You are on page 1of 28

Advanced Review

Therapeutic gold, silver, and


platinum nanoparticles
Miko Yamada,1 Matthew Foote2,3 and Tarl W. Prow1∗
There are an abundance of nanoparticle technologies being developed for use
as part of therapeutic strategies. This review focuses on a narrow class of metal
nanoparticles that have therapeutic potential that is a consequence of elemental
composition and size. The most widely known of these are gold nanoshells that
have been developed over the last two decades for photothermal ablation in super-
ficial cancers. The therapeutic effect is the outcome of the thickness and diameter of
the gold shell that enables fine tuning of the plasmon resonance. When these metal
nanoparticles are exposed to the relevant wavelength of light, their temperature
rapidly increases. This in turn induces a localized photothermal ablation that kills
the surrounding tumor tissue. Similarly, gold nanoparticles have been
developed to enhance radiotherapy. The high-Z nature of gold dramatically
increases the pho- toelectric cross-section. Thus, the photoelectric effects are
significantly increased. The outcome of these interactions is enhanced tumor
killing with lower doses of radiation, all while sparing tissue without gold
nanoparticles. Silver nanoparti- cles have been used for their wound healing
properties in addition to enhancing the tumor-killing effects of anticancer
drugs. Finally, platinum nanoparticles are thought to serve as a reservoir for
platinum ions that can induce DNA damage in cancer cells. The future is bright
with the path to clinical trials is largely cleared for some of the less complex
therapeutic metal nanoparticle systems. © 2014 The Authors. WIREs Nanomedicine and
How to cite this article:
WIREs Nanomed Nanobiotechnol 2015, 7:428–445. doi: 10.1002/wnan.1322

Nanobiotechnology published by Wiley Periodicals, Inc.

INTRODUCTION clinically relevant as many clinical trials focused on


he promise of nanoparticle technology lies in the gene therapy are ongoing or even completed.
T idea that the composition and physical size of
the material bestows properties that are unique from
However, the most heavily researched nanopar-
ticle field is drug delivery.5–9 There are a multitude
that observed in the other forms of the materials. of approved formulations that are commercially avail-
Nanoparticle research has come a long way toward able now and no doubt many more on the way. The
that in the last two decades. There is a burgeoning motivations for this research are improved pharma-
field of therapeutic nanoparticle gene delivery research cokinetics, targeting, and stability. The outcomes are
that appears to grow by the day.1–4 This research is some of the most sophisticated and well-
characterized nanoparticle technologies in play
∗ today.
Correspondence to: t.prow@uq.edu.au
1
While these fields are producing effective
Dermatology Research Centre, The University of Queensland,
School of Medicine, Translational Research Institute, Brisbane,
reagents and making poorly soluble drugs useful
Australia through encapsulation, controlled release, and tar-
2
Department of Radiation Oncology, Princess Alexandra Hospital, geting, the nanoparticles themselves are usually not
Brisbane, Australia therapeutic. They are mostly utilized for other prop-
3
Diamantina Institute, University of Queensland, Brisbane, erties, such as platforms into which functionality
Australia is engineered. Some exceptions are the sunscreen
Conflict of interest: The authors have declared no conflicts of interest nanoparticles ZnO and TiO2. These nanoparticles
for this article. selectively scatter harmful UV light to help prevent
DNA damage that can prematurely age the skin and
428 Volume 7, May/June 2015
© 2014 The Authors. WIREs Nanomedicine and Nanobiotechnology published by Wiley Periodicals, Inc.
This is an open access article under the terms of the Creative Commons Attribution-NonCommercial License, which permits use, distribution and reproduction
in any medium, provided the original work is properly cited and is not used for commercial purposes.
WIREs Nanomedicine and Nanobiotechnology Therapeutic metal nanoparticles

ultimately cause skin cancer. The sizes of these preven- not adversely affect the biological systems where they
tative nanoparticles are tuned to remain transparent will be used. Indeed, sophisticated targeting,
to visible light, meaning improved cosmetic signaling, feedback, and therapeutic systems have
outcomes, while efficiently scattering a range of UV been devel- oped with the unique properties of metal
light. nanoparticles as enabling technologies.
Researchers witnessed and participated in the von Maltzahn et al. described a multistep
generation of significant promises and speculation of nanoparticle system that described a nanoparticle
the impact nanoparticles would have on medicine communication system to amplify tumor targeting
that have largely been realized through and treatment.59 The study utilized gold nanorods
commercialized drug delivery applications. Along for photothermal activation of the coagulation path-
with this progress, there were some exciting way and magnetofluorescent iron oxide nanoworms
discoveries like the opti- for imaging. This complex feedback system shows
cal properties of quantum dots that are still finding that complex multicomponent systems enable new
their way into commercial applications.10–13 More paradigms for treating disease. The unique charac-
recently, up-converting nanoparticles have stolen teristics that some metal nanoparticles have can be
the lime light as they redefine what we know about used in quite sophisticated ways. Indeed, the future
fluorescence.14–16 These steps toward diagnostic and of medicine is bright and future therapeutics have
theranostic applications are the earliest on a very the potential for multicomponent activity that should
long road. For a review on the medical application vastly improve healthcare.
and impact of engineered nanoparticles, readers are
referred to Ref 17.
When examining the literature, it was evi- PHOTOTHERMAL ABLATION WITH
dent that the metal nanoparticle field contained well- GOLD NANOSHELLS
researched examples where the nanoparticles
themselves were uniquely used for therapeutics. The first report describing gold nanoshells was of a
Au2S nanoparticle core surrounded by a gold shell
These are not drug delivery devices per se, they are
the drug. This is the focus of this review. We follow published in 1994 by Zhou et al.18 The critical feature
the devel- opment and trends of therapeutic metal of these nanoparticles was the capacity to tune the
nanoparticles from their inception to the most recent plasmon resonance to longer light wavelengths with
larger diameters (Figure 1). The limitation of the
developments. A summary of the types of
Au2S core was that 50 nm was the diameter limit,
nanoparticles and their applications described in this
which meant that the tunable range was likewise
manuscript are shown in Table 1. The therapeutic
limited to approximately 520–900 nm. Simply put, the
properties of metals have been used for ages. Silver
kinetics of the nanoparticle core and shell synthesis
and copper have been known for their antimicrobial
prevented controlled growth of each. This synthesis
properties since ancient times. Gold has been used limitation was overcome by the development of a
in elixirs and colloids for medicinal properties for silica core nanoparticle coated in gold by the Halas
thousands of years. Gold nanoparticles have optical
laboratory at Rice University, published in 1998.19
properties that are not observed with larger This approach was founded on the capacity to coat
particulates. Indeed, gold nanoshells have become a relatively large silica nanoparticles (120 nm) with
heavily researched area with a focus on photothermal smaller gold nanoparticles (1–2 nm) in a controlled
cancer tissue ablation and are an excellent example of manner to produce shells with defined thicknesses
a unique nanoscale phenomena developed for a (14–30 nm).19 This was advantageous because the
therapeutic application. plasmon resonance could be finely tuned. In 2003, the
It appears that therapeutic metal nanoparti- Rice University collaborative group headed by Halas
cles, such as gold, are similar to chemotherapeutics and West published a landmark report on the use of
because even though they can be therapeutic, metal this technology for near-infrared thermal therapy of
nanoparticles can also be toxic.41,57,58 The presence tumors.20 This report described leveraging the relative
of this double-edged sword is common knowledge transparency of biological tissue in the near-infrared
with small molecules but the concept also applies to (820 nm) with the tuneable gold nanoshells that would
metal nanoparticles. The topic of gold nanoparticle absorb this light and increase in temperature by
toxicity is ongoing therefore it is critical to keep in approximately 37∘C in 5 min. The system was tested
mind that both therapeutic and toxic properties of in vitro and in a mouse tumor model (Figure 2). The
these nanoparticles need to be simultaneously nanoshells were composed of a 55-nm silica core and
studied. Thus, it is critical to keep in mind that both a 10-nm thick coating of 1–2 nm gold nanoparticles.
thera- peutic and toxic properties of these
nanoparticles need to be simultaneously studied. This
also highlights the need to design metal nanomaterials
for therapy that do
Volume 7, May/June 2015 © 2014 The Authors. WIREs Nanomedicine and Nanobiotechnology published by Wiley Periodicals, Inc. 429
TABL Summary of Therapeutic Metal Nanoparticles
E1
(in vivo)

Silica–gold nanoshells Cancer

H
u
m
a
n
b
r
e
a
st
c
a
r
ci
n
o
m
a
c
el
ls
(i
n
vi
tr
o
)
a
n
d
tr
a
n
s
m
is
si
bl
e
v
e
n
e
r
e
al
t
u
m
o
r
Biological Target
Ad
Nanoparticle Composition Site Model System Application Reference va
Gold sulfide, 50-nm nanoshell NA NA Plasmon resonance with Zhou et al.18 nc
ed
∼520–900 nm light
Re
Silica–gold nanoshells NA NA 19
wavelength light rangeresonance with broad
Plasmon Oldenburg et al.vi
e
Photothermal therapy Hirsch et al.20 2003w
Silica–gold nanoshells Breast cancer HER2+ breast carcinoma cells Photothermal therapy Loo
conjugated with anti-HER2 et al.21 2005
antibody
Superparamagnetic iron Head Head and neck Photothermal therapy Melancon et al.22 2011
oxide nanoparticles and cancer cell lines
coated with silica–gold neck overexpressing
nanoshells canc EGFR Photothermal therapy Kim et al.23 2006
Silica–gold nanoshells er
embedded with magnetic Head and neck
Fe3O4 nanoparticles,
Head cancer cell lines
conjugated with anti- and overexpressing
HER2 antibody
neck EGFR
canc
er
Gold-coated iron oxide Breast cancer HER2+ breast carcinoma cells Imaging Lim et al.24 2007
nanoparticles; silver- and photothermal
coated Fe3O4@Au th
nanoparticles, and er
hollow-type gold ap
nanoshells containing y
gold-coated iron oxide
nanoparticles
Silica–gold nanoshells and gold nanorods NA Tissue-simulating phantoms Photothermal therapy Patta
and Tunnel25 2012
Brain tumor Mouse brain metastatic l carcinoma cells
tumor xenografts
Silica–gold nanoshells Cancer In
vi
tr
o
m
o
d
e
l
u
si
n
g
h
e
a
d
a
n
d
n
e
c
k
s
q
u
a
m
o
u
s
c
el
Photothermal therapy Choi et al.26 2012 Photothermal therapy Trinidad et al.27 2014
Gold-branched shell Cancer HeLa or MDA-MB-231 Imaging and Topete et al. 28
2014
nanostructures (human cells (in vitro) and photother-
PLGA/doxorubicin-core breast MDA-MB-231- mal/chemoth
functionalized with a adenocar injected erapeutic
human serum cinoma) immunodeficient therapy
albumin/indocyanine BALB/c nude mice
green/folic acid
complex
Gold nanoparticles Cancer nd human prostate cancer cells (DU-145)

C
h
i
n
e
s
e

h
a
m
s
t
e
r

o
v
a
r
y

(
C
H
O
-
K
1
)
,

m
o
u
s
e

t
u
m
o
r

(
E
M
T
-
6
)

a
Radiosensitizer Herold et al.29 2000
43
0

©
20
14
Th
e
Aut
hor
s.
WI
REs
Na
no
me
dic
ine
an
d
Na
no
bio
tec
hn
olo
gy
pu
blis
he
d
by
Wil
ey
Per
iod
ica

Vo
lu
me wir
7, es.
M wil
a y/ ey.
Ju co
ne m/
20 nan
15 om
ed
Vo
lu TABLE 1 continued
me WI
7, Biological Target REs
Ma Na
Nanoparticle Composition Site Model System Application Reference Year
y/ no
Ju Gold nanoparticles targeted with Cancer Human colon carcinoma cells Radiosensitizer Hainfeld et al.30 1990 me
ne conjugated antibodies (LSl80) or control melanoma dic
20 cells (WM164) ine
15 an
Gold nanoparticles Breast cancer Mouse mammary EMT-6 tumors Radiosensitizer Hainfeld et al.31 2004 d
Gold nanoparticles Cancer In silico dose estimation Radiosensitizer Cho 32 2005 Na
33
no
© Gold nanoparticles Cancer In silico dose estimation Radiosensitizer Cho et al. 2009 bio
20 Gold nanoparticles Cancer Prostate cancer cells Radiosensitizer 34 2009 tec
Roa et al.
14 hn
35
Th Gold and iron oxide nanoparticles capped Cancer CT26 colon cancer cell lines Radiosensitizer Kim et al. 2010 olo
e with glutathione injected into mice gy
Aut
hor Gold nanoparticles capped with glucose Cancer Implanted breast MCF-7 Radiosensitizer Roa et al.36 2012
s. adenocarcinoma in mice
WI Gold nanoparticles capped with PEG Cancer : Mouse model of glioblastoma Radiosensitizer Joh et al.37,38 2013
REs glioblastoma multiforme
Na
no multiforme
me Gold nanoparticles Cancer Rat hepatoma model using JM-1 Radiofrequency ablation Cardinal et al.39 2008
dic cells
ine
an Silver nanoparticles Antimicrobial Human adipose-derived stem cells Toxicity evaluation Samberg et al.40 2012
d
Silver nanoparticles Antimicrobial Escherichia coli, Ag-resistant E. Antimicrobial assessment Sabella et al.41 2011
Na
no coli, Staphylococcus aureus,
bio methicillin-resistant S. aureus
tec (MRSA), and Salmonella sp.
hn
olo Silver nanoparticles Antimicrobial Skin and in vitro keratinocytes Toxicity evaluation Samberg et al.42 2010
gy Silver nanoparticles Antimicrobial A431 (human skin carcinoma) and Antimicrobial 43 2008
Arora et al.
pu
blis
HT-1080 (human fibrosarcoma)
he Silver nanoparticles Antimicrobial Zebrafish Health and environmental Asharani et al.44 2008
d impact
by
Silver nanoparticles capped with starch Antimicrobial Normal human lung fibroblast cells Toxicity evaluation Asharani et al.45 2009 Th
Wil era
ey (IMR-90) and human
pe
Per glioblastoma cells (U251) uti
iod
Silver nanoparticles-grafted dressings Wound healing Normal and diabetic mice Therapeutic evaluation Tian et al.46 2007 c
ica
47
me
Silver nanoparticles Wound healing Full-thickness excisional wound Therapeutic evaluation Liu et al. 2010 tal
mouse model
nan
TABLE 1 continued opa
43
rtic
1 les
Ad
va
nc
43 ed
2 Re
vi
e
radiation-enhanced therapy w
©
20
Silver nanoparticles capped with Brain cancer Human glioblastoma-astrocytoma epithelial-like cell Therapeutic evaluation Locatelli et al.50 2014
14 polyvinylpyrrolidone encapsulated line (U87MG) (in vitro); Swiss mice and severe
Th in polymer nanoparticles combined immunodeficiency mice bearing
e U87MG tumors (in vivo)
Aut
hor
Platinum nanoparticles Cancer Human colon carcinoma cells (HT29) Toxicity evaluation Gehrke et al.51 2011
s. 52
Platinum nanoparticles Cancer Human colon carcinoma cells (HT29) Toxicity evaluation Pelka et al. 2009
WI
REs Platinum nanoparticles Cancer In vitro Therapeutic evaluation Porcel et al.53 2010
Na Platinum nanoparticles capped with
no
Brain cancer Human lung fibroblasts (IMR-90) and human Toxicity evaluation Asharani et al.54 2010
polyvinyl alcohol glioblastoma cells (U251)
me
dic Au@Ag/Au nanoparticles Cancer A549 lung cancer cells in vitro and injected into mice Imaging and photothermal therapy Shi et al.55 2014
ine
an Ag@Au nanoparticles Cancer HeLa and HepG2 cells Imaging and photothermal therapy Yang et al.56 2012
d
Na
no
The Au2S particle. diameter). gol cha
FIGURE
bio
nanoparticles Each The 1 d| The
ractfirst (b) (a)
tec
hn consecutive absorbancreport
na erizof Absorbance
were then e spectragold
no atio
olo curve shows 30
coated with
gy increased gold of these nanoshell
parn. 0
pu PEG-5000 and particles synthesis
ticlPan
nanoparticle
blis injected directly is shown andes el
shell formation. in Panel foroptical
(a)
40
he into the tumor. 0
The highest
d (b) where mi sho 1
by The authors peak at
curve 1 ng ws 50
2
6 7
Wil described approximately
represent an the W
0
ey localized cell 530 nm is from irrelarg av
Per sa ele
death in vitro free gold gul e
iod solution ngt 60
and in vivo that nanoparticles ar Au2 h 0
15
ica of the sheS
was limited to and the (n
HAuCl4 ll cor
secondary peak m)
the treatment before str e
is from the gold
Vo area. This report nanoparti uctsurr
nanoshell 14
lu marked the cle ureoun
particle. wir
me beginning of a formation (45ded 70 13
(Reprinted with 0 12 Sa es.
7,
steep increase in and curve nmby 11 mp 10 wil
M permission from 10
2 shows 9 le nm ey.
a y/ the number of Ref 18. Copyright 80 C
Ju
just the 8 co
gold nanoshell 1994, APS 0
ne m/
20 nan
15 om
ed
WIREs Nanomedicine and Nanobiotechnology Therapeutic metal nanoparticles

off in the last few years. Since then there have been
(a) Laser only Nanoshells + Laser many different variations on this theme and many
have observed similar results in other models.
Calcein AM
In 2005, Loo et al. demonstrated the use of
antibody-targeted gold nanoshells for cancer imaging
and therapy applications. 21 They used 120-nm diame-
ter silica nanoparticles to achieve peak absorption
effi- ciencies in the near-infrared. Then either anti-
a b HER2 (breast cancer biomarker) or a nonspecific
antibody (anti-IgG) was conjugated to the gold
Fluorescein– Dextran

nanoshell sur- face. These antibody-capped gold


nanoshells were added to HER2-positive SKBR3
breast adenocarci- noma cell cultures and incubated
for 1 h. They inter- preted these experiments as a
proof-of-concept for simultaneous molecular imaging
c d of HER2 expres- sion and photothermal therapy in
(b) 35
vitro. The results showed that anti-HER2 nanoshells
35
presented signifi- cantly increased light-scattering-
30
30 based optical contrast compared with the two control
25
cell groups. Photother- mal therapy was also applied
25 20
by using near-infrared laser for 7 min (820 nm, 0.008
15
20 W/m2). The result showed that cell death was
T (°C)

10
observed only in cells treated with anti-HER2
15 nanoshells, thus illustrating specific molecular
targeting. Additional dose-response experiments with
10 6 min different incubation times suggested no cytotoxicity of
5 3 min anti-HER2 nanoshells to SKBr3 cells without near-
1 min infrared light exposure. This study showed that
immunotargeted gold nanoshells can pro-
0 0
0 1 2 m vide light scattering
in
4 5 6 contrast for imaging. At
D the same time, they can
(c) ep also exhibit sufficient
a b c d th
absorption to be
fr
o effective as a
m photothermal
sk
in
therapy.21,60
( An aqueous
m solution of
m superparamagnetic
)
iron oxide
nanoparticles coated
with silica–gold
nanoshells was
developed to aid in
magnetic resonance
imaging (MRI) during
photothermal ablation
treatment. Cell cultures
treated with these
1 cm multifunctional
aggregate particles
were irradiated with
810 nm continuously for
15 min. The temperature

Volume 7, May/June 2015 © 2014 The Authors. WIREs Nanomedicine and Nanobiotechnology published by Wiley Periodicals, Inc. 43
Advanced Review wires.wiley.com/nanomed

was, the a t ge in e S This time, SKBr3 cells magnetic Fe3O4


incrthtrea t cells temp damage imilarl were exposed to a nanoparticles provided
easeutme i were eratu (c′); and y, in femtosecond laser pulse high contrast in MRI
treat re a
d s nt o 2006, with 800 nm and a beam images and cancer cells
ed (ΔT) magneti
16 kiarea n with from Kim et of 1 mm for 10 seconds. incubated with
c
at lli.22 - gold 0 to resonanc al. They also tested various magnetic gold
concnThe d nanos 6 e used power of the laser nanoshells conjugated
entrgincr e hells mm imaging same ranging from 20 to 80 with anti-HER3 were
atioalease p and from tomogra SKBR mW. From this study, it rapidly
n l in e eithe the phy 3 showed the embedded
7.5 thtem n r skin image breast
× epera d treat to that destroyed upon short commonplace in oncology,
ed the cancer exposure to femtosecond
confirme but not in cell culture. The
10 cture e
with tumo cell
d laser pulse with an near- challenges include
partelwas n
near- r. cultur
irreversi infrared wavelength and reticuloendothelial removal,
icleslsconc t infrar Path ble e
/mLinentr . a low power.23 leakage into extravascular
ed ology thermal model In 2007, a similar fluid space, and low
F l | laser imag damage as Loo magnetic gold nanoshell nanoparticle accumulation
I d light es (d′). et al. that contained iron oxide into the tumor matrix. In
or are (Reprint
G but 2012, Pattani and Tunnel
n not show ed with nanoparticles (Fe3O4, 9–
U a in n in with 11 nm), as described conducted a com- parative
permissi
R n Panel Panel on from fabric above by Kim et al., was study of heating between
E o (a). (c), Ref 20. ated reported by Lim et al.24 gold nanoshells and gold
s Calce inclu Copyrigh magne Subsequently, they nanorods.25 They measured
h in AM ding t 2003, tic
2 synthesized gold-coated the heat generated by both
e signal a National gold magnetic nanoparticles types at the same optical
l is gross Academy
I prese phot
nanos coated with a silver density to determine the
l of
n p nt ograp hells nanolayer. These hollow photothermal transduction
Sciences
h and h (a of the
consis gold shells contain efficiency. They found that
v o fluor ′
); United ting of owing to the larger size of
magnetic nanoparti- cles
i t escei silver States of gold nanoshells, their absorption
t
in the hollow center that
o n- - America) nanos cross-section was much
r dextr
can provide supermag-
t stain hells
o netic characteristics for larger than nanorods. As a
h an is ed embe strong MRI contrast. In result, gold nanoshells
e exclu secti
r ded dded addi- tion, composite produced more heat than
a on to
n m in revea with nanoparticle-treated gold nanorods. But when
d a viabl l the magne SKBr3 cells were damaged they compared the
l e locati tic within 3 min of near- effectiveness of converting
i t cells. on of Fe3O4 light radiation into thermal
infrared laser at the rel-
n u The the nanop energy (photothermal
arro
atively low exposure of
m gold efficiency), the nanorod
ar- 12.7 W/cm at 808 nm.24
v o w in nano
ticles, However, despite shape was two times more
i r Panel shells
a (a) 7 nm these successful in vitro efficient. This study brought
v in
o b indic brow stabili mod- els of gold to light the need to consider
l ates n (b zed nanoshells, in the next not only the amount of heat
a wher ′
); with few years there was a gener- ated per particle but
e
t e hema oleic also the size of the particle
x noticeable lack of
i laser toxyli needed to target the tumor.
a acid, reports focused on the
o light n The nanorods generated less
m and efficacy of gold
n was and heat, per particle, but have
p conjug nanoshells in vivo after
. direc eosin
l ated systematic adminis- the potential to be more
A ted. -
e effective
d Panel stain with tration. This is due to
h (b) ed anti- the number of biological at targeting owing to their
o
f
e illust secti HER2 bar- riers inhibiting smaller size.25
r rates on to antibo nanoparticle circulation In the same year, Choi
g
e the show et al. reported the successful
o dy. and target- ing that are
n chan tissu in vivo model of delivering
43 © 2014 The Authors. WIREs Nanomedicine and Nanobiotechnology published by Wiley Periodicals, Inc. Vol um e 7, Ma y/June 201 5
WIREs Nanomedicine and Nanobiotechnology Therapeutic metal nanoparticles

go 26 s and in the loaded photothermal ablation


cul syst
ld Th use brain. macrophages were therapy in an in vitro
tu emi
na ey these Macrop able to cross blood– model using head and
re c
no sy cells hage brain barrier neck squamous cell
s circ
pa nt to following injection carcinoma (HNSCC)
we ulat
r- he traver into systemic cells. Common
re ion
tic siz se the circulation. They treatments for HNSCC
inc via
le ed blood could also observe include hyperthermia
s –brain ub the and photodynamic
sili macrophages
to barrie at tail therapy. However,
ca- loaded with
br cor r ed vein hyperthermia therapy
nanoparticles in
ai e witho wi . can cause unwanted
lung and liver.
n gol ut th The toxicity to normal
Choi et al.
m d damag go se tissue around tumor.
suggested that this
et na ing it. ld gol To avoid this side
indicates a cancer
as no Becau na d effect, they used 120-
treatment
ta sh se the no nan nm silica core with a
advantage where
se ell blood sh osh 12–15 nm gold shell to
lungs and liver are
s s –brain ell ell investigate the
frequent site of
of by barrie s conj combined effect of
metastatic diseases
br the r is a for uga photothermal therapy
and not a targeting
ea hy major 3 tes and photodynamic
issue. More- over,
st dr imped da wer therapy in vitro. For
this ‘Trojan Horse’
ca oly iment ys. e photothermal therapy,
delivery system
nc sis to the Th also the cells were
became a
er of delive e fluo irradiated with 𝜆 = 810
successful active
by tet ry of res resc nm for 5 min and for
gold nanoshell
us rae therap ult entl the pho- todynamic
delivery strategy,
in th eutics ing y therapy experiment, 𝜆 =
g to and this system 670 nm for 5 min. The
ylo gol labe
m brain, could enable the result from this study
rth d led
ac osi this delivery of other showed that
na so
ro lic was nanovec- tors such macrophages loaded
no that
ph ate and as nanorods, with gold nanoshells
sh the
hollow
ag .26 still is ell load could enhance the
es a nanospheres in the effect of photodynamic
Th - ed
kn eir critica sub-100 nm and therapy because of
loa mac
o ide l sub-150 nm. Since pho- tothermal
de rop
w a unmet this report, others properties of gold
d hag
n wa clinica have utilized this nanoshells.27 HNSCCs
su es
as s l need. Trojan Horse have been found to
sp coul
‘a to Choi approach to contain significant
en d
ce loa et al. deliver gold quantities of
- be
llu d used nanoshells in other macrophages.
sio trac
la ma mice experimental Therefore, these
n ked.
r cro to cancer models. tumors should be good
wa The
Tr ph establi One of the candidates for
s res
oj ag sh examples was macrophage-mediated
th ult reported by
an es huma therapy. This Trojan
en sho Trinidad et al. in
ho wit n horse concept has
inj ws 2014.27 They
rs h breast promise for
ect that investigated that
e’ na metas nanotherapeu- tics to
ed gol the combi-
sy no tatic overcome the
int d nation therapy of
st pa tumor challenge of tumor
o nan macrophages with
e rti xenog microenvi- ronment.
th osh gold nanoshell
m. cle rafts Many publications
e ell- enabled
Volume 7, May/June 2015 © 2014 The Authors. WIREs Nanomedicine and Nanobiotechnology published by Wiley Periodicals, Inc. 43
Advanced Review wires.wiley.com/nanomed

r o f r da rad m on tumor cells and would be daunting,


e n g l able used an in vivo model to despite device or drug
p j t e th pol investigate the ability of classification. Nev-
o u h ti er ylac the nanoplat- form as ertheless, the large
r g i n ap tic- fluorescence imaging. number of academic
t a s g eu co- The results from both publications containing
t c tic. glyc models are promising. supporting data for this
i e t o 28 olic In vitro, the combination strategy under- lines the
n s e u Th acid of chemotherapy and commitment of funding
v c l ey (PL hyperthermia and bodies and scientists to
e t h d de GA) reactive oxygen species this area. In searching
s h n b vel mat from the irradiation clinicaltrials.gov, there
t a o e op rix caused enhanced cell was only one trial,
i t l c ed load toxicity. In addition, in NCT01270139, that the
g o o a ed vivo model showed that terms ‘gold nanoshell
a f g m m wit the nanoplatform was OR nanoshell OR gold
t u y b ult h localized and retained in nanoparticle’ returned.
i r . i ifu the the tumor region for a This trial utilized the
o t T n nc anti long period of time.28 Trojan horse tech-
n h o e tio can One of the major nique (with allogeneic
s e p d na cer issues with therapeutic stem cells) to traffic the
r e w l dru nanoparticle gold nanoshells (60/15-
o t it na g applications is getting a and 70/40-nm silica
n e e h no dox clinically rel- evant dose core/gold nanoshell) to
x g pl oru- to the tumor. Thus, atherosclerotic plaques
o p e o atf bici there are numerous in 60 patients per
t a t l or n. reports beyond what is group. The primary
h n d m Thi possible to describe outcome was atheroma
e d a n th s herein that focus on vol- ume, or the volume
r l a at syst different targeting of the degenerative
t . n co em strategies to maximize plaque on the inner
t h h o nsi coul tumor dose, for wall of the artery. The
y e y s ste d instance. In this review, nanoshell group
p r p h d pro we have tried to cover contained the least
e a o e of vide the most relevant number of serious
s p t ll a che strategies, including adverse events
e h t bi mot PEG capping, antibody compared with stenting
o u e e od her targeting, and the control group. The gold
f t s c eg apy, Trojan horse approach. nanoshell group also
i i h These and related had the lowest
photot light
g c z n strategies are limited by atheroma volume (108
herap capa-
o e o
y, and bilitie a number of factors. mm3), where a normal
l a d l First, scale up 53-year old61 would
therm s.
d p o production and quality have a volume of 188
othera They
p t g control of commercial mm3 and the stent
py used a
n l h y quantities is difficult at group had 178 mm3.
with cell
a i a f best. Second, Overall, the trial
fluore cultur
n c t o incorporating appeared to be
scence e
o a r antibodies into the successful with the
imagi model
s t d a equation is extremely follow up showing that
ng for to test
h i r m expensive. Finally, one the Trojan Horse
diagn cytoto
e o u u can only guess that nanoshell group had a
osis x-
l n g lt working an antibody- significantly lower risk
under icity
l s i targeted, multilayered of cardiovascular death
near- of this
t m nanoparticle through than the other groups
infrar nanop
c o a o the regulatory maze tested. This study is
ed latfor
43 © 2014 The Authors. WIREs Nanomedicine and Nanobiotechnology published by Wiley Periodicals, Inc. Vol um e 7, Ma y/June 201 5
WIREs Nanomedicine and Nanobiotechnology Therapeutic metal nanoparticles

c is ferent phere r treatment. The explored was whether


the
e c dat types s that hypotheses gen- erated this enhanced
rap
r o aba of appro from these data will absorption translated to
euti
t m se. photot ximat need to be validated in improved brain tumor
c
a p In herma ed the animal models. treatment. They
met
i l any l curren delivered 15 Gy of 120
al
n e eve therap t kVp X-rays to brain
nan ENHANCED
l t nt, y. At state- tumors in a rabbit
opa
y e it is
rticl
the of-the- RADIOSENSITIZAT model. Computerized
d pos heart art ION WITH GOLD tomography scans were
es
p sibl of this with NANOPARTICLES used to estimate the
des
r o e to hypot gold treatment enhancement
pite Radiotherapy is used to
o r atta hesis nanop with iodine at 30%.
the treat cancer with great
m in is the article Similarly, the median
co success in some
i o clin goal of s used days of survival
mpl scenarios and less in
s n ical achiev for improved with iodine
ex others (for a recent
i g ly ing photot compared with the
nat maxi herma review, see Ref 63). The
n o rele controls with just
ure mum l
g i van concept of using high-Z radiotherapy and no
of absor therap
. n t materials to enhance treatment at 38.5, 25.5,
nan
T g out ption y.62 radiotherapy began in and 3 days, respectively.
opa efficie
h co They the 1980s Herold et al. made
rticl
e t me ncy. obser with iodine.64–66 This an important
e This
r h s ved research was based on intellectual step by
fabr can be
e a wit that the exploring the use of 1.5–
icat calcul
t h shallo established use of iodine 3.0 μm diameter gold
ion. ated
a w as a contrast agent. The particles to enhance
T using
r m cancer rationale for radiotherapy in a mouse
he Mie
e a therap investigating these tumor model.29 Ten
sha theory
y y agents was to improve thousand EMT-6 mouse
pe or
l would the therapeutic ratio of tumor cells were
of discre
i n be radiotherapy to effect injected in C.B17/Icr
the te
k o most lethal damage to the SCID mice. Experimental
gol dipole
e t efficie tumor target with groups were subjected
d appro
l nt sparing of adjacent to three intra-tumor
nan ximati
y b with normal tissue. For a injections of a 1% gold
opa on.
e the review on the adverse microparticle solution
rticl Kesse
t nanos effects of radiotherapy in fetal bovine serum of
e, n- tini
o r hells. on normal tissue, see 100 𝜇L each. Irradiation
i.e., and
e The Ref 67. It was was subsequently
sph Barchi
b g nanos appreciated that low- carried out in a chamber
ere, esi
e i hells energy X-ray absorption with 8 Gy of 200 kVp X-
nan calcul
s and dose was enhanced by rays, a little more than
osh ated
o t nanor iodine. The question half of the dose used
ell, the
t e ods that Iwamoto et al. by Iwamoto
or absor
h r were
nan ption et al. described above. 64 A efficiencies of the EMT-6
e e report
oro efficie number of outcomes cells derived from treated
r d ed to
d, is ncy of were examined in both tumors. Thus, direct
have
tho nanor in vitro and in vivo comparison between the
s w simila
ugh ods, models. Herold et al. Iwamoto et al. and Herold
t i r
t to nanos reported that the average et al. data is not possible.
u t absor
be hells, dose increase with the However, we speculate that
d h ption
usef and microparticles was 42– they both strongly support
i efficie
ul hollo 43%. This effect was the hypothesis that high-Z
e t ncies
for w materials can significantly
s h for quanti- fied primarily by
dif- nanos enhance radiotherapy in the
deepe in vitro plating
Volume 7, May/June 2015 © 2014 The Authors. WIREs Nanomedicine and Nanobiotechnology published by Wiley Periodicals, Inc. 43
Advanced Review wires.wiley.com/nanomed

do cro r ng fragment strong the hypoth- esis that gold


1.4 ± 0.1% of the
se pa dose. tumors, s foundatio nanoparticle-enhanced
injected dose. It must
ra rti This while targeted n on radiotherapy was a viable
be mentioned that the
ng cle helps also to which to therapeutic strategy for
nanoparticles used in
e tre suppo enhanc tumors build.31 treating can- cer. These
this study were just
us at rt the ing as This data showed dose
gold, without any
ed me idea radioth improve report enhancement and even
targeting moieties. The
as nt that erapy. d detailed ‘cured’ some animals.
This therapeu The radiation dose was targeting strat- egy
th en gold two major
com- tics in 26 or 30 Gy using 250 was to passively use
es ha is an experime
bined 1990. In kVp X-rays. Hainfeld et al. the enhanced
e nc excell nts based
with the two used atomic absorption permeability and
do ed ent on a
the decades spectrometry to quantify retention effect, but
se the candid mouse
earlier that mammary the mass of gold in the time course for
s the ate for
efforts followed, carcinoma various tissues, including injection and treatment
ar ra this
of this model. the tumor. These results were only separated by
e pe appro
Hainfel metal The revealed that 4.9 ± 0.6% 2 min. In the end, this
m uti ach.
d in nanopar tumor- of the injected dose was was a seminal report
uc c H
e immun ticle bearing present in the tumor 5 that was very exciting
h eff
r o- therapeu mice min after injection and and confirmed earlier
hi ect
o targeti tic received a that the majority of the speculation that gold
gh to
l ng gold technolo single dose was in the kidney nanoparti- cles could
er a d nanopa gy has injection at that stage. The most be effectively used to
th si e rticles evolved, of small favorable tumor-to-tissue enhance radiotherapy.
an mil t to but has (1.9 nm) ratio was 3.5 when However, the
th ar a
tumors not gold comparing tumor-to- mechanism of action
at de l. 30
form moved nanoparti muscle that had and modeling to
us gr p
o the too far cles. The predict the optimal
ed ee
i founda from doses conditions were
in as these
n tion for ranged unknown.
st Iw founding
t today’s from 1.35 In 2005, Cho et
an am e concepts al. reported phantom-
researc to 2.7 g
da ot d . based esti- mations of
h into gold
rd o o In the dose enhancement
targete nanoparti
ra et u 2004, when using gold
d gold cles per
di al., t Hainfeld
nanopa kilogram nanoparticles for
ot wit t
h rticles et al. injected enhanced
he h for publishe via the tail radiotherapy.32 They
a
ra X- enhanc d ‘The vein. Both put forth a two-part
t
py ra s ed use of tumor rationale for gold
. ys m radioth gold volume nanoparticle radio-
H of a erapy. nanopar and therapy enhancement.
er hig ll Hainfel ticles to survival First, gold has a high-Z
ol he e d et al. enhance were number (Z = 79)
d r r suggest radiothe moni- compared with the
et en g rapy’
ed tored, with previous elements eval-
o
al. er using with survival uated for dose
l
sh gy d radioac studies data enhancing with iodine
o bu nanop tive like exceeding (Z = 53) and
w t article gold Herold 1 year gadolinium (Z = 64).
ed at s hold nanopa et al.29 (Figure 3). This translates to an
th a promi rticles and The increased
at mu se in labeled Regulla resulting photoelectric cross-
go ch terms with et data section which is
ld lo of antibod al.68form strongly founded on pho-
mi we targeti y ing a supported toelectric effects, or

43 © 2014 The Authors. WIREs Nanomedicine and Nanobiotechnology published by Wiley Periodicals, Inc. Vol um e 7, Ma y/June 201 5
WIREs Nanomedicine and Nanobiotechnology Therapeutic metal nanoparticles

m r s bl . - imal cutoff of 40 was, however, a


o e c e e Sec kDa in terms of maximal dose
r l o s ele ond molecular weight enhancement of 1.3
e a r h m ly, or 10 nm in when using 192Ir 𝛾
t o a en the diameter, a rays. This exposure
s i l r t size maximal diameter level is less energetic
i o l p in of of 400 nm, weakly and likely to interact
m n a l thi the negative to neutral with the high-Z
p s r y s part surface charge, and material given that
l h y w co icle a long circulation the
y i it nt was time. Cho et al. 𝛾 emission is in the
p i h ex criti focused on only a range of 0.2–0.6 MV.
t s a t cal handful of possible This dose
h i t th for variable states enhancement
e s t o an esca using phantoms increased toward the
h m io pin irradiated with edge of the target
i b a i di g either 140 kVp X-
n a t c ne the rays, 4 or 6 MV
t s m or tum photon beams, or
e e t a ga or 192
Ir 𝛾 rays. The
r d h s do vas most effective
a a s. lin cula scenario was with
c o t T iu ture the 140 kVp X-rays
t n h m. usin on a superficial
i t e As g target loaded with
o t h r it the 30 mg gold
n h e e ha enh nanoparticles per
s e f pp anc gram tumor
p o en ed resulted in a dose
o a h r s, per enhancement
f t o e go mea factor of 5.6. This
o t , ld bilit level of gold
p m o g als y loading may not be
h i e o o and within realistic
o c l l ha rete ranges, but it does
t e d sa ntio illustrate that
o m c w lo n enhancement gains
n a t o ng phe are possible. Cho
s s r u his no et al. also reported
s i l tor me that the photon
a c d y na beam approach did
n a b of (for not result in any
d n e e be a appreciable
d f a in rece improvements in
a f m g nt dose enhancement.
t e e u no revi Given the much
o n c c n- ew, higher energy of
m e t h to see the photon beams
s r s m xic Ref this is not a
. g o in 69). surprise as they
y i r liv Dat may pass through
T . n e in a the high-Z material
h c s g sup without the
i T r u sy port photoelec- tric or
s h e it ste the Compton
e a a ms min interactions. There

Volume 7, May/June 2015 © 2014 The Authors. WIREs Nanomedicine and Nanobiotechnology published by Wiley Periodicals, Inc. 43
(a)

(b) 600
(c)
Gold only Irradiation only 100
Tumor volume (mm3)

500
90
No treatment 86%
80

Percent survival
400
70
300 60
50 50%
200 40
100 Gold + irradiation 30
20 20%
0 10
0 10 20 30 0 0%
Days 0 50 100 150 365
Days

FIGURE 3 Gold nanoparticle-enhanced radiotherapy in a mouse model. There was a clear difference in X-ray imaging of the animals before (left
| after (right panel) injecting gold nanoparticles (Panel a). After irradiation, the tumor volume shrank dramatically in the gold
panel) and 2 min
nanoparticle and radiotherapy group compared with the other controls (Panel b). This was also the case with percent survival shown in Panel (c).
(Reprinted with permission from Ref 31. Copyright 2004, IOP Publishing Ltd)

before falling off completely outside the target. Again this field. The progress of gold nanoparticle-enhanced
the dose enhancement was closely tied to the amount
of gold present, the more gold, the more the dose was
enhanced. The overall conclusion was that the
amount of gold per gram of tumor was the most
important fac- tor. They suggested that 30 mg gold
per gram of tumor would be key for clinically relevant
dose enhancement with 192Ir 𝛾 rays. The varied dose
enhancement with differing energies of X-rays and 𝛾
rays supports the hypothesis that energy of the
photon is a critical factor for achieving a clinically
relevant response.
The potential for enhanced radiotherapy with
much lower doses was explored by Cho et al. in 2009
through a Monte Carlo-based study where they sim-
ulated several low-energy approaches.33 Their sim-
ulations included 125I, 50 kVp, and 169Yb sources
(250–350 keV). The estimates showed promising dose
enhancement ratios for each source based on 7 and
18 mg gold nanoparticles per gram tumor. These ratios
were most favorable for the 169Yb source at 2.1 at the
center of the tumor for the 18 mg gold nanoparticles
per gram tumor dose. These and the authors’
previous publication supported the overall use of gold
nanopar- ticles in the context of radiotherapy
enhancement, but importantly also suggested
possible dose goals and sources for the elusive
‘clinically relevant dose’.
The term ‘clinically relevant dose’ is a critical
concern and a topic of considerable debate within
radiotherapy technology has gone through two tumor nanoparticle dose. If successful, this would
major shifts in focus. The above reports are enable lower radiotherapy doses to have higher
focused on establishing the basic feasibility of gold impact on tumor killing. The nanoparticle-targeting
nanoparticle-enhanced radiotherapy. The work was strategies reported to date are all based on attaching
conducted on both theoretical and biological fronts. targeting or cell entry moieties onto the surface of
It is clear from these relatively early reports that gold nanoparticles, which is a relatively
there is indeed the potential for gold nanoparticle- straightforward approach.
enhanced radiotherapy. In order to achieve the Hainfeld et al. had foreshadowed targeted gold
‘clinically rel- evant dose’, the variables need to be nanoparticle approaches in their 1990 manuscript
optimized. A practical way to optimize this approach entitled ‘Radioactive gold cluster immunoconjugates:
would be to systematically optimize the energy and potential agents for cancer therapy’ 30 Targeted gold
dose photon and the nanoparticle dose. The second nanoparticle approaches are now being sought as a
focus in this field is on targeting nanoparticles in means to improve the mass of gold present in tumors
hopes of achiev- ing a more favorable tumor to as was suggested by Cho et al. One approach reported
nontumor dose ratio and of course elevating the was to utilize glucose-capped gold nanoparticles by
Roa et al.34 The hypothesis was that the increased surprisingly high dose and would be expected to kill all
metabolic rate of tumors would facilitate uptake exposed cells calling into question the study design and
of these nanoparticles relative to the normal tissue. interpretation. Metabolic function, as measured by MTT
The 10-nm gold nanoparticles were capped with 6- assay, decreased in a dose-dependent manner in all
deoxy-6-fluoro-1-thio-d-glucose via a reductive nanoparticle-treated cells but was surprisingly unchanged
reaction with the thiol group and the gold nanopar- without irradiation. Data for irradiated cells without
ticle. These nanoparticles showed improved uptake nanoparticles were not presented, mak- ing interpretation
in a prostate carcinoma cell line (DU-145) com- difficult. In the animal tumor model, mice were injected
pared with a fibroblast cell line. The treated cells (15- with CT26 (colon cancer cell line) cells and tumors allowed
nm gold nanoparticles) were irradiated with a to grow to 6–11 mm. Tail vein injections of nanoparticles
137
Cs (1.176 MeV that is slightly higher energy than were done at 100 or 300 mg/kg body weight. Nanoparticle
the previously discussed experiments) source for a 2- distribution was assessed 30 min postinjection by inductively
Gy dose. Roa et al. monitored both surviv- ing cou- pled plasma. Unfortunately, a breakdown of gold and
fractions and metabolic activity with the MTT (3-(4,5-
dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide)
assay. They also evaluated cell cycle per- turbations
with flow cytometry. The results showed that the
targeted nanoparticles alone had no serious effects,
but when combined with 2 Gy irradiation there was
increased cell death and a G2/M delay attributed to
the inhibition of cyclin A. The authors suggested that
this could help to synchronize the cell cycle of
tumor cells making them an easier tar- get for
therapy. The dose enhancement factor for these in
vitro experiments appeared highest at 6 h post-
treatment at a 1.38.
Kim et al.35 reported using both targeted and
diethylenetriamine pentaacetic acid (DTPA) with
cysteine-conjugated gold nanoparticles. 70 The authors
explored the use of these and similar iron oxide
nanoparticles within in vitro and in vivo cancer mod-
els. The models were focused on survival outcomes as
previously discussed. They used 1.9 (as in Ref 31) or
13-nm gold nanoparticles with the larger ones capped
with DTPA (glutathione) for improved uptake. The
in vitro doses ranged from 0.2 to 2.0 mg/mL. Cellular
uptake was reported to range from 3.6 to 176.6 μg
gold/106 cells for both the 1.9- and 13-nm gold
nanoparticles without much difference between the
two. The cells received a 100-Gy dose, which is a
tissue was not presented. The only values given 8.4 days with radiotherapy alone to 27.2 ± 6.5 days
were of the tumor (0.28%) for the highest dose with gold nanoparticles and radiotherapy.
of gold nanoparticles which is 10 times less than There is still a long way to go to reach the levels
that observed by Hainfeld et al. using the same projected by Cho et al. at 7 and 18 mg/g tumor. Only
1.9-nm uncapped gold nanoparticles.31 These Hainfeld et al.31 have reported doses approaching
divergent results illustrate the inherent those suggested as achievable by Cho et al.32,33 at
difficulty in animal models. This high- lights
that there is little understanding of the physical
interaction of the photons, range of energies
used, and the concentration and size of the gold
nanoparticles.
Physical interactions are likely to be a
significant confounder in these biological
systems. Obviously, the biology of one tumor
model will be different from the next, but
importantly, there could be even greater
clinical differences. The other notable differ-
ence between these studies was that Hainfeld et
al. used 1.35 g/kg and Kim et al. used a
maximum of
0.3 g/kg. It is not clear why Kim et al. used a 4.5
times lower dose, but the results clearly show
that there is a far less effective response.
Together these data show two approaches, one
with a high dose of untargeted particles and
another with a far lower dose of capped
particles. It appears that the latter has an effect,
but not anywhere near the former despite
using three times more irradiation (100 Gy in
Kim et al. and 30 Gy in Hainfeld et al.).
More recently, Roa et al. showed
significant improvements in tumor uptake
with the addi- tion of PEG-6000 capping to
their glucose-coated gold nanoparticles to the
naked, citrate-stabilized nanoparticles36 (Figure
4). Noncancerous tissue uptake was 0.043 μg/g
and the maximal tumor intake was 0.198 μg/g.
Joh et al. also reported a PEG-capped gold
nanoparticle that was tested as a means to
enhance radiosensitization in a mouse model of
glioblastoma.37 The authors found an in vitro
dose enhancement of 1.3 that agrees well with
previous reports. They also found that the
radiotherapy (20 Gy, 7–14 days before
nanoparticle injection) increased brain blood
vessel permeability to gold nanoparti- cles and
proposed that this phenomenon could help
passive brain tumor targeting. The amount of
gold found in this tissue was reported to be
approximately 1.5% (increased from ∼0.4%
with radiotherapy pretreatment) of the injected
mass (0.4 g/kg) per gram of tissue. So,
approximately 12 mg would have been
injected into a 30 g mouse, resulting in
0.18 mg/g tissue. In this study, the mean mouse
sur- vival significantly increased from 18.3 ±
(a) F

O GNPs
H O PEG
SS
HO SH PEG
S
OH S
FDG S
S PEG
PEG S
O S
O S
S
H3CO N SH
H PEG
n 100 nm

PEG PEG–FDG–GNPs
(c) 6.00
12 Days
5.00

Normalized tumor volume (mm3)


(b) 100 19 Days
4.00
80
Cell survival rate (%)

3.00
60

2.00
40

1.00
20

0 0.00
Thio-6-FDG-GNPs X-ray X-ray + GNPs Control GNPs X-ray GNPs + X-ray

FIGURE 4 The synthesis and use of PEG- and glucose-coated gold nanoparticles for targeted enhancement of radiotherapy. Glucose (FDG) and
| onto the gold nanoparticles (GNPs) as schematically shown in Panel (a). These nanoparticles were shown to effectively enhance
PEG were coated
uptake and radiotherapy in vitro (Panel b) and in a mouse tumor model (Panel c). (Reprinted with permission from Ref 36. Copyright 2012, IOP
Publishing Ltd)

6.5 mg/g in the periphery of one tumor. The lack of correspondingly low irradiation dose
additional data showing this level of tumor-specific
gold can be due to many factors. It appears that
recent reports are using relatively low concentrations
of gold nanoparticles in their models and some do
not specifically measure gold content in tumors.
These studies also appear to be reporting dose
enhancement factors in the range of 1.5 despite a
variety of models, nanoparticle capping agents, and
endpoint assays. One clear trend is that the
irradiation dose range has settled in at 2–10 Gy. This
is a clinically relevant dose of radiotherapy in the
treatment of many cancers.
The statement that is clearly repeated through-
out the literature is that this approach is expected to
be most effective in superficial tumors and that the
mode of nanoparticle targeting needs to be tuned to
the disease. It appears that this approach has an
upward trajectory, but is mired in animal models
owing to a lack of physical modeling of the
interactions needed to predict more relevant
conditions. Interestingly, an early gold foil report by
Regulla et al. showed signifi- cantly higher dose
enhancement (up to 114 using mGy doses) than more
recent nanoparticle-based reports using much higher
photon doses.68 The low toxicity of gold and the
needed to examine possible enhancement lend itself nanoparticle thermal ablation in a manner similar to
to further testing in volunteers. The question is that of photothermal and radiosensitization
whether nanoparticles, microparticles, or some other therapies. Essentially, the gold nanoparticle is
modality will be optimal. Perhaps, each type of cancer hypothesized to function as a tiny resistor during
will have an optimal enhancement technology? radiofrequency exposure, heating the nanoparticle to
Regardless, vol- unteer studies are a necessity. Only induce ther- mal damage.71 For example, Cardinal et
then will we know the true potential for gold al. used a solid-state radiowave to transmit at 13.56
nanoparticle-enhanced radiotherapy. MHz into cultured HepG2 cells and a rat hepatoma
model.39 Gold nanoparticles were used to treat the
cultures and the animals at 4 nmol/L in vitro and a
RADIOFREQUENCY ABLATION WITH 0.5-mL injection of gold nanoparticles (13 nmol/L).
GOLD NANOPARTICLES There was significant thermal increases in the gold-
Radiofrequency fields can be used to induce gold treated
groups that resulted in thermal injury. The data were diameters of 14 ± 10 nm. The concentration of 1 mM in the
promising but additional targeted studies are needed presence of citrate as a stabilizer agent was used. This study
to support the further development of this strategy also showed that silver nanoparticles not only worked as
for human use. the efficient antimicrobial property but also suppressed
both local and systemic inflammation in vivo.46
Later in 2010, there was another study done by Liu
ANTIMICROBIAL SILVER et al., examining skin wound healing in vivo with silver
NANOPARTICLES nanoparticles.47 They specifically inves- tigated the
Silver nanoparticles represent the most common mechanical would healing process by focusing on two
man-made nanomaterials used in commercial medical different cell types: keratinocytes for re-epithelialization
and consumer products including household antisep- and fibroblasts for would contraction. The mean silver
tic sprays and antimicrobial bandages. For a review, nanoparticle diame- ter was 10 nm (ranging from 5 to 15
see Rizzello and Pompa.72 Silver nanoparticles have nm) and final
been used as powerful antimicrobial agents73,74 and
the toxicity of silver nanoparticles has been well
documented.40,75,42 Silver ions appear to block the
respiratory enzyme pathways and alter microbial
DNA and the cell wall, resulting in antimicrobial
effect. Silver ions from silver nanoparticles exhibit
toxicity; however, there is still no conclusive evidence
to show whether metallic nanoparticles themselves
exert the particle-specific toxicity. In fact, some
studies showed that silver nanoparticles were
generally more toxic than gold nanoparticles by
showing that cell
exposure to silver nanoparticles actually enhanced
cytotoxicity.43–45 Silver nanoparticles have been
shown to act as antifungal, antiviral, antiprotozoal,
and antiarthropod, indicating a potential treatment
and control of infectious disease.76 There is still a
large amount of research required in the field of
therapeutic application of silver nanoparticles. One
commonly described application is the use of silver
nanoparticles to improve wound healing.
In 2007, Tian et al. demonstrated that topical
delivery of silver nanoparticles promoted wound
heal- ing and reduced scar appearance in a dose-
dependent manner in mice.46 They used a murine
thermal injury model and compared silver
nanoparticles to silver sul- fadiazine, which has been
the standard treatment for burns (Figure 5). In this
study, silver nanoparticles were spherical, with
concentration of 1 mM containing sodium sub- sequently exposed to either UV light (375–410 nm
citrate was used in this experiment. They for 5–7 J/cm2) or 160 kV X-rays (0–25 Gy). The
found that sil- ver nanoparticles could results from this study showed that composite
increase the rate of wound closure by nanoparti- cles were significantly cytotoxic to the
promoting the proliferation and migra- tion of cells for both types of radiation compared with the
keratinocytes.47 Moreover, silver nanoparticles cells irradiated only with ionizing/nonionizing
appeared to drive the differentiation of radiation.49 The dose
fibroblasts into myoblasts, thereby promoting
wound contraction.
Around the same time in 2009, there was
a study shown for the first time that silver
nanopar- ticles could penetrate into human
stratum corneum and the outermost surface of
the epidermis.48 This has opened up another
possibility of using a topical application of
silver nanoparticles compared with the zinc
oxide or titanium dioxide particles, which are
often accumulated on the external surface of
the stra- tum corneum.77 The size of silver
nanoparticles ranged from 25 to 49 nm and the
stabilization was achieved by coating
nanoparticles with polyvinylpyrrolidone. The
final concentration of 0.14 wt% in ethanol was
used in this experiment. They used excised
human abdominal full thickness skin and used
an electrical conductometer at 300 Hz to
differentiate between intact and damaged skin.
The results showed that a median amount of
0.46 ng/cm2 of silver nanoparticles could
penetrate through intact human skin. On the
other hand, a median amount of 2.32 ng/cm 2 of
sil- ver nanoparticles could pass through
damaged human skin. Also smaller than 30-nm
silver nanoparticles were able to reach to the
deepest layer of the stra- tum corneum and the
outermost surface of the epi- dermis by passive
permeation.48 These results suggest that silver
nanoparticles could at least adhere to the skin
surface and give relatively long-term
therapeutic benefit.
Silver, like gold, is a high-Z element that has
potential for both enhancing photothermal
ablation or radiation-enhanced therapy. In
2013, Kleinauskas et al. reported the synthesis
of a silver nanoshell with a carbon core.49
They tested the capacity of these silver
nanoshells for photothermal ablation or
radiation-enhanced therapy. An in vitro
prostate ade- nocarcinoma cell line model was
utilized to evalu- ate the killing effects of each
therapeutic modality. Carbon-core silver-shell
nanorods were prepared in an aqueous solution
from PEGylated carbon core parti- cles (26 ± 12
nm) that had a mean diameter of 60 nm. They
tested this composite nanoparticle at a final
concentration of 1.5 mM in the cells that were
(a) (b)

Ratio compared with normal skin


IL-6 AgNP Silvadene
35
30
25
20

0
15
10
5
0

Days after injury


1 3 5 7 10 15 20 25 30
Days after injury
Ratio compared with normal skin

TGF-
35
30
25
20
15

25
10
5
0
1 3 5 7 10 15 20 25 30
Days after injury

FIGURE 5 Silver
| nanoparticle effects on wound cytokine levels and healing. (a) mRNA levels of IL-6 and TGF-𝛽 are shown over 1–30 days
postinjury. The diamond indicates silver nanoparticle treatment, the triangle shows silvadene (silver sulfadiazine) data, and the square represents no
treatment. (b) Photographs of silver nanoparticle (AgNP)- and silvadene-treated wounds. (Reprinted with permission from Ref 46. Copyright 2007,
Wiley)

enhancement ratios for the radiotherapy were in line were detectable concentrations of the radiolabel in
with the reports above with the highest dose (25 the mouse tumors, but the stability studies showed
Gy) showing a dose enhancement ratio of 1.3. The
that at 6 h only a fraction of the complete
dose enhancement of the UV exposure was greater at
nanocompos- ite remained. It is encouraging that a
approximately 15. This complex report showed that
six-component nanocomposite can be synthesized
these silver nanoshells are capable of enhancing both
and used in vivo with a positive outcome (−34 ± 12%
UV and radiotherapy, but interpretation is limited by
change in tumor size). There does appear to be a clear
the in vitro nature of the models investigated.
disconnect between the stability data with the 48 day
Recently, to aim to develop multifunctional
onset of tumor regression. There is also the need to
tumor-targeted silver nanoparticles, Locatelli et al.
under- stand the effects silver nanoparticles have
synthesized polymeric nanoparticles with smaller sil-
on cellu- lar biochemistry versus that of the silver
ver nanoparticles embedded within in addition to a
ions. It is well known that silver nanoparticles have
cytotoxic drug, alisertib.50 This composite nanoparti- been widely used as novel therapeutic agents,
cle was also coated with a chlorotoxin-derived pep- including uses in anti-inflammatory or would healing.
tide for targeting glioma cells that express matrix A very promis- ing prospect of silver nanoparticles is
metalloproteinase-2 (MMP-2). The core nanoparti- its use in cancer therapy. The potential of
cle, PLGA-block-PEG-carboxylic acid (PLGA-b-PEG)
multitargeted silver nanopar- ticles can be possible by
copolymer, was previously reported by Ding et al.78 developing variety systems.
The nanocomposite evaluated by Locatelli et al. was
smaller in size at 112 nm compared with Ding et al.,
which was ranged between 150 and 180 nm. Brain
METAL-CORE-METAL
cancer cells often overexpress MMP-2, and there-
fore, they hypothesized that peptide targeting should NANOPARTICLES
be specific in the glioma cell line model they used. Gold and silver nanoparticles are not mutually
They also injected radiolabeled composite nanoparti- exclusive therapeutics. They can also be combined
cles into immunodeficient mice bearing glioblastoma. within one nanoparticle to provide improved func-
The results from in vitro study indicated that there tionality. For example, Shi et al. recently reported the
was little to no cytotoxicity from composite nanopar- use of Au@Ag/Au nanoparticles that were investigated
ticle treatment up to 72 h incubation. In vivo, tumor for use in image-guided photothermal therapy for
size reduction only occurred after day 48.50 Also, cancer.55 They used an aptamer with a quencher as a
there fluorescent sensor probe specific for A549 tumor
cells.
Shi et al. used mice injected with A549 cells as the in associated with platinum nanoparticle treatment in
vivo model. The mice were injected with 0.1 mL con-
vitro.52 Pelka et al. observed that DNA integrity
taining 2.2 × 109 nanoparticles. The results showed was compromised and that cellular glutathione was
that the targeted photothermal therapy resulted in
decreased at 1 ng/cm2 platinum nanoparticles. Their
necrosis in the area where the tumor was injected.
data showed that there was an increase in toxicity
The outcome of this study showed that even a com-
with a decrease in nanoparticle diameter where <20-,
plex theragnostic system is possible using Au@Ag/Au
<100-, and >100-nm particles were evaluated in vitro
nanoparticles.
using a human colon carcinoma cell line (HT29).
Interestingly, Ag@Au nanoparticles can also
Finally, the authors observed that the toxic effects
be used for photothermal therapy. Yang et al.
did not appear to be due to reactive oxygen forma-
investigated the photothermal killing potential for
tion, leading to the hypothesis that platinum ions
Ag@Au@phenol formaldehyde resin nanoparticles in
could be the cause. Platinum ions from nanoparti-
vitro with HeLa and HepG2 cells.56 The cells were cles could then be used as anticancer therapies with
treated with 130 μg/mL of as-synthesized nanopar- a similar strategy to cisplatin. Asharani et al. observed
ticles for 6 h. There were significant increases in that cultured cells treated with platinum
temperature with 808-nm irradiation. Cell viability nanoparticles showed activated p53 and subsequent
fell to less than 5% in the two highest nanoparticle p21 activation that was hypothesized to be stimulated
dose groups. The Ag@Au nanoparticles have tune- by genotoxic stress.54 Porcel et al. suggested that
able optical properties that increase the potential platinum nanopar- ticles could be used for enhanced
for multifunctional applications. In summary, many
radiosensitization.53 They evaluated this hypothesis
different combinations of materials can be explored,
in vitro with promis- ing results. Together, these data
but the potential for translation to clinical appli-
present a picture of a potential cancer therapeutic.
cation decreases with increasing complexity of the
The development of a therapeutic platinum
material used. Therefore, it may be the simpler metal
nanoparticle is far earlier than that of gold, but is
nanoparticle therapies that reach the clinic first.
promising none the less.

DNA DAMAGE WITH PLATINUM


CONCLUSIONS
NANOPARTICLES
Metal nanoparticles are being used in a wide array
In 1965, Rosenberg et al. published a report on the of applications that reach far beyond therapeutics.
inhibition of Escherichia coli division, not grown, However, in the therapeutic arena it is clear that
by platinum electrolysis products. 79 Four years later, gold and silver nanomaterials are the most promising
Rosenberg et al. revealed that a square planar form of agents. This is largely owing to their unique interac-
platinum, what we now know as cisplatin, was highly tions with light and radiation. The exception is of
effective against rat sarcomas.80 The drug entered course the interesting effects of silver nanoparticles
clinical trials (1971) and was finally approved for use have in wound healing and potentially for amplify-
by the USA Food and Drug Administration in 1978. ing the effects of anticancer drugs. It can be expected
However, cisplatin is not a nanoparticle. Plat- that the next decade will reveal clinical effectiveness
inum nanoparticles have been shown to possess of the more robust but less complicated systems.
the capacity to enter cells.51 There is also evidence Overall, there is real potential for metal nanoparticles
for DNA damage and antioxidant response changes to cross the regulatory barrier into clinical use as
effective ther- apeutics.

REFERENCES 3887–3894.

1. Patnaik S, Gupta KC. Novel polyethylenimine-derived


nanoparticles for in vivo gene delivery. Expert Opin
Drug Deliv 2013, 10:215–228.

2. Ensign LM, Schneider C, Suk JS, Cone R, Hanes J.


Mucus penetrating nanoparticles: biophysical tool and
method of drug and gene delivery. Adv Mater 2012, 24:
Drug Deliv 2010, 7: 1191–1207.
4. Pissuwan D, Niidome T, Cortie MB. The forthcom-
3. Duceppe N, Tabrizian M. Advances in using ing applications of gold nanoparticles in drug and
chitosan- based nanoparticles for in vitro and gene delivery systems. J Controlled Release 2011, 149:
in vivo drug and gene delivery. Expert Opin 65–71.
5. Kiriyama A, Iga K, Shibata N. Availability of 18. Zhou HS, Honma I, Komiyama H, Haus JW. Con-
polymeric nanoparticles for specific enhanced and trolled synthesis and quantum-size effect in gold-
targeted drug delivery. Ther Deliv 2013, 4:1261– coated nanoparticles. Phys Rev B 1994, 50:12052–
1278. 12056.
6. Sridhar R, Ramakrishna S. Electrosprayed nanoparti- 19. Oldenburg SJ, Averitt RD, Westcott SL, Halas NJ.
cles for drug delivery and pharmaceutical applications. Nanoengineering of optical resonances. Chem Phys Lett
Biomatter 2013, 3:pii:e24281. doi: 10.4161/biom. 1998, 288:243–247.
24281. (Epub March 19, 2013).
7. Tosi G, Bortot B, Ruozi B, Dolcetta D, Vandelli MA,
Forni F, Severini GM. Potential use of polymeric
nanoparticles for drug delivery across the blood-brain
barrier. Curr Med Chem 2013, 20:2212–2225.
8. Cheng R, Meng F, Deng C, Klok HA, Zhong Z. Dual
and multi-stimuli responsive polymeric nanoparticles
for programmed site-specific drug delivery. Biomaterials
2013, 34:3647–3657.
9. Prow TW, Grice JE, Lin LL, Faye R, Butler M, Becker
W, Wurm EM, Yoong C, Robertson TA, Soyer HP, et
al. Nanoparticles and microparticles for skin drug
delivery. Adv Drug Deliv Rev 2011, 63:470–491.
10. Geszke-Moritz M, Moritz M. Quantum dots as versatile
probes in medical sciences: synthesis, modification and
properties. Mater Sci Eng C Mater Biol Appl 2013,
33:1008–1021.
11. Petryayeva E, Algar WR, Medintz IL. Quantum dots
in bioanalysis: a review of applications across various
platforms for fluorescence spectroscopy and imaging.
Appl Spectrosc 2013, 67:215–252.
12. Probst CE, Zrazhevskiy P, Bagalkot V, Gao X. Quan-
tum dots as a platform for nanoparticle drug delivery
vehicle design. Adv Drug Deliv Rev 2013, 65:703–
718.
13. Chang JC, Kovtun O, Blakely RD, Rosenthal SJ. Label-
ing of neuronal receptors and transporters with quan-
tum dots. WIREs Nanomed Nanobiotechnol 2012,
4:605–619.
14. Gorris HH, Wolfbeis OS. Photon-upconverting
nanoparticles for optical encoding and multiplex-
ing of cells, biomolecules, and microspheres. Angew
Chem 2013, 52:3584–3600.
15. Prow TW. Multiphoton microscopy applications in
nanodermatology. WIREs Nanomed Nanobiotechnol
2012, 4:680–690.
16. Shen J, Zhao L, Han G. Lanthanide-doped upcon-
verting luminescent nanoparticle platforms for optical
imaging-guided drug delivery and therapy. Adv Drug
Deliv Rev 2013, 65:744–755.
17. Sabella S. Impact of bionanointeractions of engineered
nanoparticles for nanomedicine. In: Monteiro-Riviere
NA, Tran CL, eds. Nanotoxicology: Progress Toward
Nanomedicine. 2nd ed. Boca Raton, Florida, USA: CRC
Press; 2014, 21–36.
20. Hirsch LR, Stafford RJ, Bankson JA, Sershen SR, breast cancer using a cellular Trojan horse. Cancer
Rivera B, Price RE, Hazle JD, Halas NJ, West JL. Nanotechnol 2012, 3:47–54.
Nanoshell-mediated near-infrared thermal therapy of 27. Trinidad AJ, Hong SJ, Peng Q, Madsen SJ, Hirschberg
tumors under magnetic resonance guidance. Proc Natl H. Combined concurrent photodynamic and gold
Acad Sci U S A 2003, 100:13549–13554. nanoshell loaded macrophage-mediated photothermal
21. Loo C, Lowery A, Halas N, West J, Drezek R. Immuno- therapies: an in vitro study on squamous cell head and
targeted nanoshells for integrated cancer imaging and neck carcinoma. Lasers Surg Med 2014, 46:310–318.
therapy. Nano Lett 2005, 5:709–711. 28. Topete A, Alatorre-Meda M, Iglesias P, Villar-Alvarez
22. Melancon MP, Lu W, Zhong M, Zhou M, Liang EM, Barbosa S, Costoya JA, Taboada P, Mosquera V.
G, Elliott AM, Hazle JD, Myers JN, Li C, Stafford Fluorescent drug-loaded, polymeric-based, branched
RJ. Targeted multifunctional gold-based nanoshells for gold nanoshells for localized multimodal therapy
magnetic resonance-guided laser ablation of head and and imaging of tumoral cells. ACS Nano 2014,
neck cancer. Biomaterials 2011, 32:7600–7608. 8:2725–2738.
23. Kim J, Park S, Lee JE, Jin SM, Lee JH, Lee IS, Yang I, 29. Herold DM, Das IJ, Stobbe CC, Iyer RV, Chapman JD.
Kim JS, Kim SK, Cho MH, et al. Designed fabrication Gold microspheres: a selective technique for producing
of multifunctional magnetic gold nanoshells and their biologically effective dose enhancement. Int J Radiat
application to magnetic resonance imaging and pho- Biol 2000, 76:1357–1364.
tothermal therapy. Angew Chem 2006, 45:7754–7758. 30. Hainfeld JF, Foley CJ, Srivastava SC, Mausner LF,
24. Lim YT, Cho MY, Kim JK, Hwangbo S, Chung BH. Feng NI, Meinken GE, Steplewski Z. Radioactive gold
Plasmonic magnetic nanostructure for bimodal imag- cluster immunoconjugates: potential agents for cancer
ing and photonic-based therapy of cancer cells. Chem- therapy. Int J Radiat Appl Instrum B Nucl Med Biol
biochem 2007, 8:2204–2209. 1990, 17: 287–294.
25. Pattani VP, Tunnell JW. Nanoparticle-mediated pho- 31. Hainfeld JF, Slatkin DN, Smilowitz HM. The use of
tothermal therapy: a comparative study of heating gold nanoparticles to enhance radiotherapy in mice.
for different particle types. Lasers Surg Med 2012, Phys Med Biol 2004, 49:N309–N315.
44:675–684. 32. Cho SH. Estimation of tumour dose enhancement due
26. Choi MR, Bardhan R, Stanton-Maxey KJ, Badve S, to gold nanoparticles during typical radiation
Nakshatri H, Stantz KM, Cao N, Halas NJ, Clare treatments:
SE. Delivery of nanoparticles to brain metastases of
a preliminary Monte Carlo study. Phys Med Biol radiation therapy of human sarcoma cells and tumors. Transl
2005, 50:N163–N173. Oncol 2013, 6:722–731.
33. Cho SH, Jones BL, Krishnan S. The dosimetric fea- 39. Cardinal J, Klune JR, Chory E, Jeyabalan G, Kanz- ius JS,
sibility of gold nanoparticle-aided radiation therapy Nalesnik M, Geller DA. Noninvasive radiofre- quency
(GNRT) via brachytherapy using low-energy 𝛾-/x-ray ablation of cancer targeted by gold nanoparti- cles. Surgery
sources. Phys Med Biol 2009, 54:4889–4905. 2008, 144:125–132.
34. Roa W, Zhang X, Guo L, Shaw A, Hu X, Xiong 40. Samberg ME, Loboa EG, Oldenburg SJ, Monteiro- Riviere
Y, Gulavita S, Patel S, Sun X, Chen J, et al. Gold NA. Silver nanoparticles do not influence stem cell
nanoparticle sensitize radiotherapy of prostate cancer differentiation but cause minimal toxicity. Nanomedicine
cells by regulation of the cell cycle. Nanotechnology 2012, 7:1197–1209.
2009, 20:375101. 41. Sabella S, Brunetti V, Vecchio G, Galeone A, Maiorano G,
35. Kim JK, Seo SJ, Kim KH, Kim TJ, Chung MH, Kim Cingolani R, Pompa PP. Toxicity of citrate-capped AuNPs:
KR, Yang TK. Therapeutic application of metallic an in vitro and in vivo assessment. J Nanopart Res 2011,
nanoparticles combined with particle-induced x-ray 13:6821–6835.
emission effect. Nanotechnology 2010, 21:425102. 42. Samberg ME, Oldenburg SJ, Monteiro-Riviere NA. Evaluation
36. Roa W, Xiong Y, Chen J, Yang X, Song K, Yang of silver nanoparticle toxicity in skin in vivo and
X, Kong B, Wilson J, Xing JZ. Pharmacokinetic and keratinocytes in vitro. Environ Health Perspect 2010,
toxicological evaluation of multi-functional thiol-6- 118:407–413.
fluoro-6-deoxy-D-glucose gold nanoparticles in vivo. 43. Arora S, Jain J, Rajwade JM, Paknikar KM. Cellular responses
Nanotechnology 2012, 23:375101. induced by silver nanoparticles: in vitro stud- ies. Toxicol Lett
37. Joh DY, Sun L, Stangl M, Al Zaki A, Murty S, San- 2008, 179:93–100.
toiemma PP, Davis JJ, Baumann BC, Alonso-Basanta 44. Asharani PV, Lian WY, Gong Z, Valiyaveettil S. Tox- icity of
M, Bhang D, et al. Selective targeting of brain tumors silver nanoparticles in zebrafish models. Nan- otechnology
with gold nanoparticle-induced radiosensitization. 2008, 19:255102.
PLoS One 2013, 8:e62425.
45. Asharani PV, Low KMG, Hande MP, Valiyaveettil S.
38. Joh DY, Kao GD, Murty S, Stangl M, Sun L, Al Cytotoxicity and genotoxicity of silver nanoparticles in human
Zaki A, Xu X, Hahn SM, Tsourkas A, Dorsey JF. cells. ACS Nano 2009, 3:279–290.
Theranostic gold nanoparticles modified for durable
systemic circulation effectively and safely enhance the
46. Tian J, Wong KK, Ho CM, Lok CN, Yu WY, 2011, 85:799–812.
Che CM, Chiu JF, Tam PK. Topical delivery of 52. Pelka J, Gehrke H, Esselen M, Turk M, Crone M,
silver nanoparticles promotes wound healing. Brase S, Muller T, Blank H, Send W, Zibat V, et al.
ChemMed- Chem 2007, 2:129–136. Cellular uptake of platinum nanoparticles in human
47. Liu X, Lee PY, Ho CM, Lui VC, Chen Y, Che colon carcinoma cells and their impact on cellular
CM, Tam PK, Wong KK. Silver nanoparticles redox systems and DNA integrity. Chem Res Toxicol
mediate differential responses in keratinocytes 2009, 22:649–659.
and fibroblasts during skin wound healing. 53. Porcel E, Liehn S, Remita H, Usami N, Kobayashi K,
ChemMedChem 2010, 5:468–475. Furusawa Y, Le Sech C, Lacombe S. Platinum nanopar-
48. Larese FF, D’Agostin F, Crosera M, Adami G, ticles: a promising material for future cancer ther-
Renzi N, Bovenzi M, Maina G. Human skin apy? Nanotechnology 2010, 21:85103. doi: 10.1088/
penetration of silver nanoparticles through 0957-4484/21/8/085103. (Epub January 26, 2010).
intact and damaged skin. Toxicology 2009, 54. Asharani PV, Xinyi N, Hande MP, Valiyaveettil S. DNA
255:33–37. damage and p53-mediated growth arrest in human
49. Kleinauskas A, Rocha S, Sahu S, Sun YP, Juzenas cells treated with platinum nanoparticles.
P. Carbon-core silver-shell nanodots as Nanomedicine (Lond) 2010, 5:51–64.
sensitizers for phototherapy and radiotherapy. 55. Shi H, Ye XS, He XX, Wang KM, Cui WS, He DG,
Nanotechnology 2013, 24:325103. Li D, Jia XK. Au@Ag/Au nanoparticles assembled
50. Locatelli E, Naddaka M, Uboldi C, Loudos G, with activatable aptamer probes as smart "nano-
Fra- gogeorgi E, Molinari V, Pucci A, Tsotakos doctors" for image-guided cancer thermotherapy.
T, Psi- madas D, Ponti J, et al. Targeted Nanoscale 2014, 6:8754–8761.
delivery of silver nanoparticles and alisertib: in 56. Yang P, Xu QZ, Jin SY, Lu Y, Zhao Y, Yu SH. Synthesis
vitro and in vivo syner- gistic effect against of multifunctional Ag@Au@phenol formaldehyde resin
glioblastoma. Nanomedicine (Lond) 2014, particles loaded with folic acids for photothermal ther-
9:839–849. apy. Chemistry 2012, 18:9294–9299.
51. Gehrke H, Pelka J, Hartinger CG, Blank H, 57. Khlebtsov N, Dykman L. Biodistribution and toxicity of
Bleimund F, Schneider R, Gerthsen D, Brase S, engineered gold nanoparticles: a review of in vitro and
Crone M, Turk M, et al. Platinum nanoparticles in vivo studies. Chem Soc Rev 2011, 40:1647–1671.
and their cellular uptake and DNA platination
58. Vecchio G, Galeone A, Brunetti V, Maiorano G, Sabella
at non-cytotoxic concentrations. Arch Toxicol
S, Cingolani R, Pompa PP. Concentration-dependent,
size-independent toxicity of citrate capped AuNPs in Radiother Oncol 1987, 8:161–170.
Drosophila melanogaster. PLoS One 2012, 7:e29980. 65. Cochran ST, Iwamoto K, Winter J, Norman A.
doi: 10.1371/journal.pone.0029980. (Epub January 4, Radiation-dose enhancement with iodine in brain-
2012). tumors. Med Phys 1986, 13:615–615.
59. von Maltzahn G, Park JH, Lin KY, Singh N, Schwoppe 66. Mello RS, Callisen H, Winter J, Kagan AR, Norman
C, Mesters R, Berdel WE, Ruoslahti E, Sailor MJ, A. Radiation-dose enhancement in tumors with iodine.
Bhatia SN. Nanoparticles that communicate in vivo to Med Phys 1983, 10:75–78.
amplify tumour targeting. Nat Mater 2011, 10:545–
67. Hubenak JR, Zhang Q, Branch CD, Kronowitz SJ.
552.
Mechanisms of injury to normal tissue after radio-
60. Liu X, Cao J, Li H, Li J, Jin Q, Ren K, Ji J. Mussel- therapy: a review. Plast Reconstr Surg 2014, 133:
inspired polydopamine: a biocompatible and 49e–56e.
ultrastable coating for nanoparticles in vivo. ACS
68. Regulla DF, Hieber LB, Seidenbusch M. Physical
Nano 2013, 7:9384–9395.
and biological interface dose effects in tissue due
61. Sipahi I, Tuzcu EM, Schoenhagen P, Wolski KE, to X-ray-induced release of secondary radiation
Nicholls SJ, Balog C, Crowe TD, Nissen SE. Effects of from metallic gold surfaces. Radiat Res 1998, 150: 92–
normal, pre-hypertensive, and hypertensive blood 100.
pres- sure levels on progression of coronary
69. Maeda H, Nakamura H, Fang J. The EPR effect
atherosclerosis. J Am Coll Cardiol 2006, 48:833–838.
for macromolecular drug delivery to solid tumors:
62. Kessentini S, Barchiesi D. Quantitative compari- improvement of tumor uptake, lowering of systemic
son of optimized nanorods, nanoshells and hollow
nanospheres for photothermal therapy. Biomed Opt
Expr 2012, 3:590–604.
63. Hawley L. Principles of radiotherapy. Br J Hosp
Med
2013, 74:C166–C169.
64. Iwamoto KS, Cochran ST, Winter J, Holburt E,
Higashida RT, Norman A. Radiation-dose enhance-
ment therapy with iodine in rabbit Vx-2 brain-tumors.
toxicity, and distinct tumor imaging in vivo. Adv Drug
Deliv Rev 2013, 65:71–79.
70. Park JA, Reddy PA, Kim HK, Kim IS, Kim GC,
Chang Y, Kim TJ. Gold nanoparticles functionalised
by Gd-complex of DTPA-bis(amide) conjugate of glu-
tathione as an MRI contrast agent. Bioorg Med Chem
Lett 2008, 18:6135–6137.
71. Glazer ES, Curley SA. Non-invasive radiofrequency
ablation of malignancies mediated by quantum dots,
gold nanoparticles and carbon nanotubes. Ther Deliv
2011, 2:1325–1330.
72. Rizzello L, Pompa PP. Nanosilver-based antibacte-
rial drugs and devices: mechanisms, methodological
drawbacks, and guidelines. Chem Soc Rev 2014, 43:
1501–1518.
73. Loomba L, Scarabelli T. Metallic nanoparticles and
their medicinal potential. Part I: gold and silver
colloids. Ther Deliv 2013, 4:859–873.
74. Mijnendonckx K, Leys N, Mahillon J, Silver S, Van
HR. Antimicrobial silver: uses, toxicity and potential for
resistance. Biometals 2013, 26:609–621.
75. Samberg ME, Orndorff PE, Monteiro-Riviere NA.
Antibacterial efficacy of silver nanoparticles of
different sizes, surface conditions and synthesis
methods. Nan- otoxicology 2011, 5:244–253.
76. Rai M, Kon K, Ingle A, Duran N, Galdiero S, Galdiero
M. Broad-spectrum bioactivities of silver nanoparticles:
the emerging trends and future prospects. Appl Micro-
biol Biotechnol 2014, 98:1951–1961.
77. Filipe P, Silva JN, Silva R, Cirne de Castro JL, Marques
GM, Alves LC, Santus R, Pinheiro T. Stratum corneum
is an effective barrier to TiO2 and ZnO nanoparticle per-
cutaneous absorption. Skin Pharmacol Physiol 2009,
22:266–275.
78. Ding H, Yong KT, Roy I, Hu R, Wu F, Zhao L,
Law WC, Zhao W, Ji W, Liu L, et al. Bioconjugated
PLGA-4-arm-PEG branched polymeric nanoparticles as
novel tumor targeting carriers. Nanotechnology 2011,
22:165101.
79. Rosenberg B, Vancamp L, Krigas T. Inhibition of cell
division in Escherichia coli by electrolysis products
from a platinum electrode. Nature 1965, 205:698–699.
80. Rosenberg B, VanCamp L, Trosko JE, Mansour VH.
Platinum compounds: a new class of potent antitumour
agents. Nature 1969, 222:385–386.

You might also like