissue dissections. The advantages of
GNPs lie in its superior optical property, biocompatibility and
versatile conjugation chemistry, which are promising to develop
diagnostic probes and drug delivery systems. 198Au is used as a
radiotracer because it simultaneously emits beta and gamma
radiations with proper energy and half-life; therefore, 198Au can
be used for bioanalytical purposes. The 198Au-tagged radioactive
gold nanoparticles (198Au-GNPs) were prepared simply by
irradiating the GNPs in a nuclear reactor through the 197Au-
(n,γ)198Au reaction and subsequently the 198Au-GNPs were
subjected to surface modication with polyethylene glycol to form PEGylated 198Au-GNPs. The 198Au-GNPs retained
physicochemical properties that were the same as those of GNP before neutron irradiation. Pharmacokinetic and biodisposition
studies were performed by intravenously injecting three types of 198Au-GNPs with or without PEGylation into mice; the γ
radiation in blood specimens and dissected organs was then measured. The 198Au-radiotracer technique enables rapid
quantication freed from tedious sample preparation and shows more than 95% recovery of injected GNPs. Clinical gamma
scintigraphy was proved feasible to explore spatial- and temporal-resolved biodisposition of 198Au-GNPs in living animals.
Moreover, autoradiography, which recorded beta particles from 198Au, enabled visualizing the heterogeneous biodisposition of
198Au-GNPs in dierent microenvironments and tissues. In this study, the 198Au-radiotracer technique facilitated creating a
trimodality analytical platform for tracing, quantifying and imaging GNPs in animals.
Gold nanoparticles (GNPs) have been developed for uses
of sensing devices because of outstanding optical
14
and
electrical properties.
5,6
Colloidal gold based pregnancy test has
been developed and went on the market in 1980s for the
detection of human chorionic gonadotropin (hCG) in
urine.
710
In this decade, GNPs have been reported to detect
and quantify nucleic acid with high-sensitivity.
1115
In addition
to in vitro diagnostic devices, conjugating anticancer drugs with
GNPs have demonstrated the enhanced permeability and
retention (EPR) eect, which give rise to accumulate
therapeutic molecules in tumor and lead to amelioration of
therapy. The CytImmune Sciences Inc. developed the rst
GNP-based anticancer drug, CYT-6091, and completed phase I
clinical trial in 2010.
16
CYT-6091 comprises 30 nm-diameter
gold nanoparticle and immune-avoiding molecule (thiol-
derivatized polyethylene glycol) to deliver tumor necrosis
factor-alpha (TNF-α),
1719
which can be used to treat patients
with advanced-stage tumors eectively. More recently, Wang et
al. and You et al. bound doxorubicin with GNPs to treat cancer
because GNPs can carry the doxorubicin into the tumor
region.
2022
To develop diagnostic agents for in vivo
biomedical applications, Zheng and colleagues produced
Received: August 31, 2014
Accepted: November 26, 2014
Published: November 26, 2014
Article
pubs.acs.org/ac
© 2014 American Chemical Society 601 dx.doi.org/10.1021/ac503260f |Anal. Chem. 2015, 87, 601608
ultrasmall (2.5 nm-diameter) gold nanoparticles conjugated
with glutathione (GS) and/or polyethylene glycol (PEG).
2325
The ultrasmall GS-GNPs presented superior features in rapid
diusion, relatively long circulation time and predominantly
renal clearance. The distribution half-life (T1/2α)and
elimination half-life (T1/2β) of GS-GNPs are 5.0 min and 10.0
h, comparable to small-molecule diagnostic probes, suggesting
potential for clinical application.
23
As for PEG conjugated with
ultrasmall GNPs (PEG-GNPs), the nanoparticle showed
signicant increase of area under curve (AUC) in pharmaco-
kinetic analysis, which may change disposition of GNPs in
tumor region.
24
The percentage of injection dose per gram
tissue weight (%ID/g) for the PEG-GNPs (8.3%ID/g) was
three times that of GS-GNPs (2.3%ID/g) in a tumor 12 h
postinjection.
24,25
Targeting delivery of long-circulating anticancer drugs should
be a promising strategy for cancer therapy. With the EPR eect,
drugs encapsulated or conjugated with nanoparticles have been
proved eectively delivered and accumulated in solid
tumor.
1719,26
More recently, Kataoka and colleagues indicated
delivering nanomedicine through EPR eect should depend on
not only particle size but also tumor permeability.
27
When used
to treat hypervascular and hyperpermeable colon adenocarci-
noma, drug-loaded nanoparticles 30, 50, 70, and 100 nm in
diameter exhibited neither size-dependent penetration nor
dierences in tumor suppression. Regarding pancreatic
adenocarcinoma BxPC3, a type of hypovascular tumor that is
dicult to penetrate, nanomedicine 30 nm in diameter had the
highest penetration capability, which facilitated achieving the
optimal tumor suppression ecacy. Although GNPs have been
extensively applied for in vitro biomedical applications, the
particle size and surface modication of GNPs should be
carefully adjusted for in vivo use in compatible patients with
dierent tumor vasculature and permeability. To design the
optimal compatible drug formulations for dierent tumors,
evaluating how physicochemical properties of GNPs can alter
the biological responses and ecacy of the formulations is
essential. Therefore, bioanalytical approaches are necessary for
determining the pharmacokinetics and biodispositions of GNPs
in blood, tissues, and organs.
This study aims to establish and evaluate 198Au-radiotracer
technique by using PEGylated 198Au-GNPs to accomplish
trimodality analytical application of GNPs, including in vivo
imaging/tracing, rapid quantication, and ex vivo visualization
of GNPs in animals, organs, and tissue dissections. We tried to
produce 198Au-GNPs without the requirement of radioactive
precursors. Scheme 1 illustrates an approach used in this work,
in which gold nanoparticles were prepared by reducing auric
acid (H197AuCl4) based on the Turkevich method,
2830
and
were subsequently subjected to neutron irradiation in a nuclear
reactor. A particular portion of the 197Au atoms in GNP could
capture thermal neutrons, and were converted into radioactive
198Au atoms. Two types of polyethylene glycols were
subsequently immobilized onto the surface of 198Au-GNPs for
exploring biological fates of GNP formulations.
The 198Au-GNP simultaneously emits γrays (412 keV) and
beta particles (Emax = 0.96 MeV) that have a half-life of 2.69
days.
31,32
γ-Rays facilitate imaging, tracing and rapidly
quantifying GNP-based drug carriers in biological systems.
Gamma scintigraphy of 198Au-GNPs for living animals was
performed using a clinical equipment of single photon emission
computed tomography (SPECT). γ-Rays can readily penetrate
soft tissues free from interferences; thus, determining 198Au-
GNPs in biological specimens is feasible, and sample
preparation does not require labor-intensive procedures. In
terms of beta radiations emitting from 198Au-GNPs, the kinetic
energy of beta particles can sensitize commercially available
imaging plate, enabling the presence of GNPs to be visualized
through autoradiography. Autoradiography of 198Au-GNPs can
also reveal the heterogeneous biodisposition of GNPs in the
microenvironments of various dissected tissues. Therefore, we
anticipated that, by using an 198Au-radiotracer technique as an
analytical platform, we could comprehensively evaluate of the
optimal GNPs formulation for the specic tumor model.
EXPERIMENTAL SECTION
Preparation of GNPs and 198Au-GNPs. Auric acid
(H197AuCl4, Sigma-Aldrich) aqueous solution (1.0 mM, 500
mL) was heated to boil with vigorous stir and sodium citrate
solution (38.8 mM, 62.5 mL) was subsequently added for
reducing Au(III) to Au0. This solution was boiled with stirring
for another 10 min until its color turned into burgundy (λmax:
520 ±1 nm). Average size of gold nanoparticles was conrmed
by both of dynamic light scattering (DLS) spectroscopy and
transmission electron microscope (TEM). These GNPs were
encapsulated in polyethylene containers and brought to Tsing
Hua Open-pool Nuclear Reactor (THOR, National Tsing Hua
University, Hsinchu, Taiwan) for 15 min neutron irradiation.
The thermal neutron ux and fast neutron ux were 1.23 ×1012
and 2.93 ×1011 neutrons·cm2·s1, respectively. The non-
radioactive gold nanoparticles were transformed to 198Au-GNPs
through 197Au(n,γ)198Au reaction.
3234
Characterization of 198Au-GNPs. The solutions of gold
nanoparticles before and after neutron activation were
dispensed into cuvettes (SARSTEDT, Germany) with a 2
mm light path. The UVvis spectra were recorded using a
NanoDrop 2000c spectrophotometer (Thermo Fisher Scien-
tic, Wilmington, DE 19810, USA) within 300 to 840 nm.
Aliquot of gold nanoparticles (0.2 mL) after neutron activation
was ltered using an ultraltration device (Vivaspin 500, GE
Healthcare, Uppsala, Sweden) with 20 kDa molecular weight
cutomembrane at 5000 gfor 10 min. The concentrated
particulate fraction on ultraltration membrane was obtained
and subjected to a high-purity germanium (HPGe) detector
(GC1020, CANBERRA, CT, USA) to acquire gamma energy
spectrum of 198Au-GNPs. Characteristic peaks in energy
spectrum were compared with the decay scheme of 198Au.
31
Furthermore, the concentrated 198Au-GNP was resuspended
into double deionized water for determination of hydrodynamic
size. The 198Au-GNP suspension was dispensed into a cuvette
at 25 °C for 2 min of thermal equilibrium. Dynamic light
scattering measurements (Zetasizer Nano ZS, Malvern Instru-
ments, Worcestershire, UK) were performed using a HeNe
laser (633 nm) at an angle of 173°to inspect the consistency of
hydrodynamic sizes before and after neutron activation. The
Scheme 1. Approach for Preparation of Radioactive Gold
Nanoparticles
Analytical Chemistry Article
dx.doi.org/10.1021/ac503260f |Anal. Chem. 2015, 87, 601608602
above-mentioned 198Au-GNPs were further dripped and dried
on copper grids for TEM measurement (H-7650, Hitachi,
Tokyo, Japan). All of the copper grids were preserved in a dry
cabinet before analysis. Primary particle size of GNPs and
198Au-GNPs were counted using SigmaScan Pro 5 software
(Systat Software Inc., San Jose, CA) and averaged for at least
300 nanoparticles.
Surface Modications of 198Au-GNPs. Two types of
polyethylene glycols were conjugated onto the surface of 198Au-
GNPs through ligand exchange procedure. The carboxymethyl-
poly(ethylene glycol)-thiol (cPEG, Laysan Bio Inc., Alabama,
USA) and methoxyl-poly(ethylene glycol)-thiol (mPEG, NOF
Co., Tokyo, Japan) molecules with 5 kDa of average molecular
weight were, respectively, employed to exchange citrate ions on
198Au-GNPs. The cPEG or mPEG solution (2.0 mM, 0.6 mL)
was added into 30 mL of 198Au-GNPs solution for 10 min of
vigorous mixing and another 10 min of incubation. After
PEGylation, double deionized water was employed to wash
twice. The PEGylated 198Au-GNPs were consequently
suspended in double deionized water to obtain a desired
radioactivity (10 μCi in 0.1 mL) prior to intravenous injection.
Pharmacokinetics and Biodisposition. The male ICR
mice (6 weeks old) were purchased from BioLASCO (Taiwan)
and acclimated in laboratory animal center for 2 weeks at the
National Health Research Institutes (NHRI). All mice were
under 12-h light/dark cycle, 23 ±1°C, 3943% relative
humidity; water and food were available ad libitum. Treatments
and experimental protocols for this study were reviewed and
approved by the Institutional Animal Care and Use Committee
(IACUC) of NHRI (approval number: NHRI-IACUC-097045-
A). Mice were randomly divided into three groups with ve
mice in each group. The mice were anesthetized using
isourane and subsequently intravenous injected with 10 μCi
(0.37 MBq) of 198Au-GNPs, cPEG-198Au-GNPs, and mPE-
G-198Au-GNPs through tail vein. Mice were sacriced at 5 min,
30 min, 1 h, 1 day, 2 days, 3 days, 4 days, and 7 days after
dosing gold nanoparticles. Blood and tissue samples including
brain, heart, lung, liver, spleen, gastrointestinal (GI) track,
kidney, pancreas, and carcass were collected for counting γrays
of 198Au.
Determination of 198Au-GNPs in Organs. Liver, GI track
and carcass, were collected into 20 mL plastic vials; blood,
heart, lung, spleen, kidney, and pancreas were collected into 3
mL plastic tubes for measuring tissue wet weights and counting
rate (counts per minute) of 198Au. Counting rates of 198Au,
proportional to amounts of 198Au-GNPs in organs, were
determined by automatic gamma counter (2480 WIZARD2,
PerkinElmer, Turku, Finland). Data were collected for 60 s with
82 keV energy window centered at the 412 keV. Percentage of
injection dose (%ID) was used to present biodisposition of
198Au-GNPs, cPEG-198Au-GNPs, and mPEG-198Au-GNPs. The
%ID was calculated based on the radioactivity measured for
each organ divided by the initially injected radioactivity to each
mouse. For pharmacokinetic study, concentration of radioactive
gold nanoparticles in blood was presented by percentage of
injection dose per gram sample wet weight (%ID/g).
Pharmacokinetic parameters for three types of 198Au-GNPs
were analyzed by noncompartment model of WinNonlin
software (Pharsight Corp., Mountain View, CA) and results
were presented in mean ±standard deviations.
In Vivo Imaging of 198Au-GNPs by Gamma Scintig-
raphy. Tumor-bearing mice (male NU/NU, 6 weeks old,
BioLASCO, Taiwan) were established by subcutaneous trans-
plantation 2 ×106cells of C6 glioma cell line (BCRC, Taiwan)
in 200 μL of phosphate buer saline at the dorsal region of the
right thigh. Mice were subjected to gamma scintigraphy while
tumor size grew to near 2000 mm3. Mice were anesthetized
with Zoletil (12.5 mg/kg) and Xylazine (5.0 mg/kg) through
intraperitoneal injection. Then the mPEG-198Au-GNPs (60 μCi
in 50 μL) was subsequently intravenously injected for acquiring
gamma scintigraphy at one and 120 h post injection by a
clinical SPECT system (E. CAM plus HD3 Detector,
SIEMENS) equipped single camera head and pinhole
collimator (5 mm I.D.). Mice were positioned on their backs
with legs extended and 11.5 cm below the pinhole collimator
for whole body imaging. Events of 412 keV gamma photons
were accumulated for reaching 3 ×104counts to explore the
biodisposition of mPEG-198Au-GNPs.
Ex Vivo Imaging of 198Au-GNPs by Autoradiography.
Two types of tumor-bearing mice (male NU/NU, 6 weeks old,
BioLASCO, Taiwan) were established by subcutaneous trans-
plantation of C6 glioma and CT26 colon carcinoma cells (2 ×
106cells in 200 μL of phosphate buer saline) at the dorsal
region of the right thigh. Mice were injected with mPEG-198Au-
GNPs (60 μCi in 50 μL) through tail vein while tumor size
grew to near 2000 mm3. Furthermore, mice were sacriced at
144-h postinjection of mPEG-198Au-GNPs followed by
immersed into 2.5% of carboxymethyl cellulose sodium salt
(Sigma-Aldrich) solution and then frozen at 20 °C for 3 h.
Animal bodies were subjected to a cryostat (OTF-5000, Bright
Instrument Co. Ltd., England) and frozen sliced into 30 μm-
thick specimens. The sliced specimens covered with PE lm
were placed onto imaging plate (BAS-IP SR 2040, Fuji Photo
Film Co. Ltd., Japan) for sensitization in a light-shielding
cassette at 20 °C for 120 h. Imaging plates after sensitization
were subsequently scanned using an Image Analyzing System
(FLA-5000, Fuji Photo Film Co. Ltd., Japan) to obtain 16 bits
planar images with 50 μm of pixel size.
RESULTS AND DISCUSSION
Critical Role of 198Au-GNPs in Bioanalytical Studies.
Radiotracing is among the optimal methods for investigating
the absorption, distribution, metabolism, and excretion of
molecules of interests.
3538
Radioactive nuclides, such as 3H
and 14C, have been incorporated into numerous compounds
that are commercially available for tracing molecules and their
metabolites in biological systems. Recently, Georgin and
colleagues fabricated 14C-labeled multiwall carbon nanotubes
(14C-MWNT) to extend the use of radiolabeling technique for
tracing nanomaterials.
39
However, 14C emits pure beta radiation
and biodispositions of 14C-MWNT in tissue dissections can
only be presented by autoradiography. In this study, we
introduced the use of 198Au as a tracer incorporated into GNPs
to conduct a series of bioanalytical studies. By measuring the
characteristic γradiation of 198Au, we traced, imaged, and
quantied 198Au-GNPs in biological systems without encoun-
tering interference caused by matrix substances.
198Au-GNPs are eective radiotracers because of the
characteristic properties of 198Au, which simultaneously emits
γrays (412 keV) and beta particles (Emax: 0.96 MeV) with a
reasonable half-life of 2.69 days. The half-life of 198Au is
comparable to 67Ga (t1/2 = 3.26 days), 90Y(t1/2 = 2.67 days),
111In (t1/2 = 2.80 days), and 201Tl (t1/2 = 3.04 days), which have
been frequently employed for clinical diagnosis. The emitted
gamma photons can readily penetrate tissue free from
Analytical Chemistry Article
dx.doi.org/10.1021/ac503260f |Anal. Chem. 2015, 87, 601608603
interferences, enabling rapid quantication of 198Au-GNPs in
biological specimens. Gamma scintigraphy is also feasible for
tracing 198Au-GNPs in vivo because the gamma energy of 198Au
is comparable to that of 131I (364 keV, t1/2 = 8.02 days) which
has been popularly used for clinical gamma scintigraphy.
Furthermore, autoradiography is feasible for imaging 198Au
nuclide because the emission of beta particles can sensitize
imaging plate to generate two-dimensional images, and can thus
be used for elucidating the biodisposition of 198Au-GNPs in
microenvironments. Therefore, 198Au-GNPs can facilitate rapid
quantication and visualization of the dynamics of GNP-based
drug carriers in living animals, and be used as a trimodality
analytical platform for imaging, tracing and quantifying GNPs.
According to recent literatures, Katti et al. has rst prepared
radioactive gold nanoparticles in which they put gold foil in
nuclear reactor for neutron capture reaction to transform 197Au
to 198Au and followed by dissolving radioactive gold foil in aqua
regia to generate radioactive auric acid (H198AuCl4).
40
The
resulted 198Au(III) solution was further reacted with reducing
agent in a heated aqueous system to reduce 198Au(III) to form
radioactive gold nanoparticles. With this method, it is inevitable
to involve a series of chemical treatments, and would thus raise
risk of radiation exposure. In this study, we demonstrated a
direct approach to prepare 198Au-GNPs simply by irradiating
gold nanoparticles in a nuclear reactor. PEGylation of 198Au-
GNPs is conducted after the irradiation of gold nanoparticles to
avoid damage to PEG molecules from intense radiation eld in
the nuclear reactor.
Characterization of Physicochemical Properties for
198Au-GNPs. The 197Au is the only stable isotope of natural
gold element, which can be transformed to radioactive 198Au by
capturing thermal neutrons through 197Au(n,γ)198Au reaction in
a nuclear reactor. Therefore, ensuring that the physical and
chemical identities of 198Au-GNPs remain the same as those of
the original GNPs is critical. The following is a discussion of the
physicochemical properties of GNPs before and after neutron
irradiation. As shown in Figure 1a, the surface plasmon
resonance (SPR) band of GNPs at 519520 nm did not
present any perceivable wavelength shift before and after
neutron irradiation. Subsequently, ultraltration was employed
to separate the nanoparticle fraction from the neutron-activated
aqueous fraction. The radionuclidic purity of nanoparticle
fraction was measured by a high-purity germanium detector.
37
Figure 1b shows the energy spectrum of the gamma photons
emitted from the nanoparticle fraction, displaying a major peak
at 412 keV, accompanied by minor peaks at 674 and 1082 keV.
All of these peaks are in good agreement with the decay scheme
of 198Au, demonstrating perfect radionuclidic purity of 198Au-
GNPs prepared in this study.
31,32
Furthermore, transmission
electron microscopy (TEM) and dynamic light scattering
(DLS) spectroscopy were also used to conrm the size and size
distribution of gold nanoparticles before and after the nuclear
reaction. The TEM micrograph in Figure 1c shows that the
particle size and size distribution of the 198Au-GNPs (13.2 ±
1.0 nm) were comparable to that of the GNPs (13.4 ±1.0 nm)
before neutron irradiation; this similarity can be cross-validated
according to the SPR bands shown in Figure 1a. Additionally,
Figure 1d displays the hydrodynamic sizes of the GNPs and
198Au-GNPs; z-average values are 14.0 nm (PDI = 0.031) and
14.4 nm (PDI = 0.054), respectively. These results clearly
indicate that the 198Au-GNPs remained equivalent to the
original nonradioactive GNPs, and therefore can be employed
as a radiotracer for evaluating the biological fates, including
absorption, distribution, metabolism, and excretion of various
types of GNPs in biological systems.
Pharmacokinetics and Biodisposition of PEGylated
198Au-GNPs. Fluorescent tags conjugated with nanoparticles
have been frequently used for reporting the presence of
nanoparticles in cells and tissues, which facilitate in vivo tracing
of nanoparticles.
4143
In addition, chemical analysis can be
performed to evaluate concentrations of the component
elements of nanoparticles in of biological specimens. As for
gold nanoparticles, the excitation or emission wavelengths of
uorescent tags might be attenuated by GNPs or masked by the
autouorescence of living animals. For chemical analysis, acidic
digestion of tissue samples is necessary before gold can be
quantied using atomic emission spectroscopy or mass
spectrometry, which cannot be used to determine spatial- or
temporal-resolved information on GNPs in tissues.
44,45
In this study, we used an 198Au-radiotracer to rapidly quantify
of GNPs in biological specimens without encountering the
aforementioned drawbacks. The PEGylated-GNPs were used as
model drug carriers by conjugating the 198Au-GNPs with
carboxymethyl-poly(ethylene glycol)-thiol (cPEG) and me-
thoxyl-poly(ethylene glycol)-thiol (mPEG) molecules, respec-
tively. As is well-known, PEG-conjugated pharmaceuticals can
provide a shielding layer to prevent nonspecic adhesion of
numerous molecules in blood and avoid capturing by immune
system, which consequently lead to prolonging circulation of
PEGylated molecules.
46,47
Since PEGylation is necessary to
render GNP a practical anticancer carrier,
1719
both cPEG and
mPEG molecules (averagely 5 kDa of molecular weight) were
immobilized on 198Au-GNPs in this study through AuS
binding. The material characterization indicated that the
hydrodynamic sizes of cPEG-198Au-GNPs and mPEG-198Au-
GNPs were 34.8 ±3.4 and 37.3 ±1.3 nm, respectively. The ζ
potential values were 39.1 ±8.9 and 26.5 ±8.5 mV for the
cPEG-198Au-GNPs and mPEG-198Au-GNPs, respectively.
The eectiveness of using 198Au-radiotracing techniques for
pharmacokinetics and biodisposition analysis was evaluated.
Three types of 198Au-GNPs (with dierent surface moieties)
Figure 1. Physicochemical characterization using (a) UVvis spectra
of bare GNPs before and after neutron irradiation, (b) gamma photon
energy spectrum of bare GNPs after neutron irradiation, (c)TEM
micrograph of bare GNPs after neutron irradiation, and (d) particle
size determined by dynamic light scattering before and after neutron
activation.
Analytical Chemistry Article
dx.doi.org/10.1021/ac503260f |Anal. Chem. 2015, 87, 601608604
were injected intravenously into mice, followed by withdrawing
blood for counting 412 keV gamma photons. Figure 2 displays
the concentration proles of bare 198Au-GNPs, cPEG-198Au-
GNPs, and mPEG-198Au-GNPs, in which the concentrations of
GNPs in blood are shown as the percentage of the injected
dose per gram of blood (%ID/g). The concentration of
bare198Au-GNPs in the blood diminished rapidly, and was
approximately 2.0% ID/g at 5 min postinjection. By contrast,
concentrations of cPEG-198Au-GNPs and mPEG-198Au-GNPs
in blood were maintained at the level of 30% ID/g at 5 min
postinjection. Therefore, we could estimate 60% of injected
PEGylated-GNPs persisted in blood because of nearly 2 g of
total blood in each mouse. Through radioactivity measurement,
it also showed bare radioactive GNPs were rapidly washed out
of blood within less than 5 min and very quickly accumulated in
liver (Table 2). Table 1 lists comparative results of
pharmacokinetic parameters of bare198Au-GNPs, cPEG-198Au-
GNPs and mPEG-198Au-GNPs formulations. As a drug carrier,
signicant improvements occurred for both of cPEG-198Au-
GNPs and mPEG-198Au-GNPs in comparison to bare 198Au-
GNPs. Obviously, two PEGylated-198Au-GNPs show higher
maximum concentration (Cmax) and larger area under curve
(AUC) than the bare 198Au-GNPs. However, evaluating
whether the cPEG-198Au-GNPs or mPEG-198Au-GNPs are
more eective drug carriers only according to pharmacokinetic
information is insucient.
In addition to pharmacokinetic data, biodisposition informa-
tion is required. Table 2 lists biodisposition data for the bare
198Au-GNPs, cPEG-198Au-GNPs, and mPEG-198Au-GNPs,
showing the percentage of injected dose (% ID) in each
organ at 1 h, 1 day, and 1 week after administration. There was
more than 85% ID of bare 198Au-GNPs accumulated in liver,
persisting for 7 days, which was in good agreement with rapid
decrease of bare 198Au-GNPs in blood (Figure 2). By contrast,
less than 15% ID of PEGylated-198Au-GNPs presented in liver
at 24-h postinjection, accompanied by more than 10%ID/g of
cPEG-198Au-GNPs and mPEG-198Au-GNPs in blood indicated
the practice of prolong circulation. Retention of cPEG-198Au-
GNPs and mPEG-198Au-GNPs in liver, spleen, and carcass
gradually increased for the period from 1 h to 7 days after
administration. More than 85% ID of cPEG-198Au-GNPs and
mPEG-198Au-GNPs were distributed in liver, spleen, and
carcass at 7-day postinjection. For spleen, there was no
signicant dierence between cPEG-198Au-GNPs and mPE-
G-198Au-GNPs (6.4 ±1.3% ID versus 6.2 ±1.4% ID).
However, biodisposition of cPEG-198Au-GNPs (32.9 ±3.6%
ID) in liver was signicantly higher than that of mPEG-198Au-
GNPs (26.0 ±5.4% ID). Conversely, the mPEG-198Au-GNPs
exhibited a marked tendency toward higher accumulation in the
carcass than the cPEG-198Au-GNPs did (65.4 ±5.5% ID versus
49.8 ±4.5% ID). To prevent from overestimating the
biodisposition of GNPs in carcass, we count tail tissue
separated from carcass, showing very few (<3%) GNPs
remained in tail tissue. Also, we do not include radioactivity
of tail tissue into total injection dose to avoid artifact from
performing an i.v. tail vein injection. The amounts of the
cPEG-198Au-GNPs and mPEG-198Au-GNPs in the heart, lung,
kidney, and brain decreased during the period from 1 h to 7
days, which can be attributed to continuous wash-out through
blood circulation. In addition, a small portion of 198Au-GNPs
were excreted from the body within 7 days because the
recovery rates for the three radioactive GNPs in the blood,
organs, and carcass were more than 95% of the injection dose.
In comparison with published works, zinc oxide nanoparticles
(10 and 71 nm),
37,48
CdSe-based quantum dots (13 nm),
49,50
and mesoporous silica nanoparticles (50100 nm)
5153
could
be predominantly retained in liver, spleen, lung and kidney.
The above-mentioned nanoparticles might be excreted through
gastrointestinal (GI) track/feces and/or kidney/urine, which
were far dierent from the observation in this study. Not only
particle size but also physicochemical nature, particle shape,
surface modication and administration route all play a role in
excretion pathways of nanoparticles from the body.
48
Imaging 198Au-GNPs in Vivo and ex Vivo. In addition to
rapid quantication of 198Au-GNPs for pharmacokinetic and
biodisposition investigation, the 198Au-radiotracer technique is
applicable for imaging and tracing GNPs in living animals and
dissected tissues. Simultaneous emission of γrays and beta
particles facilitates imaging 198Au-GNPs by two modalities,
gamma scintigraphy by clinical SPECT and autoradiography by
βparticle responsive imaging plate. Gamma scintigraphy and
autoradiography are not only useful to verify the biodisposition
of PEGylated-198Au-GNPs but also allow screening optimal
portfolio of GNP-based drug carriers for the specic tumor-
bearing animal model. Based on above-mentioned pharmaco-
kinetics and biodisposition evaluations, mPEG-198Au-GNPs was
superior to cPEG-198Au-GNPs for treating animals with
subcutaneous tumors because of longer circulation time and
less retention in liver. Figure 3 shows the gamma scintigraphy
Figure 2. Concentration proles of bare198Au-GNPs, cPEG-198Au-
GNPs, and mPEG-198Au-GNPs in blood after intravenous injection.
Error bars demonstrate standard deviation of GNP concentration in
blood.
Table 1. Pharmacokinetic Parameters of Radioactive Gold
Nanoparticles with and without Conjugation of Polyethylene
Glycols (Mean ±SD, n=5)
a
bare198Au-
GNPs cPEG-198Au-GNPs mPEG-198Au-GNPs
T1/2 (h) 35.64 ±13.15 14.39 ±3.82 14.54 ±1.60i
C0(% ID/g) 2.22 ±1.11 37.26 ±3.19 34.99 ±3.36
AUC
(% ID h/g) 10.19 ±7.10 800.34 ±248.82 1008.58 ±102.35
CL (g/h) 13.13 ±8.66 0.13 ±0.04 0.10 ±0.01
MRT (h) 53.77 ±16.34 13.72 ±6.74 17.27 ±2.36
a
Abbreviations: T1/2 = biological half-life of 198Au-GNPs in blood; C0
= the extrapolated concentration of 198Au-GNPs in blood at t=0;
AUC = the area under the curve of blood concentrationtime curve;
CL = total body clearance; MRT = mean residence time.
Analytical Chemistry Article
dx.doi.org/10.1021/ac503260f |Anal. Chem. 2015, 87, 601608605
acquired by clinical SPECT platform after intravenous injection
of mPEG-198Au-GNPs, which demonstrate the spatial- and
temporal-resolved biodispositions of mPEG-198Au-GNPs in
glioblastoma-bearing mice. The mPEG-198Au-GNPs were
predominantly observed in the circulatory system 1-h
postadministration (left panel, Figure 3); this observation
strongly corresponds to the pharmacokinetic information
shown in Figure 2; more than 70% of the injected
mPEG-198Au-GNPs were estimated to have remained in the
blood. At 120-h postadministration (right panel, Figure 3),
mPEG-198Au-GNPs were evenly distributed in abdomen and
tumor region. The gamma scintigraphy shown in Figure 3
could be performed using a clinical SPECT imaging system.
The centimeter-level spatial resolution is unideal for tracing
GNPs in small animals. Nevertheless, using a clinical SPECT
imaging platform for tracing 198Au-GNPs in humans would be
less problematic, and more GNP-based pharmaceuticals for
clinical treatments are expected to be developed.
In addition to the γrays, the beta particles emitted from the
mPEG-198Au-GNPs were applied in autoradiography, a practical
imaging technique for exploring the biodisposition of GNPs in
microenvironments in tumors and organs. The mPEG-198Au-
GNPs were intravenously injected into mice, and then allowed
to distribute for 144 h before sacrice for collecting tissue
dissection. By placing a tissue dissection containing mPE-
G-198Au-GNPs onto a beta particles responsive imaging plate,
the kinetic energy of beta particles could be converted to
detectable signals on a planar display. Biodisposition of
mPEG-198Au-GNPs were therefore blotted from the dissected
tissue to a two-dimensional image. Therefore, we can prove the
concept of visualizing mPEG-198Au-GNPs in dissected tissues
from C6 glioblastoma (Figure 4a) and CT26 colorectal
carcinoma (Figure 4b) tumor-bearing animals to distinguish
eciency of drug delivery in dierent tumor models. As shown
in the gures, it presents half of mPEG-198Au-GNPs
successfully delivered into C6 tumor, which might imply that
C6 tumor is more hyper-vascular as compared to that of the
CT26 tumor. Regarding the biodisposition of mPEG-198Au-
GNPs in dierent microenvironments, Figure 4a reveals
absence of mPEG-198Au-GNPs in the center of tumor. Previous
studies have demonstrated the presence of necrotic center of
glioblastoma in human and animal models.
5456
We determined that the region was necrotic center because
the GNPs could not be delivered. Moreover, the concentrations
of PEGylated-198Au-GNPs in spleen were apparently higher
than that in liver for both of C6 and CT26 tumor-bearing mice
models (Figure 4). Table 2 displays the amounts of gold
nanoparticles in liver much more than that in spleen; however,
the concentration of PEGylated-198Au-GNPs in spleen should
be higher than that in liver for more than 1 day after injection
because liver weights were nearly 20 times more than spleen
weights. Results from autoradiography revealed the heteroge-
neous biodisposition of the mPEG-198Au-GNPs in livers of C6
and CT26 tumor models and aorded abundant information
on GNPs in various microenvironments. The 198Au-radiotracer
technique enables trimodality analytical applications, including
quantication, imaging, and tracing, that are unachievable
through conventional chemical analysis or uorescent imaging.
CONCLUSIONS
In this study, an approach for establishing a trimodality
analytical platform for imaging, tracing, and quantifying GNPs
in mice was developed using radiotracer techniques. 198Au-
GNPs were prepared by irradiating nonradioactive GNPs in a
nuclear reactor through the nuclear transformation of 197Au
into 198Au. The obtained 198Au-GNPs retained the same
intrinsic properties as did the original GNPs and were further
synthesized through surface modication with PEG molecules.
The PEGylated 198Au-GNPs were then injected intravenously
Table 2. Biodispositions of Radioactive Gold Nanoparticles Presenting in % ID for Each Organ (Mean ±SD, n=5)
bare 198Au-GNPs cPEG-198Au-GNPs mPEG-198Au-GNPs
1 h 1 day 7 day 1 h 1 day 7 day 1 h 1 day 7 day
liver 87.6 ±5.5 88.4 ±6.8 87.6 ±3.8 14.5 ±3.3
a
28.9 ±16.7
a
32.9 ±3.6
a
,
b
12.1 ±4.0
a
15.1 ±2.8
a
26.0 ±5.4
a
,
b
spleen 3.8 ±1.0 3.1 ±0.7 3.0 ±0.3 0.2 ±0.1
a
3.3 ±1.8 6.4 ±1.3
a
0.3 ±0.1
a
1.5 ±0.6
a
6.2 ±1.4
a
carcass 4.9 ±2.2 5.8 ±2.6 3.8 ±1.3 39.3 ±4.0
a
47.6 ±7.8
a
49.8 ±4.5
a
,
b
43.3 ±4.4
a
53.8 ±4.6
a
65.4 ±5.5
a
,
b
heart <0.1 <0.1 <0.1 0.7 ±0.2
a
0.4 ±0.1
a
0.3 ±0.1
a
0.8 ±0.3
a
0.6 ±0.3
a
0.3 ±0.1
a
lung 0.9 ±0.6 0.4 ±0.3 0.3 ±0.2 1.6 ±0.4 0.8 ±0.3 0.4 ±0.1 1.7 ±0.4
a
1.1 ±0.3
a
0.6 ±0.2
a
kidney 0.1 ±0.1 0.1 ±0.0 0.1 ±0.0 2.9 ±0.5
a
1.6 ±0.7
a
0.6 ±0.1
a
3.1 ±0.2
a
2.3 ±0.6
a
0.8 ±0.1
a
brain <0.1 <0.1 <0.1 0.3 ±0.1
a
0.1 ±0.1
a
<0.1 0.3 ±0.0
a
0.2 ±0.0
a
<0.1
GI 0.2 ±0.1 0.1 ±0.0 0.2 ±0.1 2.9 ±0.5
a
4.1 ±1.1
a
5.0 ±0.9
a
2.8 ±0.2
a
4.1 ±0.2
a
5.5 ±0.8
a
pancreas <0.1 <0.1 <0.1 0.3 ±0.0
a
0.3 ±0.0
a
0.3 ±0.1
a
0.3 ±0.0
a
0.3 ±0.1
a
0.3 ±0.1
a
recovery 97.7 ±3.6 97.9 ±6.0 95.0 ±3.8 100.0 ±5.9 96.9 ±3.3 95.7 ±3.4 101.9 ±6.1 96.6 ±5.6 105.3 ±8.9
a
Signicant dierence (pvalue < 0.05) between bare 198Au-GNPs and cPEG-198Au-GNPs or between bare 198Au-GNPs and mPEG-198Au-GNPs for
the same time postinjection.
b
Signicant dierence (pvalue <0.05) between cPEG-198Au-GNPs and mPEG-198Au-GNPs for the same time
postinjection.
Figure 3. Gamma scintigraphy done by clinical SPECT system for tail
vein injection of mPEG-198Au-GNPs on subcutaneously C6
glioblastoma-bearing nude mouse. The radioactive gold nanoparticles
were circulating in whole body at 1-h postinjection (left panel) and
were almost evenly distributed in abdomen and tumor at 120-h
postinjection (right panel).
Analytical Chemistry Article
dx.doi.org/10.1021/ac503260f |Anal. Chem. 2015, 87, 601608606
into mice, and gamma photon measurement of blood
specimens and separated tissues was performed to quantify
the GNPs. The simultaneous beta and γradiation emissions
from the 198Au-GNPs can enable exploring the dynamics and
biodisposition of GNPs in animals. Imaging and tracing GNPs
in vivo is possible through gamma scintigraphy, which implies
the potential for clinical use. In addition to being used to
determine the macroscopic distribution of GNPs, auto-
radiography enables visualizing various biodispositions in
microenvironments. 198Au-radiotracer techniques facilitate
comprehensive achievements in rapid quantication, real-time
monitoring in animals, and microscopic visualization of GNPs
in tissues; thus, further development of GNP-based drugs for
clinical applications is feasible.
AUTHOR INFORMATION
Corresponding Authors
*Fax: +886-37-586447. E-mail: cyang@nhri.org.tw.
*E-mail: jkchen@nhri.org.tw.
Author Contributions
C.-H.C., F.-S.L., amd W.-N.L. contributed equally. The
manuscript was written through contributions of all authors.
All authors have given approval to the nal version of the
manuscript.
Notes
The authors declare no competing nancial interest.
ACKNOWLEDGMENTS
This work was supported by the grants of National Nano-
science and Nanotechnology Program for the Innovative
Molecular Biomedical Nano-Imaging Open Facility, National
Health Research Institutes (NM-99-PP-11), Nuclear Science
and Technology Development Center of National Tsing Hua
University (NTHU-100N7520E1) and Ministry of Science and
Technology (NSC 101-2113-M-400-001-MY2 and 102-2113-
M-007-010) in Taiwan. This study was supported in part by a
surcharge of tobacco products, funding a grant from the
Ministry of Health and Welfare (MOHW103-TD-B-111-12) of
Taiwan. Authors are grateful of animal housing supports from
Laboratory Animal Center of NHRI (98A1-LAPP01-014) and
Eva Yu-Ching Chen for TEM analysis (NM-101-PP-04,
Electron Microscopes Core Facility, NHRI). We heartedly
thank Jeng-Yan Yang (NTHU), Chien-Tien Liu (NTHU),
Fiona Mei-Hui Shih (NHRI), and Matthew Chih-Hui Liu
(NHRI) for handling radioactive materials.
REFERENCES
(1) Cheng, S. F.; Chau, L. K. Anal. Chem. 2003,75,1621.
(2) Lai, N. S.; Wang, C. C.; Chiang, H. L.; Chau, L. K. Anal. Bioanal.
Chem. 2007,388, 901907.
(3) Darbha, G. K.; Singh, A. K.; Rai, U. S.; Yu, E.; Yu, H.; Chandra
Ray, P. J. Am. Chem. Soc. 2008,130, 80388043.
(4) Lin, J. H.; Chang, C. W.; Tseng, W. L. Analyst 2010,135, 104
110.
(5) Tseng, C. W.; Tao, Y. T. J. Am. Chem. Soc. 2009,131, 12441
12450.
(6) Chen, Y. S.; Hong, M. Y.; Huang, G. S. Nat. Nanotechnol. 2012,7,
197203.
(7) Leuvering, J. H.; Thal, P. J.; Schuurs, A. H. J. Immunol. Methods
1983,62, 175184.
(8) Vaitukaitis, J. L. Ann. N.Y. Acad. Sci. 2004,1038, 220222.
(9) Tanaka, R.; Yuhi, T.; Nagatani, N.; Endo, T.; Kerman, K.;
Takamura, Y.; Tamiya, E. Anal. Bioanal. Chem. 2006,385, 14141420.
(10) Posthuma-Trumpie, G. A.; Korf, J.; van Amerongen, A. Anal.
Bioanal. Chem. 2009,393, 569582.
(11) Glynou, K.; Ioannou, P. C.; Christopoulos, T. K.; Syriopoulou,
V. Anal. Chem. 2003,75, 41554160.
(12) Li, H.; Rothberg, L. Anal. Chem. 2005,77, 62296233.
(13) Paliwoda, R. E.; Li, F.; Reid, M. S.; Lin, Y.; Le, X. C. Anal. Chem.
2014,86, 61386143.
(14) Liu, P.; Yang, X.; Sun, S.; Wang, Q.; Wang, K.; Huang, J.; Liu, J.;
He, L. Anal. Chem. 2013,85, 76897695.
(15) Liu, Y. C.; Li, Y. J.; Huang, C. C. Anal. Chem. 2013,85, 1021
1028.
(16) Libutti, S. K.; Paciotti, G. F.; Byrnes, A. A.; Alexander, H. R., Jr.;
Gannon, W. E.; Walker, M.; Seidel, G. D.; Yuldasheva, N.; Tamarkin,
L. Clin. Cancer Res. 2010,16, 61396149.
(17) Paciotti, G. F.; Myer, L.; Weinreich, D.; Goia, D.; Pavel, N.;
McLaughlin, R. E.; Tamarkin, L. Drug Delivery 2004,11, 169183.
(18) Goel, R.; Swanlund, D.; Coad, J.; Paciotti, G. F.; Bischof, J. C.
Mol. Cancer Ther. 2007,6, 20392047.
(19) Goel, R.; Shah, N.; Visaria, R.; Paciotti, G. F.; Bischof, J. C.
Nanomedicine 2009,4, 401410.
(20) Wang, F.; Wang, Y. C.; Dou, S.; Xiong, M. H.; Sun, T. M.;
Wang, J. ACS Nano 2011,5, 36793692.
(21) You, J.; Zhang, R.; Xiong, C.; Zhong, M.; Melancon, M.; Gupta,
S.; Nick, A. M.; Sood, A. K.; Li, C. Cancer Res. 2012,72, 47774786.
(22) Gautier, J.; Allard-Vannier, E.; Munnier, E.; Souce, M.; Chourpa,
I. J. Controlled Release 2013,169,4861.
(23) Zhou, C.; Hao, G.; Thomas, P.; Liu, J.; Yu, M.; Sun, S.; Oz, O.
K.; Sun, X.; Zheng. J. Angew. Chem. Int. Ed. 2012,51, 1011810122.
Figure 4. Whole-body autoradiography was performed at 144-h
postinjection of mPEG-198Au-GNPs into (a) C6 glioblastoma-bearing
nude mouse and (b) CT26 colon carcinoma-bearing nude mouse.
Analytical Chemistry Article
dx.doi.org/10.1021/ac503260f |Anal. Chem. 2015, 87, 601608607
(24) Liu, J.; Yu, M.; Ning, X.; Zhou, C.; Yang, S.; Zheng, J. Angew.
Chem., Int. Ed. 2013,52, 1257212576.
(25) Liu, J.; Yu, M.; Zhou, C.; Yang, S.; Ning, X.; Zheng, J. J. Am.
Chem. Soc. 2013,135, 49784981.
(26) Matsumura, Y.; Maeda, H. Cancer Res. 1986,46, 63876392.
(27) Cabral, H.; Matsumoto, Y.; Mizuno, K.; Chen, Q.; Murakami,
M.; Kimura, M.; Terada, Y.; Kano, M. R.; Miyazono, K.; Uesaka, M.;
Nishiyama, N.; Kataoka, K. Nat. Nanotechnol. 2011,6, 815823.
(28) Turkevitch, J.; Stevenson, P. C.; Hillier. J. Discuss. Faraday Soc.
1951,11,5575.
(29) Frens, G. Nat. Phys. Sci. 1973,241,2022.
(30) Kimling, J.; Maier, M.; Okenve, B.; Kotaidis, V.; Ballot, H.;
Plech, A. J. Phys. Chem. B 2006,110, 1570015707.
(31) Erdtmann, G. Neutron Activation Tables; Verlag Chemie:
Weinheim, Germany, 1976.
(32) Huang, X. L. Nucl. Data Sheets 2009,110, 25332688.
(33) Isler, D.; Moeglen, C.; Gains, N.; Meier, M. K. Br. J. Nutr. 1995,
73, 851862.
(34) Semmler-Behnke, M.; Kreyling, W. G.; Lipka, J.; Fertsch, S.;
Wenk, A.; Takenaka, S.; Schmid, G.; Brandau, W. Small 2008,4,
21082111.
(35) Isler, D.; Moeglen, C.; Gains, N.; Meier, M. K. Br. J. Nutr. 1995,
73, 851862.
(36) Niu, Y. G.; Hauton, D.; Evans, R. D. J. Physiol. 2004,558, 225
237.
(37) Chen, J. K.; Shih, M. H.; Peir, J. J.; Liu, C. H.; Chou, F. I.; Lai,
W. H.; Chang, L. W.; Lin, P.; Wang, M. Y.; Yang, M. H.; Yang, C. S.
Analyst 2010,135, 17421746.
(38) Rouessac, F.; Rouessac, A. Chemical Analysis: Modern
Instrumentation Methods and Techniques; John Wiley & Sons, Ltd.:
Chichester, England, 2000.
(39) Georgin, D.; Czarny, B.; Botquin, M.; Mayne-Lhermite, M.;
Pinault, M.; Bouchet-Fabre, B.; Carriere, M.; Poncy, J. L.; Chau, Q.;
Maximilien, R.; Dive, V.; Taran, F. J. Am. Chem. Soc. 2009,131,
1465814659.
(40) Katti, K. V.; Kannan, R.; Katti, K.; Kattumori, V.;
Pandrapraganda, R.; Rahing, V.; Cutler, C.; Boote, E. J.; Casteel, S.
W.; Smith, C. J.; Robertson, J. D.; Jurrison, S. S. Czech. J. Phys. 2006,
56, D23D34.
(41) Gaumet, M.; Gurny, R.; Delie, F. Int. J. Pharm. 2007,342, 222
230.
(42) Zhou, X.; Zhou, J. Anal. Chem. 2004,76, 53025312.
(43) Kim, S. E.; Ahn, K. Y.; Park, J. S.; Kim, K. R.; Lee, K. E.; Han, S.
S.; Lee, J. Anal. Chem. 2011,83, 58345843.
(44) De Jong, W. H.; Hagens, W. I.; Krystek, P.; Burger, M. C.; Sips,
A. J.; Geertsma, R. E. Biomaterials 2008,29, 19121919.
(45) Glazer, E. S.; Zhu, C.; Hamir, A. N.; Borne, A.; Thompson, C.
S.; Curley, S. A. Nanotoxicology 2011,5, 459468.
(46) Otsuka, H.; Nagasaki, Y.; Kataoka, K. Adv. Drug Delivery Rev.
2003,55, 403419.
(47) Akiyama, Y.; Mori, T.; Katayama, Y.; Niidome, T. J. Controlled
Release 2009,139,8184.
(48) Yeh, T. K.; Chen, J. K.; Lin, C. H.; Yang, M. H.; Yang, C. S.;
Chou, F. I.; Peir, J. J.; Wang, M. Y.; Chang, W. H.; Tsai, M. H.; Tsai,
H. T.; Lin, P. Nanotechnology 2012,23, 085102.
(49) Yang, R. S.; Chang, L. W.; Wu, J. P.; Tsai, M. H.; Wang, H. J.;
Kuo, Y. C.; Yeh, T. K.; Yang, C. S.; Lin, P. Environ. Health Perspect.
2007,115, 13391343.
(50) Schipper, M. L.; Iyer, G.; Koh, A. L.; Cheng, Z.; Ebenstein, Y.;
Aharoni, A.; Keren, S.; Bentolila, L. A.; Li, J.; Rao, J.; Chen, X.; Banin,
U.; Wu, A. M.; Sinclair, R.; Weiss, S.; Gambhir, S. S. Small 2009,5,
126134.
(51) Lee, C. H.; Cheng, S. H.; Wang, Y. J.; Chen, Y. C.; Chen, N. T.;
Souris, J.; Chen, C. T.; Mou, C. Y.; Yang, C. S.; Lo, L. W. Adv. Funct.
Mater. 2009,19, 215222.
(52) Meng, H.; Xue, M.; Xia, T.; Ji, Z.; Tarn, D. Y.; Zink, J. I.; Nel, A.
E. ACS Nano 2011,5, 41314144.
(53) Huang, X.; Li, L.; Liu, T.; Hao, N.; Liu, H.; Chen, D.; Tang, F.
ACS Nano 2011,5, 53905399.
(54) Wen, P. Y.; Kesari, S. N. Engl. J. Med. 2008,359, 492507.
(55) Verhoeff, J. J.; Stalpers, L. J.; Claes, A.; Hovinga, K. E.; Musters,
G. D.; Peter Vandertop, W.; Richel, D. J.; Leenders, W. P.; van Furth,
W. R. Eur. J. Cancer 2009,45, 30743080.
(56) Jacobs, V. L.; Valdes, P. A.; Hickey, W. F.; De Leo, J. A. ASN
Neuro 2011,3, No. e00063.
Analytical Chemistry Article
dx.doi.org/10.1021/ac503260f |Anal. Chem. 2015, 87, 601608608
... Zhou et al. showed the synthesis of glutathione-coated radioactive gold nanoparticles through a one-step reaction using 198 Au as the precursor and glutathione as a reducing agent [38]. Chen et al. demonstrated a method of GNP prepared using the Turkevich method followed by thermal neutron irradiation to produce 198 Au-GNP [39]. ...
... From the viewpoint of medical application, the 198 Au-GNP takes advantage of dual functions since simultaneous emission of γ photons (412 keV) and β particles (E βmax : 0.96 MeV) with a half-life of 2.69 days [41]. The γ photon scintigraphy of 198 Au-GNP in living animals using single-photon-emission-computed tomography (SPECT) has been demonstrated in previous work [39]. The β particles emitting from 198 Au-GNP can provide 0.61 MeV of maximal kinetic energy, 58% higher than those from 131 I, thereby indicating better penetration in soft tissues of locoregional radiotherapy. ...
... They then employed H 198 AuCl 4 as a precursor to synthesize RGNP using the conventional method; however, this procedure was tedious, and extensive radiation safety concerns were notified from starting materials, products, and waste. We have developed two approaches to synthesizing RGNP without using radioactive precursors ( Figure S2) [39]. The onepot/one-step reaction and the three-step reaction demonstrated the same radiochemical property along with similar physical properties despite that reaction types and surface ligands were totally different (Table S1). ...
Article
Full-text available
Nuclear fission reactions can release massive amounts of energy accompanied by neutrons and γ photons, which create a mixed radiation field and enable a series of reactions in nuclear reactors. This study demonstrates a one-pot/one-step approach to synthesizing radioactive gold nanoparticles (RGNP) without using radioactive precursors and reducing agents. Trivalent gold ions are reduced into gold nanoparticles (8.6–146 nm), and a particular portion of 197Au atoms is simultaneously converted to 198Au atoms, rendering the nanoparticles radioactive. We suggest that harnessing nuclear energy to gold nanoparticles is feasible in the interests of advancing nanotechnology for cancer therapy. A combination of RGNP applied through convection-enhanced delivery (CED) and temozolomide (TMZ) through oral administration demonstrates the synergistic effect in treating glioblastoma-bearing mice. The mean survival for RGNP/TMZ treatment was 68.9 ± 9.7 days compared to that for standalone RGNP (38.4 ± 2.2 days) or TMZ (42.8 ± 2.5 days) therapies. Based on the verification of bioluminescence images, positron emission tomography, and immunohistochemistry inspection, the combination treatment can inhibit the proliferation of glioblastoma, highlighting the niche of concurrent chemoradiotherapy (CCRT) attributed to RGNP and TMZ.
... PEGylated AuNPs were produced by linking mPEG-thiol or cPEG-thiol (averaging 5 kDa) to AuNPs with core sizes of 13 and 30 nm, respectively [20,21]. The core sizes, average hydrodynamic sizes, and ζ potentials of 13 and 30 nm mPEG-and cPEG-AuNPs are shown in Figure 2e. ...
... cPEG-AuNPs were more negatively charged than mPEG-AuNPs, as expected, but were not substantially different. Results of material characterization were generally in agreement with our previous studies [20,21]. Representative DLS results (Figure 2a,c) and TEM images (Figure 2b,d) of 13 nm mPEG-AuNPs and 30 nm cPEG-AuNP analyses are shown. ...
... cPEG-AuNPs were more negatively charged than mPEG-AuNPs, as expected, but were not substantially different. Results of material characterization were generally in agreement with our previous studies [20,21]. ...
Article
Full-text available
Polyethylene glycol (PEG) coating of gold nanoparticles (AuNPs) improves AuNP distribution via blood circulation. The use of PEG-coated AuNPs was shown to result in acute injuries to the liver, kidney, and spleen, but long-term toxicity has not been well studied. In this study, we investigated reporter induction for up to 90 days in NF-κB transgenic reporter mice following intravenous injection of PEG-coated AuNPs. The results of different doses (1 and 4 µg AuNPs per gram of body weight), particle sizes (13 nm and 30 nm), and PEG surfaces (methoxyl-or carboxymethyl-PEG 5 kDa) were compared. The data showed up to 7-fold NF-κB reporter induction in mouse liver from 3 h to 7 d post PEG-AuNP injection compared to saline-injected control mice, and gradual reduction to a level similar to control by 90 days. Agglomerates of PEG-AuNPs were detected in liver Kupffer cells, but neither gross pathological abnormality in liver sections nor increased activity of liver enzymes were found at 90 days. Injection of PEG-AuNPs led to an increase in collagen in liver sections and elevated total serum cholesterol, although still within the normal range, suggesting that inflammation resulted in mild fibrosis and affected hepatic function. Administrating PEG-AuNPs inevitably results in nanoparticles entrapped in the liver; thus, further investigation is required to fully assess the long-term impacts by PEG-AuNPs on liver health.
... Positron emission tomography (PET) imaging using 64 Cu [9] and single photon emission computed tomography (SPECT) using 99m Tc [10] were conducted for enhanced tracking of gold nanoparticles in cancer cells. Using functionalized nanomaterials, labeled with proper interfaces and 198 Au radioisotope, our group and others have already demonstrated efficient SPECT imaging of tumors in-vivo and in-vitro [11,12]. ...
... Another study by Chen et al. showed that the addition of functionalized groups to nanoparticles resulted in a significant reduction in accumulation of gold nanoparticles in liver and increasing the presence of nanomaterial in kidney. This study established a trimodality analytical platform for imaging, tracing, and quantifying gold nanoparticles in mice using 198 Au radiotracer [12]. ...
Article
Full-text available
The influence of coating a CdTe quantum dots (QDs) layer on the 198 Au nanoparticles (NPs) in biodistribution of 198 Au nanoparticles was investigated. The 198 Au nano-particles were prepared by irradiating the highly pure metallic gold in Tehran research nuclear reactor and subsequently 198 Au-NPs were synthesized and subjected to surface modification with cysteamine and CdTe QDs to form an adduct. The prepared nanomaterials were characterized with X-ray diffraction, radio thin layer chromatography, transmission electron microscopy, and scanning electron microscopy. In-vivo biodistribution and tumor avidity studies were performed by intravenously injecting of cysteamine@ 198 AuNPs: CdTe QDs nanocomposite into rats. The %ID/g (percent of the initial dose per gram tissue weight) in dissected organs and Fibro-sarcoma tumor specimens was then measured. The hydro-philicity of the cysteamine@ 198 AuNPs was increased by surface modification with CdTe QDs. Rapid excretion from body and high tumor uptake for cysteamine@ 198 AuNPs: CdTe QDs revealed that this radiotracer could potentially be used in nuclear medicine as a theranostic agent.
... Besides acting as contrast agents, by fast passivation and functionalized surfaces, very small nanoparticles, ∼nm size, producing bright fluorescent visible light are ideal for labeling [178] for multimodal imaging important for SYNAPSE. Moreover, stable 197 Au, can be transformed into 198 Au and 199 Au through neutron irradiation using a research reactor [180,181] producing β and γ -rays. The γ -rays that can be captured to reveal the emission location in an animal body with high accuracy, again a unique feature facilitating multimodal imaging. ...
Article
Full-text available
Since 2020, synchrotron radiation facilities in several Asia-Pacific countries have been collaborating in a major project called “SYNAPSE” (Synchrotrons for Neuroscience: an Asia-Pacific Scientific Enterprise). They use x-ray imaging to attack in a coordinated fashion one of the major issues in modern science: the structure of animal and human brains, including neurons and connections. The objective is to develop Google-like maps also including detailed structural and functional information for selected regions of interest. The sheer mass of data needed for the objective poses huge problems for the acquisition, processing, storage and use of images. In order to complete the task within a reasonable time, the key element of the SYNAPSE strategy is the parallel and coordinated work of several facilities on the same specimens. This article reviews different aspects of the enterprise, including the foundations of synchrotron radiation, coherence and of its role in advanced imaging, electron accelerators, x-ray optics and detectors. This will provide the foundation for an extensive presentation of the different components of SYNAPSE, with an overview of results already obtained within the consortium.
... [25][26][27] Neutron activation of AuNPs in a nuclear reactor and visualization by SPECT were attempted by Chen et al., and they visualized C6 glioma in mice. 28 Gold has only one stable isotope, 197 Au. When it captures a thermal neutron, it becomes 198 Au through 197 Auðn; cÞ 198 Au. ...
Article
Full-text available
The visualization of drugs is essential for cancer treatment. Although several methods for visualizing drugs have been proposed, a versatile method that can be easily applied to various drugs has not yet been established. Therefore, we propose “activation imaging,” in which a drug is irradiated with thermal neutrons and becomes radioactive, enabling visualization using emitted x rays and/or gamma rays. Activation imaging does not require the conjugation of specific tracers with drugs. Therefore, it can be easily applied to a variety of drugs, drug carriers (e.g., metal nanoparticles), and contrast agents. In this study, neutron activation, gamma-ray spectroscopy, and imaging of drug carriers, anticancer drug, and contrast agents were performed. Gold nanoparticles (AuNPs) and platinum nanoparticles were used as drug carriers, cisplatin was used as an anticancer drug, and gadoteridol and iohexol were used as contrast agents. As a neutron source, the RIKEN accelerator-driven compact neutron source II (RANS-II) was utilized. The imaging was performed using a hybrid Compton camera (HCC). The HCC can visualize x rays and gamma rays ranging from a few keV to nearly 1 MeV, which enables the imaging of various x rays and gamma rays emitted from the activated drugs. As a result, the gamma-ray spectra indicated the generation of radioisotopes through neutron irradiation, and AuNPs and iohexol were visualized.
... In vivo imaging of AuNP-based drug carriers is possible since the -rays may enter soft tissues without interference and are ineffective for single-photon emission computed tomography (SPECT) scanners [77]. PEGylated 198AuNP was developed by Chen and colleagues and used to visualise and track AuNPs in vivo animals and dissected tissues [79]. 198AuNPs were synthesised for Cerenkov luminescence imaging in addition to SPECT imaging [80]. ...
Article
Full-text available
Because of their unique basic features, gold nanoparticles (AuNPs) have been extensively explored and employed in the field of tumour diagnostics and therapy. There must be a thorough investigation of all aspects of AuNPs' natural features and the connections between these qualities to improve their use for tumour detection or therapy. Physical and chemical aspects of the natural features of AuNPs were discussed. The LSPR, radioactivity, and high X-ray absorption coefficient of AuNPs are among the physical features that are commonly exploited in tumour diagnosis and therapy. Unlike many other nanoparticles in chemicals, AuNPs may establish stable chemical interactions with Sand N-containing groups, which is a distinct benefit. For example, ligands or polymers with specified functions can be attached to AuNPs using this method. AuNPs' biocompatibility, targeting, and drug delivery are greatly improved as a result of these surface changes. The intrinsic connections and physicochemical features of AuNPs were comprehensively summarised in this review. The most recent advances in fundamental research and clinical studies that make use of these features will be discussed next. We also go through the challenges to be faced and possible solutions along the way from basic laboratory research to clinical application. Finally, it was determined whether or not the findings could be translated into clinical trials. Our goal is to help oncologists better leverage the remarkable physicochemical features of gold nanoparticles.
... Once within the tumor matrix, the most beneficial scenario is that the radiolabeled AuNPs diffuse and spread uniformly throughout the tumor tissue [7]. Several studies assessed the local intratumoral distribution of the radiolabeled AuNPs using nuclear imaging, microscopic examination or autoradiography.In the majority of the studies, AuNPs are usually observed in the periphery of the tumor mass close to the vasculature where they generally display heterogeneous distribution [69,70,75,82,90,102,110,117,[120][121][122]. The intratumoral diffusion of AuNPs depends on the characteristics of both the AuNPs (such as size, charge and shape) and the tumor tissue (such as the cellular density and the extracellular matrix stiffness) [123,124]. ...
Article
Full-text available
Thanks to their unique optical and physicochemical properties, gold nanoparticles have gained increased interest as radiosensitizing, photothermal therapy and optical imaging agents to enhance the effectiveness of cancer detection and therapy. Furthermore, their ability to carry multiple medically relevant radionuclides broadens their use to nuclear medicine SPECT and PET imaging as well as targeted radionuclide therapy. In this review, we discuss the radiolabeling process of gold nanoparticles and their use in (multimodal) nuclear medicine imaging to better understand their specific distribution, uptake and retention in different in vivo cancer models. In addition, radiolabeled gold nanoparticles enable image-guided therapy is reviewed as well as the enhancement of targeted radionuclide therapy and nanobrachytherapy through an increased dose deposition and radiosensitization, as demonstrated by multiple Monte Carlo studies and experimental in vitro and in vivo studies.
... Numerous publications validate the use of ex vivo γ radiotracing for biodistribution studies of drugs and their carriers. [25][26][27][28][29][30] Yet, like other methods, radiotracing may suffer from a noise signal that originates from the presence of free label. Free label, for example, radio-iodine and other radiohalogens, may arise by detachment from DDSs upon interaction with the body or degradation of biodegradable DDS components. ...
Article
Full-text available
Biodistribution studies are essential in drug carrier design and translation, and radiotracing provides a sensitive quantitation for this purpose. Yet, for biodegradable formulations, small amounts of free‐label signal may arise prior to or immediately after injection in animal models, causing potentially confounding biodistribution results. In this study, we refined a method to overcome this obstacle. First, we verified free signal generation in animal samples and then, mimicking it in a controllable setting, we injected mice intravenously with a radiolabeled drug carrier formulation (¹²⁵I‐antibody/3DNA) containing a known amount of free radiolabel (¹²⁵I), or free ¹²⁵I alone as a control. Corrected biodistribution data were obtained by separating the free radiolabel from blood and organs postmortem, using trichloroacetic acid precipitation, and subtracting the confounding signal from each tissue measurement. Control free ¹²⁵I‐radiolabel was detected at ≥85% accuracy in blood and tissues, validating the method. It biodistributed very heterogeneously among organs (0.6–39 %ID/g), indicating that any free ¹²⁵I generated in the body or present in an injected formulation cannot be simply corrected to the free‐label fraction in the original preparation, but the free label must be empirically measured in each organ. Application of this method to the biodistribution of ¹²⁵I‐antibody/3DNA, including formulations directed to endothelial target ICAM‐1, showed accurate classification of free ¹²⁵I species in blood and tissues. In addition, this technique rendered data on the in vivo degradation of the traced agents over time. Thus, this is a valuable technique to obtain accurate measurements of biodistribution using ¹²⁵I and possibly other radiotracers.
Article
The visualization of drug distribution in the body plays an essential role in cancer therapy. Although various imaging methods have been proposed, a versatile method for visualizing multiple types of drugs is yet to be established. Therefore, we propose “activation imaging” as a new method for visualizing various drugs. Activation imaging activates drugs with protons or neutrons, enabling the visualization of drugs using gamma rays and/or X-rays emitted from the activated drugs. This method does not require labeling drugs with specific tracers because the constituent elements of the drugs become radiotracers through proton/neutron irradiation. Therefore, activation imaging can be used to visualize various types of existing drugs, including anticancer drugs, drug carriers, and contrast agents. In this study, we activated drugs using low-energy neutrons and performed gamma-ray spectroscopy and imaging. We used several types of drugs, such as cisplatin, a platinum-based anticancer drug; gold and platinum nanoparticles, which are promising drug carriers; and gadoteridol, a gadolinium-based contrast agent. Imaging was performed using a hybrid Compton camera, which can visualize X-rays and gamma rays ranging from a few tens of keV to nearly 1 MeV. With this wide-band X-ray and gamma-ray imager, various X-rays and gamma rays emitted from drugs can be visualized. Moreover, high-performance liquid chromatography (HPLC) was performed for cisplatin and gadoteridol to investigate whether the molecules of the drugs were degraded by neutron irradiation. Consequently, gamma rays from the activated drugs were observed in the spectra, and all drugs were successfully visualized. Furthermore, the HPLC results showed that most gadoteridol and cisplatin molecules retained their structure through activation.
Article
Studies of radioactive isotopes at the liquid-solid or gas-solid interface are enabling a detailed mechanistic understanding the effects of radioactive decay on physical, biological, and chemical systems. In recent years, there has been a burgeoning interest in using radioactive isotopes for both imaging and therapeutic purposes by attaching them to the surface of colloidal nanoparticles. By merging the field of nanomedicine with the more mature field of internal radiation therapy, researchers are discovering new ways to diagnose and treat cancer. In this perspective, we discuss state-of-the-art radioactive thin films as applied to both well-defined surfaces and more complex nanoparticles. We highlight the design considerations that are unique to radioactive films, which originate from the damaging and potentially self-destructive emissions produced during radioactive decay and highlight future opportunities in the largely underexplored area between radioisotope chemistry and nanoscience.
Article
Functionalizing nanomaterials for diverse analytical, biomedical, and therapeutic applications requires determination of surface coverage (or density) of DNA on nanomaterials. We describe a sequential strand displacement beacon assay that is able to quantify specific DNA sequences conjugated or co-conjugated onto gold nanoparticles (AuNPs). Unlike the conventional fluorescence assay that requires the target DNA to be fluorescently labelled, the sequential strand displacement beacon method is able to quantify multiple unlabelled DNA oligonucleotides using a single (universal) strand displacement beacon. This unique feature is achieved by introducing two small unlabeled DNA probes for each specific DNA sequence and by performing sequential DNA strand displacement reactions. Varying the relative amounts of the specific DNA sequences and random DNA sequences during their co-conjugation onto AuNPs results in different densities of the specific DNA on AuNP, ranging from 90 to 230 DNA molecules per AuNP. Results obtained from our sequential strand displacement beacon assay are consistent with those obtained from the conventional fluorescence assays. However, labelling of DNA with some fluorescent dyes, e.g., tetramethylrhodamine, alters DNA density on AuNP. The strand displacement strategy overcomes this problem by obviating direct labelling of the target DNA. This method has broad potential to facilitate more efficient design and characterization of novel multifunctional materials for diverse applications.
Article
Got it PEGged: PEGylation and zwitterionization have distinct effects on the pharmacokinetic and tumor-targeting properties of luminescent gold nanoparticles (AuNPs), although both strategies lead to effective renal clearance. High tumor-targeting efficiency and specificity were obtained with PEGylated AuNPs, whereas rapid tumor detection was more readily achieved with zwitterionic AuNPs. HD=hydrodynamic diameter, GS=glutathione.
Article
A novel, high sensitive and specific DNA assay based on gold nanoparticles (AuNP) colorimetric detection and hybridization chain reaction (HCR) amplification has been demonstrated in this article. Two hairpin auxiliary probes were designed with single-stranded DNA (ssDNA) sticky ends which stabilize AuNPs and effectively prevent them from salt-induced aggregation. The target DNA hybridized with the hairpin auxiliary probes and triggered the formation of extended double-stranded DNA (dsDNA) polymers through HCR. As a result, the formed dsDNA polymers provide less stabilization without ssDNA sticky ends, and AuNPs undergo aggregation when salt concentration is increased. Subsequently, a pale purple-to-blue color variation is observed in the colloid solution. The system is simple in design and convenient in operation. The novel strategy eliminates the need of enzymatic reactions, separation processes, chemical modifications, and sophisticated instrumentations. The detection and discrimination process can be seen with the naked eye. The detection limit of this method is lower than or at least comparable to previous AuNP-based methods. Importantly, the protocol offers high selectivity for the determination between perfectly matched target oligonucleotides and targets with single base-pair mismatches.
Article
Orlistat, a selective inhibitor of gastrointestinal lipases, was used to investigate triacylglycerol absorption. Using mice and a variety of emulsified dietary lipids we found that the absorption of radiolabelled tripalmitin (containing the fatty acid 16:0), but not of triolein (18: 1n−9) or tri-γ-linolenin (18:3n−6), was incomplete from meals rich in esterified palmitate. Further, the absorption of radiolabelled tri-γ-linolenin, from both saturated and unsaturated dietary triacylglycerols, was 1·3- to 2-fold more potently inhibited by orlistat than that of triolein and tripalmitin. These radiolabelled triacylglycerols, which have the same fatty acid in all three positions, may not always be accurate markers of the absorption of dietary triacylglycerols. Orlistat was more effective at inhibiting the absorption of radiolabelled triacylglycerols with which it was codissolved than those added separately, which indicates that equilibration between lipid phases in the stomach may not always be complete. The saturation of the dietary lipid had little or no effect on the potency of orlistat. Orlistat provides a novel approach for studying the role of triacylglycerol hydrolysis in the overall process of triacylglycerol absorption.
Article
Hybrid (organic/inorganic) nanoparticles emerged as a simple solution to build "theranostic" systems. Due to their physical properties, superparamagnetic iron oxide nanoparticles (SPIONs) and plasmonic gold nanoparticles (Au-NPs) are extensively studied as a part of diagnostic and therapeutic strategies in cancer treatments. They can be used as agents for in vitro or in vivo imaging, for magnetic drug targeting and/or thermal therapy. Their functionalization with organic shells enhances their potential performance in tumor targeting and drug delivery. The advances in such hybrid nanocarriers are well illustrated with the example of the anticancer drug doxorubicin (DOX). The aim of this review is to give a multidisciplinary overview of such smart nanosystems loaded with DOX, based on examples taken from recent publications. From a physico-chemical point of view, we discuss the choices for the strategies for loading DOX and the consequences on drug release. From a biological point of view, we analyse the in vitro and in vivo assays concerning tumor imaging, targeted drug delivery and anticancer efficiency. Future opportunities and challenges are also addressed.
Article
Glutathione-coated luminescent gold nanoparticles (GS-AuNPs) with diameters of ∼2.5 nm behave like small dye molecules (IRDye 800CW) in physiological stability and renal clearance but exhibit a much longer tumor retention time and faster normal tissue clearance, indicating that the well-known enhanced permeability and retention effect, a unique strength of conventional NPs in tumor targeting, still exists in such small NPs. These merits enable the AuNPs to detect tumor more rapidly than the dye molecules without severe accumulation in reticuloendothelial system organs, making them very promising for cancer diagnosis and therapy.
Article
In this study we employed laser desorption mass spectrometry (LDI-MS) to explore the interactions between thiolated DNA (HS-DNA) and gold nanoparticles (Au NPs). Under nanosecond-pulsed laser irradiation (Nd:YAG, 355 nm), the efficiency of Au cluster ion formation from the Au NPs decreased in the presence of HS-DNA. At the optimal laser power density (2.1 × 10(4) W cm(-2)), the intensity of the Au cluster signal was sensitive to the DNA coverage and the length of the DNA strands on the Au NPs (diameter: 13 nm). Using this information, we developed a simple and specific DNA sensor that operates through analysis of the Au cluster ions formed from the fragmentation of Au NPs under LDI conditions. The coverage of the thiolated probe DNA (pDNA) on the Au NPs increased in the presence of its perfectly matched DNA (DNApm). As a result, the intensity of the signal of Au cluster ions decreased upon increasing the concentration of DNApm. Coupling these pDNA-Au NPs with LDI-MS allowed the detection of DNApm at concentrations down to the nanomolar regime. Furthermore, we applied this pDNA-Au NP probe to the detection of single-nucleotide polymorphisms (SNPs) of the Arg249Ser unit in the TP53 gene. To the best of our knowledge, this paper provides the first example of the use of LDI to analyze the coverage and structure of DNA strands on metal NPs. This simple, rapid, high-throughput detection system, based on the coupling of biofunctional Au NPs with LDI-MS, appears to hold great practicality for bioanalyses of oligonucleotides and proteins.
Article
MANY properties of colloids and suspensions depend on the particle size. Series of monodisperse suspensions of the same chemical composition but of rather different particle sizes may be used to study particle size dependent phenomena, such as Brownian motion, light scattering, sedimentation and electrophoresis of small particles. We have used such series to demonstrate the increased tendency of metal suspensions to coagulate in the presence of electrolytes as the radius of the particles increases1.
Article
A molecular nanoprobe: Glutathione-coated near-infrared-emitting radioactive gold nanoparticles have been synthezised and behave like small-molecule contrast agents in pharmocokinetics. These nanoparticles show a rapid distribution half-life, a desirable elimination half-life, and hold promise for single-photon emission computed tomography (SPECT) and fluorescence imaging.
Article
Photothermal ablation (PTA) is an emerging technique that uses near-infrared (NIR) laser light-generated heat to destroy tumor cells. However, complete tumor eradication by PTA therapy alone is difficult because heterogeneous heat distribution can lead to sublethal thermal dose in some areas of the tumor. Successful PTA therapy requires selective delivery of photothermal conducting nanoparticles to mediate effective PTA of tumor cells, and the ability to combine PTA with other therapy modalities. Here, we synthesized multifunctional doxorubicin (DOX)-loaded hollow gold nanospheres (DOX@HAuNS) that target EphB4, a member of the Eph family of receptor tyrosine kinases overexpressed on the cell membrane of multiple tumors and angiogenic blood vessels. Increased uptake of targeted nanoparticles T-DOX@HAuNS was observed in three EphB4-positive tumors both in vitro and in vivo. In vivo release of DOX from DOX@HAuNS, triggered by NIR laser, was confirmed by dual-radiotracer technique. Treatment with T-DOX@HAuNS followed by NIR laser irradiation resulted in significantly decreased tumor growth when compared with treatments with nontargeted DOX@HAuNS plus laser or HAuNS plus laser. The tumors in 6 of the 8 mice treated with T-DOX@HAuNS plus laser regressed completely with only residual scar tissue by 22 days following injection, and none of the treatment groups experienced a loss in body weight. Together, our findings show that concerted chemo-photothermal therapy with a single nanodevice capable of mediating simultaneous PTA and local drug release may have promise as a new anticancer therapy. Cancer Res; 72(18); 4777-86. ©2012 AACR.