Graf Cancer

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

Biomaterials 33 (2012) 2206e2214

Contents lists available at SciVerse ScienceDirect

Biomaterials
journal homepage: www.elsevier.com/locate/biomaterials

The influence of surface chemistry and size of nanoscale graphene oxide


on photothermal therapy of cancer using ultra-low laser power
Kai Yang a, Jianmei Wan b, Shuai Zhang a, Bo Tian a, Youjiu Zhang b, Zhuang Liu a, *
a
Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, Suzhou,
Jiangsu 215123, China
b
School of Radiation Medicine and Public Health, Soochow University, Suzhou, Jiangsu 215123, China

a r t i c l e i n f o a b s t r a c t

Article history: Photothermal therapy as a physical treatment approach to destruct cancer has emerged as an alternative
Received 5 November 2011 of currently used cancer therapies. Previously we have shown that polyethylene glycol (PEG) function-
Accepted 22 November 2011 alized nano-graphene oxide (nGO-PEG) with strong optical absorption in the near-infrared (NIR) region
Available online 12 December 2011
was a powerful photothermal agent for in vivo cancer treatment. In this work, by using ultra-small
reduced graphene oxide (nRGO) with non-covalent PEG coating, we study how sizes and surface
Keywords:
chemistry affect the in vivo behaviors of graphene, and remarkably improve the performance of
Nano-reduced graphene oxide
graphene-based in vivo photothermal cancer treatment. Owing to the enhanced NIR absorbance and
Non-covalent functionalization
Size effect
highly efficient tumor passive targeting of nRGO-PEG, excellent in vivo treatment efficacy with 100% of
Photothermal therapy tumor elimination is observed after intravenous injection of nRGO-PEG and the followed 808 nm laser
Ultra-low laser power irradiation, the power density (0.15 W/cm2, 5 min) of which is an order of magnitude lower than that
Cancer treatment usually applied for in vivo tumor ablation using many other nanomaterials. All mice after treatment
survive over a period of 100 days without a single death or any obvious sign of side effect. Our results
highlight that both surface chemistry and sizes are critical to the in vivo performance of graphene, and
show the promise of using optimized nano-graphene for ultra-effective photothermal treatment, which
may potentially be combined with other therapeutic approaches to assist our fight against cancer.
Ó 2011 Elsevier Ltd. All rights reserved.

1. Introduction achieved with an ultra-low laser power, we could avoid unneces-


sary heating of normal tissues, and importantly, may be able to
Photothermal therapy (PTT) using photo-absorbing agents to ablate larger or certain internal tumors by the phototherapy since
generate heat from optical energy has been widely developed as such a lower optical power could still be delivered to tumors even
a minimally invasive treatment methodology in comparison with after substantial light absorption and scattering by the covering
other cancer therapies such as surgery, chemotherapy and radio- tissues.
therapy. In past years, a wide range of nanomaterials with strong Graphene, a class of two-dimensional carbon nanomaterials
optical absorption in the near-infrared (NIR) tissue transparency with fascinating physical and chemical properties, has attracted
window, such as gold nanorods [1], nanoshells [2,3], nanocages tremendous interests in many different fields including biomedi-
[4,5], as well as carbon nanotubes [6e9], have been developed as cine [14e19]. A large number of groups have developed a variety of
photothermal agents for PTT treatment of cancer. However, one of graphene-based biosensing platforms for detection of biological
major limitations of using PTT for clinical cancer treatment is the molecules via different mechanisms [20e26]. We and others have
limited light penetration depth that leads to the ineffective ablation used functionalized nano-graphene as drug carriers for in vitro
of large tumors or tumors deeply located inside the body, and the intracellular delivery of anticancer chemotherapy drugs and genes
relatively high optical power (usually 2e4 W/cm2 for gold nano- [16,17,27,28]. Recently, the in vitro and in vivo toxicology of gra-
materials) [1e5,10e13] that required to obtain sufficient heating for phene oxide (GO) has also received significant attention. It is now
cancer cell killing. Thus, if effective PTT cancer destruction can be generally accepted that the in vitro cellular toxicity of graphene is
closely related to its surface functionalization, showing improved
biocompatibility for those with better coatings [29,30]. In a recent
* Corresponding author. study, we uncovered that covalently PEGylated nano-GO (nGO-
E-mail address: zliu@suda.edu.cn (Z. Liu). PEG) did not induce appreciable toxicity at our tested dose (20 mg/

0142-9612/$ e see front matter Ó 2011 Elsevier Ltd. All rights reserved.
doi:10.1016/j.biomaterials.2011.11.064
K. Yang et al. / Biomaterials 33 (2012) 2206e2214 2207

kg) to mice in a period of 3 months and could be gradually excreted from Fluka Inc.) was then added to the mixture in two equal
by both renal and fecal excretion [31]. portions to give a final concentration of 1 mg/ml totally. The reac-
Phototherapies with graphene have also shown promise in tion was allowed overnight, yielding a nGO-PEG solution which
a number of in vitro and in vivo studies [18,32e34]. In a previous was purified by filtration through 100 kDa MWCO filters
work by our group, nGO-PEG that exhibit high NIR absorbance was (Millipore).
used for effective photothermal therapy of tumors in a mouse Ultra-small nRGO-PEG was synthesized using the same proce-
model. In that work, the tumors of mice intravenously injected with dure as the preparation of RGO-PEG, except that nGO-PEG was used
nGO-PEG (2 mg/ml) were completely eliminated by the NIR laser instead of GO as the starting material.
irradiation (808 nm, 2 W/cm2) [18]. The phtotothermal effect of
125
nGO-PEG could also be used to enhance the intracellular delivery of 2.2. I labeling
photosensitizers for improved photodynamic therapy [35].
Recently, Dai and co-workers reported that ultra-small reduced nGO-PEG, RGO-PEG, and nRGO-PEG were labeled by 125I using
nano-GO (nRGO) with even higher NIR absorbance could act as a standard chloramine-T oxidation method according to our
a highly efficient targeted photothermal agent for in vitro selective previous protocol [31,39]. A mixture of 200 ml GO-derivatives
cancer cell killing [36]. Herein, by using non-covalently PEGylated (2 mg/ml), w300 mCi Na125I and 100 ml of 4 mg/mL chloramine-T
nRGO (nRGO-PEG) with the optimized surface coating and size, we (SigmaeAldrich) was reacted in a pH 7.5 phosphate buffer
further improve the graphene-based in vivo PTT cancer treatment (0.05 M) for 30 min at room temperature. The labeled GO-
in animal experiments, and achieve excellent in vivo treatment derivatives were purified by centrifugation filtration through
efficacy. The power density (0.15 W/cm2, 5 min) used in this work is Amicon filters (MWCO ¼ 100 kDa) to remove excess 125I until no
an order of magnitude lower than that usually applied for in vivo detachable gamma activity in the filtrate solution. A radiolabeling
tumor ablation using various gold nanostructures [1,2,4,5,10e13]. yield of w50% was achieved.
Our nRGO-PEG with ultra-small sizes and non-covalent PEG
coating appears to be an extremely effective agent for in vivo PTT 2.3. Tumor models
ablation of cancers.
4T1 murine breast cancer cells were cultured in RPMI-1640
2. Experimental section supplemented with 10% FBS and 1% penicillin-streptomycin. Balb/
c mice were obtained from Nanjing Peng Sheng Biological Tech-
2.1. Synthesis of reduced graphene oxide nology Co, Ltd and performed under protocols approved by Soo-
chow University Laboratory Animal Center. The 4T1 tumor models
Graphene oxide was synthesized by the modified Hummer were generated by subcutaneous injection of 2  106 cells in 50 ml
method, using flake expandable graphite as the original material PBS into the right shoulder of female BALB/c mice. The mice were
according to our previous protocol [16,18,37]. To prepare reduced used for treatment when the tumor volume reached w50 mm3.
graphene oxide, hydrazine hydrate (w1 ml) was added to the GO
solution (10 ml) at a concentration w10 mg/ml and heated in an oil 2.4. Blood circulation and biodistribution study
bath at 95  C under stirring for 24 h [41]. The resulting solution was
filtrated to remove hydrazine hydrate and washed with deionized Healthy female Balb/c mice were intravenously injected with
water, yielding an insoluble RGO black solid. 125
I-nGO-PEG, 125I-RGO-PEG, and 125I-nRGO-PEG (100 ml of 20 mCi
PEG grafted poly(maleic anhydride-alt-1-octadecene) (C18PMH- per mouse, a dose of 4 mg/kg). Blood circulation was measured by
PEG5000) was synthesized following our previously reported drawing w10 ml of blood from the tail of balb/c mice. The radio-
procedure [8,38]. In brief, 1 M equivalent (eq.) of 5 k mPEG-NH2 activity in blood was measured by gamma counter (Science and
(PEG Bio, China) were reacted with poly(maleic anhydride-alt-1- Technology Institute of China in Jia Branch Innovation Co., Ltd.). In
octadecene) (PMHC18, SigmaeAldrich) at the PEG: MHC18 mono- order to test the tumor uptake, mice bearing 4T1 tumor were
mer molar ratio of 1:1 in 2 ml dichloromethane for one day in the injected with 125I-labeled GO-derivatives at the same dose and
presence of 2 eq. of N-(3-Dimethylaminopropyl)- N0 -ethyl- sacrificed 2 days after injection. The major organs were weighed
carbodiimide hydrochloride (EDC, SigmaeAldrich) and 4 eq. trie- and collected for gamma counting.
thylamine. The reaction solution was blown-dry by nitrogen,
yielding a solid product which was then dissolved in water. The 2.5. Photothermal therapy
amphiphilic polymer solution was dialyzed against water using
a 14 kDa molecular weight cut-off (MWCO) membrane and then Female Balb/c mice bearing 4T1 tumors were injected with
lyophilized. 200ml of 2 mg/ml nRGO-PEG or nGO-PEG. For the laser treatment
To functionalize RGO, 5 ml of C18PMH-PEG5000 (2 mg/ml) was groups, the tumors of mice were irradiated by an optical fiber
mixed with 1 mg RGO and sonicated for 90min. The suspension was coupled 808 nm NIR laser (Hi-Tech Optoelectronics Co., Ltd. Beijing,
then centrifugated at 14,800 rpm for 3 h to remove any unstable China) at the power density of 0.15 W/cm2 for 5 min. The exact
aggregates. The supernatant was collected and washed through optical powers of lasers used in our experiments were calibrated
100 nm filter membrane to remove excess C18PMH-PEG. and measured by an LPE-1B laser power/energy meter (Physcience
The covalently functionalized nGO-PEG was synthesized OptoElectronics Co. Ltd. Beijing). The tumor sizes were measured by
following our established protocol [16,18,31,37]. GO aqueous a caliper every the other day and calculated as the volume ¼ (tumor
suspension (5 ml) at a concentration of w3 mg/ml was sonicated length)  (tumor width)2/2. Relative tumor volumes were calcu-
for about 30 min to give a clear solution. NaOH (0.12 g/ml) was lated as V/V0 (V0 was the tumor volume when the treatment was
added to the GO suspension and heated at 40  C for 4 h. The initiated).
resulting solution was neutralized, purified by repeated rinsing and
centrifugation. A solution of 6-arm-polyethylene glycol-amine 2.6. Histology examinations
(Sunbio Inc.) (3 mg/ml) was added to the base treated GO solu-
tion (0.5 mg/ml) and the mixture was sonicated for 5 min. N-(3- Control untreated and nRGO-PEG treated mice were sacrificed
dimethylaminopropyl-N0 -ethylcarbodiimide) hydrochloride (EDC, 100 days after treatment. Major organs of those mice were
2208 K. Yang et al. / Biomaterials 33 (2012) 2206e2214

collected, fixed in 4% formalin, conducted with paraffin embedded conjugating GO with 6-armed-PEG-Amine (SunBio Inc) via carbo-
sections, stained with hematoxylin and eosin (H&E), and examined diimide catalyzed amide formation under sonciation in an aqueous
under a digital microscope (Leica QWin). solution. Different from as-prepared GO which was in a light brown
color and rapidly aggregated in the presence of salts, nGO-PEG
3. Results and discussion showed a darker brownish color and was stable in a range of phys-
iological solutions (Fig. 1b). Strong CeH and OeH stretch peaks were
3.1. Preparation and characterization of GO-derivatives with observed in the infrared (IR) spectrum of nGO-PEG sample (Fig. 1c).
different sizes and surface chemistry To enhance the optical absorption in the NIR region, GO was
reduced into RGO by hydrazine hydrate reduction following liter-
GO was synthesized from graphite by a modified Hammers’ ature protocols [40]. RGO with much less surface functional groups
method. Three PEGylated GO-derivatives with different surface became water-insoluble (Fig. 1b, Supporting Fig. S1). We thus
coatings and sizes were prepared (Fig. 1a). Following our established functionalized RGO by sonicating it in a solution of an amphiphilic
protocol [16e18,31], nGO-PEG were synthesized by covalently polymer, PEG grafted poly(maleic anhydride-alt-1-octadecene)

Fig. 1. Preparation of PEGylated GO-derivatives. (a) A scheme showing the preparation of nGO-PEG, RGO-PEG, and nRGO-PEG from GO. (b) Photos of GO, nGO-PEG, RGO, RGO-PEG,
nRGO and nRGO-PEG solutions in water, 9% NaCl and fetal bovine serum. (c&d) FT-IR spectra of nGO-PEG, nRGO, nRGO-PEG, RGO and RGO-PEG after purification to remove excess
polymers. The strong CeH stretch peak (w2800 cm1) and CeO stretch peaks (1100e1500 cm1) clearly indicate the presence of PEG in the PEGylated samples. The absence of PEG
signals in the nRGO sample (c) suggested that the N2H4 treatment of nGO-PEG was able remove its initial covalent PEG coating.
K. Yang et al. / Biomaterials 33 (2012) 2206e2214 2209

(C18PMH-PEG) (Fig. 1a), which was previously employed to func- C18PMH-PEG, obtaining nRGO-PEG which was again stable in
tionalize single-walled carbon nanotubes (SWNTs) to effectively physiological solutions (Fig. 1b).
prolong their blood circulation half-lives [8,38]. The strong hydro- As expected [16e18,31] and shown by the atomic force micro-
phobic interactions between numerous C18 hydrocarbon chains in scope (AFM) image, nGO-PEG produced by this method exhibited
the C18PMH-PEG polymer and the hydrophobic graphene panel ultra-small sheet sizes (Fig. 2a). Compared to nGO-PEG synthesized
allows stable PEG coating of RGO, which was also evidenced by IR by covalent PEGylation, the non-covalently functionalized RGO-
spectra (Fig. 1d). The obtained RGO-PEG was a black solution stable PEG showed obviously larger sheet sizes (Fig. 2b). The third one,
in various biological buffers (Fig. 1b). nRGO-PEG, was much smaller than RGO-PEG and had similar sizes
The third PEGylated GO-derivative, nRGO-PEG, was obtained via to that of nGO-PEG (Fig. 2c). The AFM measured averaged flake
a three step method (Fig. 1a). We first synthesized nGO-PEG, and areas were 408, 3280, and 578 nm2, which were equivalent to sheet
then treated it by hydrazine reduction, which was able to eliminate diameters of 23, 65, and 27 nm (assuming them in round shape), for
the oxygen groups as well as to remove the PEGylation. The nRGO nGO-PEG, RGO-PEG, and nRGO-PEG, respectively (Fig. 2d). The
sample after reduction was not water soluble anymore (Fig. 1b). IR sheet thickness of nRGO-PEG, on the other hand, was similar to that
spectra also suggested that the PEG coating of nGO-PEG was of RGO-PEG and much higher than that of nGO-PEG (Fig. 2e), likely
completely removed after hydrazine treatment (Fig. 1c). To regain owing to the non-covalent PEGylation that offered more condensed
the water solubility, we further sonicated nRGO in a solution of surface polymer coating on graphene sheets [8].

Fig. 2. Characterization of PEGylated GO-derivatives. (aec) AFM images of nGO-PEG (a), RGO-PEG (b) and nRGO-PEG (c). (d&e) AFM histograms of flake areas (d) and heights (e) of
nGO-PEG, RGO-PEG and nRGO-PEG samples. The flake area was estimated as (length  width)  3.14/4. (f) UVeVISeNIR spectra of as-prepared GO, nGO-PEG, RGO-PEG and nRGO-
PEG solutions at the graphene-concentration of 0.01 mg/ml. (g) Temperature change curves of water, nGO-PEG, RGO-PEG, and nRGO-PEG solutions exposed to the 808 nm laser at
a power density of 1 W/cm2.
2210 K. Yang et al. / Biomaterials 33 (2012) 2206e2214

Consistent to their colors (Fig. 1b), UVeviseNIR spectral of as- second phase blood circulation half-lives were 0.29  0.02 and
prepared GO, nGO-PEG, RGO-PEG and nRGO-PEG solutions at the 5.8  2.8 h for nGO-PEG, 0.22  0.09 and 17.5  3.2 h for RGO-PEG,
same weight concentration clearly revealed that reduced RGO-PEG 0.51  0.2 and 16.7  1.7 h for nRGO-PEG. The areas under curves
and nRGO-PEG exhibited dramatically enhanced NIR absorption, (AUC0-N) of nGO-PEG, RGO-PEG and nRGO-PEG were 95  11,
with 3e4 folds of increase at 808 nm compared to that of un- 494  20 and 461  21 mg/L$h, respectively. Compared to nGO-
reduced nGO-PEG (Fig. 2f). Thus as expected, both RGO-PEG and PEG, RGO-PEG and nRGO-PEG functionalized by non-covalent
nRGO-PEG was more effective than nGO-PEG in terms of photo- polymer coating exhibit remarkably prolonged blood circulation,
thermal heating under the NIR laser irradiation (Fig. 2g). which allows repeated passes of nanomaterials through tumor
blood vasculatures and is favorable for passive tumor targeting via
3.2. In vivo blood circulation and biodistribution the enhanced permeability and retention (EPR) effect.
Next, we studied the biodistribution of the three GO-derivatives.
In order to study their in vivo behaviors, nGO-PEG, RGO-PEG, Balb/c mice bearing 4T1 tumors were sacrificed at 2 days post
nRGO-PEG were all labeled with 125I following our previous injection (p.i.) of radiolabeled 125I-nGO-PEG. 125I-RGO-PEG, and
protocol [31]. The radiolabeling method should be more reliable
125
I-nRGO-PEG (100 ml 20 mCi per mouse, a dose of 4 mg/kg). It was
than the fluorescence imaging method used in our earlier work [18] found that RGO-PEG and nRGO-PEG showed a significant increase
for quantitative and accurate in vivo tracking. The radiolabeling of tumor uptake by 7e8 times compared to that of nGO-PEG
stability of three GO-derivatives was tasted in mouse plasma at (Fig. 3b), owing to the improved pharmacokinetics of the former
37  C (Supporting Fig. S2), showing <20% of de-iodination within 7 two. However, the uptake of RGO-PEG by RES organs including liver
days. We then compared the blood circulation of three GO- and spleen was also drastically higher than that of nGO-PEG, likely
derivatives after intravenous injection of 125I-nGO-PEG, 125I-RGO- attributed to the increased sheet sizes of RGO-PEG. Remarkably,
PEG, and 125I-nRGO-PEG at the same dose (4 mg/kg, 20 mCi per nRGO-PEG with ultra-small sizes and long blood circulation
mouse) into Balb/c mice (Fig. 3a). Blood was drawn at different time exhibited rather efficient tumor passive accumulation which was
points post injection (p.i.), and measured by a gamma counter. The similar, if not higher, than that of RGO-PEG, along with the low RES
blood circulation curves showed that the pharmacokinetics of GO- retention close to that of nGO-PEG. As a critical parameter to judge
derivatives followed a two-compartment model. The first and the tumor targeting efficiency, the tumor to normal tissue (T/N)

a 40 RGO-PEG nRGO-PEG nGO-PEG


nGO-PEG
t 1/2 (h) 0.22 0.09 0.51 0.2 0.29 0.02
RGO-PEG
30 t’1/2(h) 17.5 3.2 16.7 1.7 5.8 2.8
nRGO-PEG
AUC(0- )
494 20 461 21 95 11
(mg/L·h)
%ID/g

20
c
nGO-PEG
10 RGO-PEG
nRGO-PEG
0 10
0 10 20 30 40 50
Time (h)
T/N

b 32 1
nGO-PEG
RGO-PEG
24 nRGO-PEG

0.1
%ID/g

16
Bo e
Th ach

n
e

ne
us n
te n
om g
He y

Sp ver
L rt

Li d

l
in

ai
e

M ki
In lee
St un

uc
oi
a
dn

Br
st
S
yr
Ki

0
e
te n

le
Th ch

Tu in
e
M kin
om g

Sp r
y

or
Lu t

d
ve
r

on
in
ne

In lee
St n

uc
oi

ra
ea

m
a

S
st
Li

B
id

yr

B
us
H
K

Fig. 3. Blood circulation and biodistribution of nGO-PEG, RGO-PEG and nRGO-PEG. (a) The blood circulation curve of 125I-labeled nGO-PEG, RGO-PEG and nRGO-PEG. The phar-
macokinetics of all samples followed the two-compartment model. Inset table: pharmacokinetic data of 125I-nGO-PEG, 125I-RGO-PEG and 125I-nRGO-PEG including circulation half-
lives and AUC0-N.(b) biodistribution of 125I-nGO-PEG, 125I-RGO-PEG and 125I-nRGO-PEG in female mice bearing 4T1 tumors. High tumor uptake of RGO-PEG and nRGO-PEG was
observed, owing to the prolonged blood circulation of these two GO-derivatives. On the other hand, nGO-PEG and nRGO-PEG with ultra-small sizes exhibited remarkably lower RES
retention compared to RGO-PEG. (c) Tumor to normal organ ratios (T/N ratios) of nGO-PEG, RGO-PEG and nRGO-PEG. The highest T/N ratios of nRGO-PEG were observed for most
organs, suggesting its superior passive cancer targeting efficiency compared to that of nGO-PEG and RGO-PEG. Error bars were based on standard deviations of 4 mice per group.
K. Yang et al. / Biomaterials 33 (2012) 2206e2214 2211

ratios were calculated and compared among the three GO- this work is clearly a more reliable approach to accurately assess the
derivatives (Fig. 3c). The T/N ratios of nRGO-PEG were signifi- in vivo biodistribution of nanomaterials.
cantly higher than that of nGO-PEG and RGO-PEG for most
measured organs, suggesting the superior passive tumor targeting 3.3. In vivo photothermal therapy
efficiency of nRGO-PEG with the optimized surface chemistry and
size. Based on our data, we propose that the tumor uptake of gra- Motivated by high tumor accumulation of nRGO-PEG and its
phene increases as the prolonging of its blood circulation, which is strong NIR absorbance, we then carried out an in vivo photothermal
closely associated to the surface coating of graphene. On the other therapy study. Ten mice bearing 4T1 tumors were intravenously
hand, the RES uptake of graphene may be size-dependent, with injected with 200 ml of nRGO-PEG at 2 mg/ml (a dose of 20 mg/kg)
reduced retention for those with smaller sizes. with tumors irradiated by the 808 nm laser for 5 min at an ultra-
In our previously work, we labeled nGO-PEG with a NIR fluo- low power density of 0.15 W/cm2 at 48 h p.i. (nRGO-PEG þ Laser,
rescent dye, Cy7, to study its in vivo behaviors in tumor-bearing n ¼ 10). As the comparison, another seven 4T1 tumor-bearing mice
mice [18]. Unfortunately, the RES uptake of GO-PEG was largely were injected with nGO-PEG at the same dose (20 mg/kg) and
underestimated in that study using the fluorescence imaging exposed to the laser at the same conditions (nGO-PEG þ Laser,
method, possibly due to the quenching of fluorescent dyes in liver n ¼ 7). Three other groups included saline injected mice (Control,
and spleen. The observed low RES accumulation of GO-PEG during n ¼ 7), saline injected mice exposed to laser (Laser only, n ¼ 7), and
fluorescence imaging leaded to the overestimation of its tumor nRGO-PEG injected mice without laser irradiation (nRGO-PEG,
passive targeting efficiency. The radiolabeling method employed in n ¼ 7), were also used as controls. The tumor sizes and mice

Fig. 4. In vivo photothermal therapy study using intravenously injected nRGO-PEG. (a) Tumor growth curves of different groups of mice after treatment. The tumor volumes were
normalized to their initial sizes. (b) Survival curves of mice bearing 4T1 tumors after various treatments indicated. nRGO-PEG injected mice after PTT treatment survived over 100
days without a single death. Seven mice were used for each group for ‘control’, ‘Laser only’, ‘nRGO-PEG only’, and ‘nGO-PEG þ Laser’ groups. Ten mice were used for the ‘nRGO-
PEG þ Laser’ group. (c) Representative photos of tumor-bearing mice after various treatments indicated. The laser irradiated tumors on the nRGO-PEG injected mice were
completely destructed. Laser wavelength ¼ 808 nm. Power density ¼ 0.15 W/cm2. Irradiation time ¼ 5 min. Error bars were based on standard deviations.
2212 K. Yang et al. / Biomaterials 33 (2012) 2206e2214

survival were monitored every the other day after treatment Gold nanomaterials are the mainstream photothermal agents
(Fig. 4a and b). used in PTT treatment of cancer [1e5,10e13]. In 2007, Gobin et al.
While the surface temperature of tumors on nRGO-PEG treated [41] designed gold nanoshells that possess high NIR absorbance for
mice reached to w 48  C after laser irradiation, that on nGO-PEG effective photothermal ablation of tumors at the laser power of
injected and untreated mice only showed slight increase to 41  C 4 W/cm2. Maltzahn et al used PEG coated gold nanorods (dose:
and 38  C, respectively. The tumors of mice treated with nRGO-PEG 20 mg/kg) for photothermal tumor destruction under 810 nm laser
disappeared 1 day after laser irradiation, leaving the original tumor irradiation at the power of 2 W/cm2 for 5min [1]. Effective in vivo
sites black scars, which were fully recovered about 1 week after PTT with gold nanomaterials has been achieved at lower power
treatment (Fig. 4c). The tumors of four control groups of mice densities down to 0.75 W/cm2 (dose: 10 mg/kg), however has to be
showed similarly rapid tumor growth (Fig. 4a and c), even for those combined with chemotherapy drugs [42]. In addition, one problem
injected with nGO-PEG and irradiated by the laser, indicating that of using gold nanomaterials for PTT is the instability or melting of
nGO-PEG at this optical power was not effective in photothermal gold nanostructures under intense NIR light, resulting in the
ablation of tumors. Importantly, mice bearing 4T1 tumors treated decrease of NIR absorbance after a certain period of laser irradiation
with nRGO-PEG and laser irradiation survived over 100 days [43e45]. In marked contrast to gold nanorods, whose absorbance
without tumor re-growth (Fig. 3b), in marked contrast to mice in spectrum dramatically changed when exposed to the NIR light over
the four control groups which survived for only an average of w16 time, we found that nRGO-PEG was extremely stable under
days post treatment (Fig. 4b), further demonstrating the excellent continuous NIR laser irradiation for hours, without any noticeable
efficacy of nRGO-PEG based in vivo photothermal therapy of cancer. change in its optical absorbance (Supporting Fig. S3).

Fig. 5. No obvious toxic effect was observed for nRGO-PEG treated mice. (a) Body weight curves after various treatments indicated. Neither nanomaterials injection nor laser
irradiation resulted in significant body weight drop to the treated mice. (b) H&E stained images of major organs. Mice intravenously injected with nRGO-PEG were sacrificed 1, 7,
and 100 days after treatment. No noticeable abnormality was observed in liver, spleen, kidney, and other major organs including heart, lung, stomach, and intestine (data not
shown).
K. Yang et al. / Biomaterials 33 (2012) 2206e2214 2213

Surveying literature, the lowest power density that applied to References


realize in vivo tumor ablation was reported to be 0.6 W/cm2 by Dai
et al. using SWNTs (dose: 3.6 mg/kg) [9]. Using a similar dose of [1] von Maltzahn G, Park J-H, Agrawal A, Bandaru NK, Das SK, Sailor MJ, et al.
Computationally guided photothermal tumor therapy using long-circulating
nRGO-PEG (4 mg/kg), we also achieved excellent PTT efficacy by gold nanorod antennas. Cancer Res 2009;69:3892e900.
employing 0.5 W/cm2 of NIR laser irradiation (Supporting Fig. S4). [2] Hirsch LR, Stafford RJ, Bankson JA, Sershen SR, Rivera B, Price RE, et al.
Notably, based on our own and others’ experiences, non-covalently Nanoshell-mediated near-infrared thermal therapy of tumors under
magnetic resonance guidance. Proc Natl Acad Sci USA 2003;100:
PEGylated SWNTs synthesized by that method has a concentration 13549e54.
limit and would become a sticky oil-like liquid that could hardly be [3] Tang FQ, Liu HY, Chen D, Li LL, Liu TL, Tan LF, et al. Multifunctional gold
injected into mice [46]. Therefore it may not be practical to further nanoshells on silica nanorattles: a platform for the combination of photo-
thermal therapy and chemotherapy with low systemic toxicity. Angew Chem
increase the dose and lower the laser irradiation power for SWNT- Int Ed 2011;50:891e5.
based PTT. Our nRGO-PEG is thus unique among various light- [4] Chen J, Wang D, Xi J, Au L, Siekkinen A, Warsen A, et al. Immuno gold
absorbing nanomaterials currently explored for in vivo PTT treat- nanocages with tailored optical properties for targeted photothermal
destruction of cancer cells. Nano Lett 2007;7:1318e22.
ment of cancer.
[5] Yavuz MS, Cheng Y, Chen J, Cobley CM, Zhang Q, Rycenga M, et al. Gold
nanocages covered by smart polymers for controlled release with near-
3.4. Histology examination infrared light. Nat Mater 2009;8:935e9.
[6] Moon HK, Lee SH, Choi HC. In vivo near-infrared mediated tumor destruc-
tion by photothermal effect of carbon nanotubes. ACS Nano 2009;3:
Our previous study revealed the minimal toxicity of nGO-PEG to 3707e13.
the treated animals [31]. In this work, neither death nor significant [7] Ghosh S, Dutta S, Gomes E, Carroll D, D’Agostino R, Olson J, et al.
body weight variation was noticed after nRGO-PEG treatment at Increased heating efficiency and selective thermal ablation of malignant
tissue with DNA-encased multiwalled carbon nanotubes. ACS Nano 2009;
a dose of 20 mg/kg (Fig. 5a). Major organs of nRGO-PEG treated 3:2667e73.
mice whose tumors were eliminated by the photothermal therapy [8] Liu X, Tao H, Yang K, Zhang S, Lee S-T, Liu Z. Optimization of surface chemistry
were sacrificed 100 days after the treatment. Histological assess- on single-walled carbon nanotubes for in vivo photothermal ablation of
tumors. Biomaterials 2011;32:144e51.
ment of hematoxylin and eosin (H&E) stained tissue slices showed [9] Robinson JT, Welsher K, Tabakman SM, Sherlock SP, Wang HL, Luong R, et al.
no appreciable normality or lesion in major organs of nRGO-PEG High performance in vivo near-IR (>1 mu m) imaging and photothermal
treated mice (Fig. 5b). Although the systematic long-term fate cancer therapy with carbon nanotubes. Nano Res 2010;3:779e93.
[10] O’Neal DP, Hirsch LR, Halas NJ, Payne JD, West JL. Photo-thermal tumor
and toxicology of nRGO-PEG in animals remain to be carefully ablation in mice using near infrared-absorbing nanoparticles. Cancer Lett
examined, our preliminary results indicate that nRGO-PEG may not 2004;209:171e6.
be obviously toxic to mice. [11] Dickerson EB, Dreaden EC, Huang X, El-Sayed IH, Chu H, Pushpanketh S,
et al. Gold nanorod assisted near-infrared plasmonic photothermal
therapy (PPTT) of squamous cell carcinoma in mice. Cancer Lett 2008;269:
4. Conclusions 57e66.
[12] Tong L, Wei Q, Wei A, Cheng J- X. Gold nanorods as contrast agents for bio-
logical imaging: optical properties, surface conjugation and photothermal
In summary, we develop nRGO-PEG with ultra-small sizes
effects. Photochem Photobiol 2009;85:21e32.
(average diameter ¼ 27 nm) and non-covalent PEG coating for [13] Huang XH, Jain PK, El-Sayed IH, El-Sayed MA. Plasmonic photothermal
ultra-low power in vivo PTT treatment of cancer. How the surface therapy (PPTT) using gold nanoparticles. Lasers Med Sci 2008;23:217e28.
coatings and sizes of graphene affect its in vivo behaviors is studied [14] Feng LZ, Liu ZA. Graphene in biomedicine: opportunities and challenges.
Nanomedicine 2011;6:317e24.
for the first time. It is uncovered that nRGO-PEG is able to effec- [15] Feng L, Zhang S, Liu Z. Graphene based gene transfection. Nanoscale 2011;3:
tively target the tumor via the ERP effect with relatively low RES 1252e7.
retention. Owing to the high tumor passive uptake and strong NIR [16] Liu Z, Robinson JT, Sun XM, Dai HJ. PEGylated nanographene oxide for delivery
of water-insoluble cancer drugs. J Am Chem Soc 2008;130:10876e7.
absorption, nRGO-PEG is shown to be an excellent photothermal [17] Sun X, Liu Z, Welsher K, Robinson JT, Goodwin A, Zaric S, et al. Nano-graphene
agent that enables highly efficient in vivo tumor ablation by using oxide for cellular imaging and drug delivery. Nano Res 2008;1:203e12.
an ultra-low laser power density (0.15 W/cm2), which is an order of [18] Yang K, Zhang S, Zhang G, Sun X, Lee S-T, Liu Z. Graphene in mice: ultra-
high in vivo tumor uptake and photothermal therapy. Nano Lett 2010;
magnitude lower than that usually applied for in vivo PTT treat- 10:3318e23.
ment of cancer using many other nanomaterials. The nRGO-PEG [19] Chen GY, Pang DW, Hwang SM, Tuan HY, Hu YC. A graphene-based platform
developed in this work is obviously a powerful photothermal for induced pluripotent stem cells culture and differentiation. Biomaterials
2012;33:418e27.
agent with great potential in phototherapies of cancer. The [20] He S, Song B, Li D, Zhu C, Qi W, Wen Y, et al. A Graphene Nanoprobe for rapid,
graphene-based PTT cancer treatment may be further combined sensitive, and multicolor fluorescent DNA analysis. Adv Fun Mater 2010;20:
with other therapeutic approaches co-delivered by graphene for 453e9.
[21] Tang LAL, Wang J, Loh KP. Graphene-based SELDI probe with ultrahigh
synergistic cancer destruction. Our findings also suggest that the
extraction and sensitivity for DNA oligomer. J Am Chem Soc 2010;132:
surface chemistry and sizes of nanomaterials have unassailable 10976e7.
effects to their behaviors in biological systems especially in vivo, [22] Jung JH, Cheon DS, Liu F, Lee KB, Seo TS. A graphene oxide based immuno-
and are critical to optimize their functionalities in biomedicine. biosensor for pathogen detection. Angew Chem Int Ed 2010;49:5708e11.
[23] Choi BG, Park H, Park TJ, Yang MH, Kim JS, Jang S-Y, et al. Solution chemistry of
self-assembled graphene nanohybrids for high-performance flexible biosen-
Acknowledgments sors. ACS Nano 2010;4:2910e8.
[24] He Q, Sudibya HG, Yin Z, Wu S, Li H, Boey F, et al. Centimeter-long and large-
This work was partially supported by the National Basic scale micropatterns of reduced graphene oxide films: fabrication and sensing
applications. ACS Nano 2010;4:3201e8.
Research Program (973 Program) of China (2012CB932601, [25] Wang Y, Shao Y, Matson DW, Li J, Lin Y. Nitrogen-doped graphene and its
2011CB911002), the National Natural Science Foundation of China application in electrochemical biosensing. ACS Nano 2010;4:1790e8.
(51132006, 51002100), and a Project Funded by the Priority [26] Loh KP, Bao Q, Eda G, Chhowalla M. Graphene oxide as a chemically tunable
platform for optical applications. Nat Chem 2010;2:1015e24.
Academic Program Development of Jiangsu Higher Education [27] Zhang L, Xia J, Zhao Q, Liu L, Zhang Z. Functional graphene oxide as a nano-
Institutions. carrier for controlled loading and targeted delivery of mixed anticancer drugs.
Small 2010;6:537e44.
Appendix. Supplementary data [28] Zhang L, Lu Z, Zhao Q, Huang J, Shen H, Zhang Z. Enhanced chemotherapy
efficacy by sequential delivery of siRNA and anticancer drugs using PEI-
grafted graphene oxide. Small 2011;7:460e4.
Supplementary data related to this article can be found online at [29] Hu W, Peng C, Lv M, Li X, Zhang Y, Chen N, et al. Protein corona-mediated
doi:10.1016/j.biomaterials.2011.11.064. mitigation of cytotoxicity of graphene oxide. ACS Nano 2011;5:3693e700.
2214 K. Yang et al. / Biomaterials 33 (2012) 2206e2214

[30] Chang Y, Yang S-T, Liu J-H, Dong E, Wang Y, Cao A, et al. In vitro toxicity [38] Prencipe G, Tabakman SM, Welsher K, Liu Z, Goodwin AP, Zhang L, et al. PEG
evaluation of graphene oxide on A549 cells. Toxicol Lett 2011;200: branched polymer for functionalization of nanomaterials with ultralong blood
201e10. circulation. J Am Chem Soc 2009;131:4783e7.
[31] Yang K, Wan JM, Zhang SA, Zhang YJ, Lee ST, Liu ZA. In vivo pharmacokinetics, [39] Zhang S, Yang K, Feng L, Liu Z. In vitro and in vivo behaviors of dextran
long-term biodistribution, and toxicology of PEGylated graphene in mice. ACS functionalized graphene. Carbon 2011;49:4040e9.
Nano 2011;5:516e22. [40] Stankovich S, Dikin DA, Piner RD, Kohlhaas KA, Kleinhammes A, Jia Y, et al.
[32] Liu Z, Robinson JT, Tabakman SM, Yang K, Dai H. Carbon materials for drug Synthesis of graphene-based nanosheets via chemical reduction of exfoliated
delivery & cancer therapy. Mater Today 2011;14:316e23. graphite oxide. Carbon 2007;45:1558e65.
[33] Markovic ZM, Harhaji-Trajkovic LM, Todorovic-Markovic BM, Kepic DP, [41] Gobin AM, Lee MH, Halas NJ, James WD, Drezek RA, West JL. Near-infrared
Arsikin KM, Jovanovic SP, et al. In vitro comparison of the photothermal resonant nanoshells for combined optical imaging and photothermal cancer
anticancer activity of graphene nanoparticles and carbon nanotubes. Bioma- therapy. Nano Lett 2007;7:1929e34.
terials 2011;32:1121e9. [42] Sailor MJ, Park JH, von Maltzahn G, Xu MJ, Fogal V, Kotamraju VR, et al.
[34] Zhang W, Guo Z, Huang D, Liu Z, Guo X, Zhong H. Synergistic effect of chemo- Cooperative nanomaterial system to sensitize, target, and treat tumors. Proc
photothermal therapy using PEGylated graphene oxide. Biomaterials 2011; Natl Acad Sci USA 2010;107:981e6.
32:8555e61. [43] Link S, Burda C, Mohamed MB, Nikoobakht B, El-Sayed MA. Laser photo-
[35] Tian B, Wang C, Zhang S, Feng L, Liu Z. Photothermally enhanced photody- thermal melting and fragmentation of gold nanorods: energy and laser pulse-
namic therapy delivered by nano-graphene oxide. ACS Nano; 2011 [Article width dependence. J Phys Chem A 1999;103:1165e70.
ASAP]. [44] Chen JY, Wiley B, Li ZY, Campbell D, Saeki F, Cang H, et al. Gold nanocages:
[36] Robinson JT, Tabakman SM, Liang YY, Wang HL, Casalongue HS, Vinh D, et al. Engineering their structure for biomedical applications. Adv Mater 2005;17:
Ultrasmall reduced graphene oxide with high near-infrared absorbance for 2255e61.
photothermal therapy. J Am Chem Soc 2011;133:6825e31. [45] Link S, Wang ZL, El-Sayed MA. How does a gold nanorod melt? J Phys Chem B
[37] Sun X, Zaric S, Daranciang D, Welsher K, Lu Y, Li X, et al. Optical properties of 2000;104:7867e70.
ultrashort semiconducting single-walled carbon nanotube capsules down to [46] Liu Z, Tabakman SM, Chen Z, Dai H. Preparation of carbon nanotube bio-
sub-10 nm. J Am Chem Soc 2008;130:6551e5. conjugates for biomedical applications. Nat Protoc 2009;4:1372e82.

You might also like