ArticlePDF Available

A 3D graphene oxide microchip and a Au-enwrapped silica nanocomposite-based supersandwich cytosensor toward capture and analysis of circulating tumor cells

Authors:
  • Jianghan University, Wuhan, China

Abstract and Figures

Determination of the presence and number of circulating tumor cells (CTCs) in peripheral blood can provide clinically important data for prognosis and therapeutic response patterns. In this study, a versatile supersandwich cytosensor was successfully developed for the highly sensitive and selective analysis of CTCs using Au-enwrapped silica nanocomposites (Si/AuNPs) and three-dimensional (3D) microchips. First, 3D microchips were fabricated by a photolithography method. Then, the prepared substrate was applied to bind graphene oxide, streptavidin and biotinylated epithelial-cell adhesion-molecule antibody, resulting in high stability, bioactivity, and capability for CTCs capture. Furthermore, horseradish peroxidase and anti-CA153 were co-linked to the Si/AuNPs for signal amplification. The performance of the cytosensor was evaluated with MCF7 breast cancer cells. Under optimal conditions, the proposed supersandwich cytosensor showed high sensitivity with a wide range of 10(1) to 10(7) cells per mL and a detection limit of 10 cells per mL. More importantly, it could effectively distinguish CTCs from normal cells, which indicated the promising applications of our method for the clinical diagnosis and therapeutic monitoring of cancers.
Content may be subject to copyright.
Nanoscale
PAPER
Cite this: Nanoscale, 2015, 7, 16354
Received 23rd July 2015,
Accepted 25th August 2015
DOI: 10.1039/c5nr04798f
www.rsc.org/nanoscale
A 3D graphene oxide microchip and a
Au-enwrapped silica nanocomposite-based
supersandwich cytosensor toward capture and
analysis of circulating tumor cells
Na Li,
a,b
Tingyu Xiao,
a,b
Zhengtao Zhang,
a
Rongxiang He,
a,b
Dan Wen,
a
Yiping Cao,*
a,b
Weiying Zhang*
a,b
and Yong Chen
a,b,c
Determination of the presence and number of circulating tumor cells (CTCs) in peripheral blood can
provide clinically important data for prognosis and therapeutic response patterns. In this study, a versatile
supersandwich cytosensor was successfully developed for the highly sensitive and selective analysis of
CTCs using Au-enwrapped silica nanocomposites (Si/AuNPs) and three-dimensional (3D) microchips.
First, 3D microchips were fabricated by a photolithography method. Then, the prepared substrate was
applied to bind graphene oxide, streptavidin and biotinylated epithelial-cell adhesion-molecule antibody,
resulting in high stability, bioactivity, and capability for CTCs capture. Furthermore, horseradish peroxidase
and anti-CA153 were co-linked to the Si/AuNPs for signal amplication. The performance of the cyto-
sensor was evaluated with MCF7 breast cancer cells. Under optimal conditions, the proposed super-
sandwich cytosensor showed high sensitivity with a wide range of 10
1
to 10
7
cells per mL and a detection
limit of 10 cells per mL. More importantly, it could eectively distinguish CTCs from normal cells, which
indicated the promising applications of our method for the clinical diagnosis and therapeutic monitoring
of cancers.
Introduction
Cancer is considered a worldwide mortal sickness and has
become a major public concern. Metastasis is the most
common cause of cancer-related deaths in patients with solid
tumors. During the progression of metastasis, tumor cells
detach from the solid primary tumor, enter the blood stream
and travel to dierent tissues of the body.
1
These break-away
tumor cells in the peripheral blood are known as circulating
tumor cells (CTCs). In addition to conventional diagnostic
imaging and serum marker detection, CTCs analysis as a
liquid biopsycan provide valuable information on prognosis,
facilitate monitoring of systemic anticancer therapy, and help
in identifying appropriate therapeutic targets.
2
However, as the
concentration of CTCs is generally extremely low (a few to
hundreds per milliliter) in the bloodstream, detection and
characterization of CTCs present a tremendous technical chal-
lenge.
3,4
Over the past decade, various technology platforms
for isolating/counting CTCs have been developed using
dierent strategies such as flow cytometry, immune magnetic
beads, mechanical separation, or microfluidic devices.
58
However, the sensitivity of these emerging technologies relies
on the degree of enrichment of CTCs. Thus, the development
of a novel and sensitive platform that enhances CTCs capture,
allows for imaging of the captured CTCs, and enables quanti-
tative analysis would dramatically increase the use of CTCs in
diagnostics and prognostics.
Graphene and its derivatives have attracted considerable
attention due to their extremely large surface area, biocompati-
bility, excellent electrocatalytic activity, fast electron transfer,
high mechanical strength and high chemical stability.
9
Owing
to these extraordinary properties, graphene and its nano-
composites have been widely applied to modify a glassy carbon
electrode (GCE) for immobilizing various biomolecules.
1014
Thus, most of the graphene based electrochemical sensors
developed to date show two designs: the assembly of two-
dimensional (2D) graphene on the GCE or multi-stacked
graphene films. Further integration of the widely available
graphene sheets as 2D-nanoscale building blocks into a func-
tional three-dimensional (3D) system is essential to extend
a
Flexible Display Mater. & Tech. Co-Innovation Center of Hubei, Institute for
Interdisciplinary Research, Jianghan University, Wuhan 430056, PR China.
E-mail: zwy2428@163.com, cyp@jhun.edu.cn
b
Key Laboratory of Optoelectronic Chemical Materials and Devices of Ministry of
Education, Jianghan University, Wuhan 430056, PR China
c
Ecole Normale Supérieure, CNRS-ENS-UPMC UMR 8640, 24 Rue Lhomond, Paris
75005, France
16354 |Nanoscale,2015,7, 1635416360 This journal is © The Royal Society of Chemistry 2015
their biomedical applications.
15
Recently, a 3D hierarchical
nanostructured graphene oxide (GO) platform combined with
a ZnO nanorod array was successfully utilized to recognize/
capture epithelial cell-adhesion molecule (EpCAM)-expressing
cancer cells.
16
The mechanism relies on the enhanced local
topographic interactions between the substrate and nanoscale
components of the cellular surface, which improve the capture
eciency of the target cells to a certain extent.
1722
Herein, we present a convenient and cost-ecient 3D
microchip coated with GO and EpCAM antibody (anti-EpCAM,
Ab1) to serve as a cytosensor for the capture and analysis of
CTCs. With the remarkable development of nanotechnology,
nanomaterials have been extensively used to enhance the
sensitivity of cytosensors. Among them, gold nanoparticles
(AuNPs) are of particular interest owing to their merits of high
surface reactivity, good solubility, unique optical and electronic
characteristics as well as their excellent biocompatibility.
2325
Thus, clever combinations of dierent types of functional
nanostructured materials will enable the development of multi-
functional nanomedical platforms for diagnosis and therapy.
Among the various integrated nanocomposite-based systems,
mesoporous silica-based nanostructured materials have
attracted great interest, since they exhibit low cytotoxicity and
excellent chemical stability, and their surfaces can be easily
modified.
2629
Therefore, they are ideal platforms for con-
structing multifunctional materials that incorporate a variety
of functional nanostructured materials.
In this study, we developed a novel CTC-sensitive quantitative
detection system that integrates two functional components: (1) a
3D microchip with GO and an anti-EpCAM coating for recogniz-
ing/capturing EpCAM-expressing cells, and (2) Au-enwrapped
silica nanocomposites (Si/AuNPs) loaded with horseradish peroxi-
dase (HRP) and anti-CA153 (Ab2) to improve the selectivity of the
target cells and amplify the electrochemical detection signal. The
3D microchip arrays were initially prepared as illustrated in
Scheme 1A. The microchips were subsequently modified by GO,
streptavidin (SA), and biotinylated anti-EpCAM, respectively,
which served as 3D bioelectronic interfaces (Scheme 1B). In
addition, the Si/AuNPs were synthesized (Scheme 1C), and then
used as matrices to load the high-density signaling enzymes HRP
and Ab2. As a proof-of-concept, MCF7 breast cancer cells were
selected as model cells because of their high levels of EpCAM and
CA153 expression to demonstrate the feasibility of the supersand-
wich strategy. Finally, when sandwich-type immunoreactions were
carried out (Scheme 1D), the HRP on the surface could catalyze
the redox of H
2
O
2
in the presence of thionine, which served as
an electron transfer mediator. The proposed strategy exhibited
excellent sensitivity and selectivity, indicating its wide applica-
bility for research and clinical evaluations of cancer progression.
Experimental
Materials and reagents
The polydimethylsiloxane (PDMS) prepolymer kit was pur-
chased from Momentive Performance Materials (Waterford,
NY). The positive photoresist AZ 40XT-11D and the developer
AZ-300MIF were obtained from AZ Electronic Materials Corp.
(Philadelphia, PA). DMEM medium for cell culture was
obtained from GIBCO. 1-(3-(Dimethylamino)-propyl)-3-ethyl-
carbodiimide hydrochloride (EDC), N-hydroxysulfosuccinimide
(NHS), HPR, fluorescein diacetate (FDA) and SA were pur-
chased from Sigma (St. Louis, MO). The biotinylated goat IgG
polyclonal anti-EpCAM antibody and mouse monoclonal anti-
CA153 antibody were obtained from R&D Systems (Minneapo-
lis, MN). GO was obtained from Graphene Laboratories Inc.
(Calverton, NY). All other chemicals used in this study were of
analytical-grade. All solutions were freshly prepared using
ultrapure water (18 MΩ, Millipore).
Fabrication and surface modification of the 3D microchip
device
All the 3D microchip devices were designed with L-Edit soft-
ware and fabricated using a lithography fabrication technique,
which is illustrated in Scheme 1. The device was approximately
1 × 1 cm. All of the microchip masters were fabricated by spin-
coating with a positive photoresist (AZ 40XT-11D) on a silicon
substrate. Briefly, step 1 (Scheme 1A a to c): a layer of AZ
Scheme 1 (A) Schematic representation of the integrated fabrication of
polydimethylsiloxane (PDMS)-based 3D microchip arrays. (B) The micro-
chips were modied by GO, SA and biotinylated anti-EpCAM, respect-
ively. (C) Si/AuNPs were synthesized and used as nanocarriers to load
HPR and anti-CA153. (D) MCF7 cells bound to biotinylated anti-EpCAM
and HRPSi/AuNPAb2 were recognized by MCF7 cells on the
microchips.
Nanoscale Paper
This journal is © The Royal Society of Chemistry 2015 Nanoscale,2015,7, 1635416360 | 16355
40XT-11D was thrown at a clean and dry silicon wafer, which
was then exposed to ultraviolet light through a high-resolution
transparency mask (25 000 dpi). Step 2 (Scheme 1A d): a PDMS
prepolymer mixture (oligomer: the curing agent mass ratio =
10 :1) was poured over the negative grooves, which were placed
in suitable Petri dishes. Step 3 (Scheme 1A e): after curing at
80 °C for 2 h and peeling othe PDMS, positive linear arrays
of 3D PDMS-based microchips were obtained. Scheme 1A(f )
shows the scanning electron microscopy (SEM) images
(Hitachi, S-3400N) of the prepared 3D microchip arrays.
A total of 100 mg of GO was dispersed in 200 mL ethanol
(with 0.05% chitosan) with ultrasonication for 30 min, and
then 1 mL of the GO solution was mixed with 400 mM EDC
and 100 mM NHS in 1 mL of MES buer ( pH 5.2) for 30 min
at room temperature to activate the carboxyl groups on the sur-
faces of GO. After washing the mixture with phosphate-
buered saline (PBS), SA (50 μgmL
1
in PBS) was added to the
solution at 4 °C for 2 h and subsequently washed three times
with PBS to remove unreacted SA. Biotinylated anti-EpCAM
(10 μgmL
1
in PBS) was finally added. The microchips were
incubated with the resulting solution for 1 h at room tempera-
ture and dried to fabricate a functional film.
Synthesis of Si/AuNP nanocomposites
The Si/AuNPs were prepared according to a published pro-
cedure with slight modification.
30
Briefly, alcohol (EtOH;
2600 μL), H
2
O (500 μL), and ammonia-water (NH
3
·H
2
O;
300 μL) were mixed in a round-flask with stirring for 5 min.
Tetraethyl orthosilicate (170 μL) was then added to the mixture
and stirred for 8 h at room temperature. After the reaction, the
mixture was centrifuged at 15 000gfor 10 min, rinsed succes-
sively with H
2
O and EtOH three times, and then dried to
obtain the silica particles.
The Au-coated silica complex was obtained by chemical
reduction of an Au(III) solution with hydroxylamine in the pres-
ence of suspended silica nanoparticles. Firstly, SiO
2
(8 mg) was
well dissolved in HAuCl
4
·3H
2
O (1 mL, 0.1 mg mL
1
), and then
the solution was adjusted to pH 78 with 2 M NaOH, resulting
in a colour change to pink. NH
2
OH·HCl (30 μL, 0.22 M) was
added for the chemical reduction of Au(III) to Au(0), and the
colour immediately turned dark brown. The mixture was
placed in an orbital shaker at 120 rpm for 10 h followed by
resting for 10 h to discard the supernatant. The resulting
nanocomplex was resuspended in H
2
O. This procedure was
repeated and the precipitated conjugates were dispersed in
double-distilled water and stored at 4 °C.
Preparation of HRPSi/AuNPAb2 conjugates
First, the Si/AuNPs were sonicated with mercaptoacetic acid
for 2 h to generate carboxylated groups, and then the mixture
was washed repeatedly with PBS. Second, the carboxylated Si/
AuNPs were added to 1.0 mL 400 mM EDC and 100 mM NHS
in MES buer (pH 5.2), and sonicated for 30 min. The mixture
was centrifuged for 5 min at 13 000gand the supernatant was
discarded. Third, 500 μL of 400 ng mL
1
anti-CA153 and
500 μLof80μgmL
1
HRP were added to the mixture, which
was stirred for 4 h at room temperature. The mixture was
washed with PBS several times to remove unbound HRP
and Ab2. Finally, the particles were dispersed in 1.0 mL of
PBS (pH 7.4) containing 3% bovine serum albumin and stored
at 4 °C.
Cell culture
MCF7 cells were cultured in DMEM supplemented with 10%
fetal bovine serum, penicillin (100 μgmL
1
), and streptomycin
(100 μgmL
1
) in an incubator (5% CO
2
, 37 °C). After the con-
centration of cells reached 1 × 10
5
cells per mL, the cells were
collected using centrifugation at 1000gfor 3 min.
Construction of the supersandwich cytosensor
The MCF7 cell suspension (100 μL) was introduced onto the
modified 3D microchip device and retained in an incubator
(5% CO
2
, 37 °C) for 50 min, followed by washing with 0.05%
Tween-20 and PBS. Then, 100 μL HRPSi/AuNPAb2 was
dropped into the device and incubated at 37 °C for 50 min.
After rinsing and staining, the immobilized cells were imaged
by using SEM and a fluorescence microscope (Zeiss, Observer
Z1), and analyzed with the CHI 660E electrochemical work-
station. As a control, a flat substrate modified with GO/SAAb1
was also examined in parallel.
Results and discussion
Characterization of Si/AuNP and HRPSi/AuNPAb2 conjugates
The electrochemical responses were greatly influenced by the
Ab2. So we first synthesized novel Si/AuNPs that were used as
antibodies and HRP carriers to amplify the signals. Fig. 1A
shows the SEM and transmission electron microscopy images
of the Si/AuNPs, indicating that the silica nanospheres were
well covered with Au nanoparticles and the Si/AuNPs were
mono-dispersed with a size around 125 nm in diameter. Such
a uniform dispersion of the Si/AuNPs was vital for their conju-
gation with HRP and Ab2. X-ray photoelectron spectroscopy
(XPS) spectra were explored to confirm formation of the HRP
Si/AuNPAb2 conjugate. As shown in Fig. 1B, the conjugate
exhibited some single sharp N, Si and Au peaks in the XPS
spectra to indicate the successful formation of the HRP
Si/AuNPAb2 conjugate.
Ecient capture of CTCs with the 3D microchip device
The 3D microstructure cell-capture device was prepared as
illustrated in Scheme 1. To test the cell-capture performance of
the 3D microchip device, we prepared a cell suspension
(10
5
cells per mL) in cell culture medium (DMEM). The cell
suspension (100 μL) was introduced onto the microchip device,
which was placed in an incubator (5% CO
2
, 37 °C) for 50 min.
As controls, 3D microchips modified with SAAb1 and flat
substrates modified with GO/SAAb1 were examined in parallel.
After rinsing and staining, the substrate-immobilized cells were
characterized through microscopy imaging and electrochemical
behaviors. Fig. 2A shows the SEM images of immobilized
Paper Nanoscale
16356 |Nanoscale,2015,7,1635416360 This journal is © The Royal Society of Chemistry 2015
MCF7 cells at dierent magnifications after glutaraldehyde fix-
ation. The immobilized MCF7 cells, which were stained with
FDA, were also counted under a fluorescence microscope. As
shown in Fig. 2B, the 3D microchips modified with GO/SA
Ab1 could capture three times more cells than the microchips
only modified with SAAb1, and eight times more cells than
the flat substrate modified with GO/SAAb1. This result
suggests that the introduction of GO and the 3D microstruc-
ture was possibly responsible for the enhanced cell-capture
yields. This eect was likely due to the synergistic interplay
between GO and the 3D microchip, which provided more
surface area for SA and antibody binding and enhanced local
topographic interactions between the microarrays and CTCs.
Characterization of the supersandwich cytosensor
As shown in Fig. 3 (curve a), the cyclic voltammogram did not
display any detectable signal on the Ab1SA/GO/microchip in
PBS (pH 7.4). However, upon addition of thionine and H
2
O
2
,a
couple of stable and well-defined redox peaks were observed at
0.248 and 0.289 V (curve b), which corresponded to the elec-
trochemical response of thionine. When the cytosensor was
incubated with MCF7 cells at 10
5
cells per mL, no obvious
change in the signal was observed (data not shown). However,
after the sensor was incubated with the HRPAb2 solution, the
reduction current obviously increased on the HRPAb2/MCF7/
Ab1SA/GO/microchip, which was due to the catalysis of the
immobilized HRP toward the reduction of H
2
O
2
. Moreover,
when replacing HRPAb2 with HRPSi/AuNPAb2 as the detec-
tion antibody, the electrocatalytic current at the HRPSi/AuNP
Ab2/MCF7/Ab1SA/GO/microchip (curve d) increased substan-
tially. As expected, the achieved amplification of the signal
was ascribed to the increased amounts of enzymes introduced
on the microchips compared with the traditionally labeled
HRPAb2.
Fig. 1 (A) TEM (a) and SEM (b) images of Si/AuNPs. Inset: SEM image of
a silica nanosphere. (B) XPS spectra of HRPSi/AuNPAb2 conjugates.
Fig. 2 (A) SEM images of microchips on which MCF7 cells were cap-
tured, shown at dierent magnications. (B) Fluorescence micrographs
of 3D microchips modied with GO/SAAb1 (left), 3D microchips
modied with SAAb1 (middle), and the at substrate modied with GO/
SAAb1 (right) on which MCF7 cells were captured.
Fig. 3 Cyclic voltammograms of (a) Ab1SA/GO/microchip in pH 7.4
PBS and (b) Ab1SA/GO/microchip, (c) HRPAb2/MCF7/Ab1SA/GO/
microchip, (d) HRPSi/AuNPAb2/MCF7/Ab1SA/GO/microchip in pH
7.4 PBS containing 50 µM thionine and 2 mM H
2
O
2
. A concentration of
10
5
MCF7 cells per mL was used.
Nanoscale Paper
This journal is © The Royal Society of Chemistry 2015 Nanoscale,2015,7, 1635416360 | 16357
Optimization of experimental conditions
The electrochemical performance of the cytosensor is influ-
enced by some parameters such as the ratio of HRP to Ab2 and
the incubation time. To improve the sensitivity, the ratio of
HRP to Ab2 was optimized first because of the co-immobili-
zation of enzymes and antibodies on the Si/AuNP nanocarrier.
As shown in Fig. 4A, a marked increase of the peak current
was observed when increasing the HRP/Ab2 ratio from 50/1 to
200/1. This increased ratio caused an increase in the total
amount of HRP loaded per Si/AuNP, which was expected to
enhance the response during the immunoassay. However, the
excessive HRP reduced the available binding sites of Si/AuNPs
with Ab2, which would decrease the immune coupling
eciency for capturing MCF7 cells and result in a decreased
response. Therefore, the HRP/Ab2 ratio of 200/1 was selected
as the optimal condition for the preparation of HRPSi/AuNP
Ab2 conjugates. The BCA protein assay
31
showed that the con-
centration of active HRP in the HRPSi/AuNPAb2 dispersion
was 4.31 μgmL
1
.
In addition, the eects of incubation time on capturing
MCF7 cells and specifically recognizing HRPSi/AuNPAb2
were investigated. With increasing incubation time in the
MCF7 solution, the peak current increased and tended to be
steady after 50 min (red curve in Fig. 4B), indicating thorough
capturing of MCF7 cells on the 3D microchip device. When
HRPSi/AuNPAb2 was incubated with MCF7 in the second
step, the current increased and reached a plateau at 50 min
(black curve in Fig. 4B), reflecting the saturation of binding
sites between MCF7 and Ab2. A longer incubation time could
result in a large nonspecific signal. Therefore, the optimal
incubation time for the first and second immunoreactions was
determined to be 50 min, respectively.
Electrochemical detection of CTCs
For the detection of CTCs, high sensitivity and selectivity play
important roles in improving the treatment. In view of this,
the proposed supersandwich cytosensor was challenged with
dierent concentrations of MCF7 cells, as shown in Fig. 5A.
The dierential pulse voltammetry currents increased with the
increasing concentration of MCF7 cells. Fig. 5B shows the
linear calibration plots of the peak current (i
p
)versus the con-
centration of MCF7 cells. A linear relationship between i
p
and
the logarithm of the cell concentration is found in the range of
10
1
10
7
cells per mL, with a correlation coecient of 0.9921
(n= 3). The detection limit at a signal-to-noise ratio of 3σ
(where σis the standard deviation of the signal in a blank solu-
tion) was estimated to be 10 cells per mL, which is comparable
and much lower than the values reported in the literature
(summarized in Table 1). The excellent sensitivity is attributed
to double signal amplifications from GO and Si/AuNPs,
which greatly accelerate the rate of electron transfer on the
cytosensor.
Performance of the supersandwich cytosensor
To investigate the specificity of the supersandwich cytosensor
toward MCF7 cells, dierent possible interfering cells such as
HeLa and A549 were used. As shown in Fig. 5A (curves b and
c), the peak current of the cytosensor showed a negligible
change with the addition of the interfering cells, whereas a
substantial electrochemical signal increase was observed with
the addition of MCF7 cells. These results demonstrated the
good selectivity of the cytosensor for MCF7 cells. Thus, the
proposed strategy can be applied to dierent types of CTCs
using the corresponding Ab2.
To evaluate the intra- and inter-assay coecients of vari-
ation of the cytosensor array, the intra-assay precision of the
proposed method was measured for six replicate measure-
ments. The coecient of variation of the intra-assay was 3.5%
at 10
5
cells per mL. Likewise, the inter-assay coecient of vari-
ation on six cytosensors was 4.3% at 10
5
cells per mL. The pre-
Fig. 4 (A) Eects of the HRP to Ab2 ratio on the current responses in
the presence of 10
5
MCF7 cells per mL. (B) Eect of incubation time on
the current responses by capturing MCF7 (red curve) and recognizing
between HRPSi/AuNPAb2 and MCF7 (black curve) in the presence of
10
5
MCF7 cells per mL.
Fig. 5 (A) Dierential pulse voltammetry responses of the supersand-
wich cytosensor incubated with (b) HeLa cells, (c) A549 cells at 10
5
cells
per mL and (a, dj) dierent concentrations of MCF7 cells: 0, 10
1
,10
2
,
10
3
,10
4
,10
5
,10
6
,10
7
cells per mL. (B) Calibration curve of MCF7.
Table 1 Comparison of the sensitivity of dierent CTC cytosensors
Cytosensor type
Linear range
(cells per mL)
Detection limit
(cells per mL) Ref.
Microchip cytosensor 10
1
to 10
7
10 Present study
LSAW aptasensor 10
2
to 10
7
32 32
PEC biosensor 10
2
to 10
6
58 33
Colorimetric aptasensor 10
2
to 10
4
40 34
Aptamer/QD cytosensor 10
2
to 10
6
50 35
Paper Nanoscale
16358 |Nanoscale,2015,7, 1635416360 This journal is © The Royal Society of Chemistry 2015
cision and reproducibility of the cytosensor are acceptable.
Moreover, when the cytosensor was stored at 4 °C, the analyti-
cal performance did not decline obviously after one week, and
93% of the initial response remained after one month. The
stability is attributed to the stable structure of the Si/AuNPs
and the strong interaction between anti-EpCAM and GO modi-
fied 3D microchips. Thus, the designed strategy shows good
performance for the detection of CTCs with a broad detection
range, low detection limit, excellent selectivity, good reproduci-
bility and stability.
In this study, the supersandwich cytosensor shows attrac-
tive performance for the quantification of MCF7 cells, such as
its wide linear range and low detection limit. First, the GO
modified 3D microchips provide large specific surface areas to
facilitate abundant binding of SA. Combined with the extra-
ordinary biocompatibility of GO, the microchips are very suit-
able for immobilizing anti-EpCAM with excellent stability and
bioactivity. Therefore, an ideal interface for cell capture is pro-
vided, which improves the sensitivity of detection compared to
previous designs. Second, the supersandwich strategy helps to
further enhance the sensitivity via the signal amplification of
Si/AuNPs. Indeed, the introduction of an HRPSi/AuNPAb2
probe led to a remarkable increase of the signal.
Conclusions
In summary, due to the increase of local topographic inter-
actions between the substrate and CTCs, the formation of
sandwich-like structures and double signal amplifications
through the use of GO and Si/AuNPs, the proposed supersand-
wich cytosensor shows excellent performance for analysis of
CTCs with a wide linear range, low detection limit, and accep-
table stability, reproducibility and accuracy. Among the various
technologies developed to specifically recognize and capture
CTCs to date, this is the first strategy to combine 3D micro-
chips and an electrochemical method for quantitative detec-
tion of CTCs. Therefore, we anticipate that this 3D microchip
device can be extended for the determination of other CTCs
and shows great potential in the field of disease diagnostics
and clinical analysis.
Acknowledgements
This work was supported by the National Natural Science
Foundation of China (no. 81402466) and the Natural Science
Foundation of Hubei (no. 2015CFB610). W. Z. would like to
acknowledge the support from a Jianghan University start-up
grant. We would like to thank Editage (http://www.editage.cn/)
for English language editing.
References
1 M. Cristofanilli, G. T. Budd, M. J. Ellis, A. Stopeck,
J. Matera, M. C. Miller, J. M. Reuben, G. V. Doyle,
W. J. Allard, L. W. M. M. Terstappen and D. F. Hayes,
N. Engl. J. Med., 2004, 351, 781.
2 K. Pantel and C. Alix-Panabieres, Trends Mol. Med., 2010,
16, 398.
3 V. Murlidhar, M. Zeinali, S. Grabauskiene, M. Ghannad-
Rezaie, M. S. Wicha, D. M. Simeone, N. Ramnath,
R. M. Reddy and S. Nagrath, Small, 2014, 10, 4895.
4 T. JunáHuang, Lab Chip, 2013, 13, 602.
5 S. Nagrath, L. V. Sequist, S. Maheswaran, D. W. Bell,
D. Irimia, L. Ulkus, M. R. Smith, E. L. Kwak,
S. Digumarthy, A. Muzikansky, P. Ryan, U. J. Balis,
R. G. Tompkins, D. A. Haber and M. Toner, Nature, 2007,
450, 1235.
6 S. K. Singh, C. Hawkins, I. D. Clarke, J. A. Squire, J. Bayani,
T. Hide, R. M. Henkelman, M. D. Cusimano and
P. B. Dirks, Nature, 2004, 432, 396.
7 S. S. Banerjee, A. J. Badhwar, K. R. Zope, K. J. Todkar,
R. R. Mascarenhas, G. P. Chate, G. V. Khutale, A. Bharde,
M. Calderon and J. J. Khandare, Nanoscale, 2015, 7,
8684.
8 X. Hu, C. W. Wei, J. Xia, I. Pelivanov, M. ODonnell and
X. Gao, Small, 2013, 9, 2046.
9 A. K. Geim, Science, 2009, 324, 1530.
10 G. S. Kulkarni, K. Reddy, Z. Zhong and X. Fan, Nat.
Commun., 2014, 5, 4376.
11 R. Alam, I. V. Lightcap, C. J. Karwacki and P. V. Kamat, ACS
Nano, 2014, 8, 7272.
12 J. Wang and X. Qu, Nanoscale, 2013, 5, 3589.
13 H. K. Choi, H. Y. Jeong, D. S. Lee, C. G. Choi and
S. Y. Choi, Carbon, 2013, 14, 186.
14 X. Zhang, Y. Zhang, Q. Liao, Y. Song and S. Ma, Small,
2013, 23, 4045.
15 H. J. Yoon, T. H. Kim, Z. Zhang, E. Azizi, T. M. Pham,
C. Paoletti, J. Lin, N. Ramnath, M. S. Wicha, D. F. Hayes,
D. M. Simeone and S. Nagrath, Nat. Nanotechnol., 2013, 8,
735.
16 S. Yin, Y. L. Wu, B. Hu, Y. Wang, P. Cai, C. K. Tan, D. Qi,
L. Zheng, W. R. Leow, N. S. Tan, S. Wang and X. Chen, Adv.
Mater. Interfaces, 2014, 1, 1300043.
17 H. J. Yoon, M. Kozminsky and S. Nagrath, ACS Nano, 2014,
8, 1995.
18 N. Zhang, Y. Deng, Q. Tai, B. Cheng, L. Zhao, Q. Shen,
R. He, L. Hong, W. Liu, S. Guo, K. Liu, H. R. Tseng,
B. Xiong and X. Z. Zhao, Adv. Mater., 2012, 24, 2756.
19 S. Hou, L. Zhao, Q. Shen, J. Yu, C. Ng, X. Kong, D. Wu,
M. Song, X. Shi, X. Xu, W. H. OuYang, R. He, X. Z. Zhao,
T. Lee, F. C. Brunicardi, M. A. Garcia, A. Ribas, R. S. Lo and
H. R. Tseng, Angew. Chem., Int. Ed., 2013, 52, 3379.
20 S. Hou, H. Zhao, L. Zhao, Q. Shen, K. S. Wei, D. Y. Suh,
A. Nakao, M. A. Garcia, M. Song, T. Lee, B. Xiong, S. C. Luo,
H. R. Tseng and H. H. Yu, Adv. Mater., 2013, 25, 1547.
21 S. Wang, K. Liu, J. Liu, Z. T. F. Yu, X. Xu, L. Zhao, T. Lee,
E. K. Lee, J. Reiss, Y. K. Lee, L. W. K. Chung, J. Huang,
M. Rettig, D. Seligson, K. N. Duraiswamy, C. K. F. Shen and
H. R. Tseng, Angew. Chem., Int. Ed., 2011, 50, 3084.
22 X. Liu and S. Wang, Chem. Soc. Rev., 2014, 43, 2385.
Nanoscale Paper
This journal is © The Royal Society of Chemistry 2015 Nanoscale,2015,7, 1635416360 | 16359
23 O. Yehezkeli, R. Tel-Vered, S. Raichlin and I. Willner, ACS
Nano, 2011, 5, 2385.
24 Z. K. Han and Y. Gao, Nanoscale, 2015, 7, 308.
25 A. Li, P. Zhang, X. Chang, W. Cai, T. Wang and J. Gong,
Small, 2015, 11, 1892.
26 W. Chen, P. H. Tsai, Y. Hung, S. Chiou and C. Y. Mou, ACS
Nano, 2013, 7, 8423.
27 D. Tarn, D. P. Ferris, J. C. Barnes, M. W. Ambrogio,
J. F. Stoddart and F. I. Zink, Nanoscale, 2014, 6, 3335.
28 N. Z. Knezevic and V. S. Y. Lin, Nanoscale, 2013, 5,
1544.
29 L. Zhu, H. Wang, X. Shen, L. Chen, Y. Wang and H. Chen,
Small, 2012, 8, 1857.
30 K. Leopold, M. Foulkes and P. J. Worsfold, Anal. Chem.,
2009, 81, 3421.
31 M. M
´evel, G. Breuzard, J. J. Yaouanc, J. C. Cĺement,
P. Lehn, C. Pichon, P. A. Jar
`es and P. Midoux, ChemBio-
Chem., 2008, 9, 1462.
32 K. Chang, Y. Pi, W. Lu, F. Wang, F. Pan, F. Li, S. Jia, J. Shi,
S. Deng and M. Chen, Biosens. Bioelectron., 2014, 60, 318.
33 F. Liu, Y. Zhang, J. Yu, S. Wang, S. Ge and X. Song, Biosens.
Bioelectron., 2014, 51, 413.
34 X. Zhang, K. Xiao, L. Cheng, H. Chen, B. Liu, S. Zhang and
J. Kong, Anal. Chem., 2014, 86, 5567.
35 H. Liu, S. Xu, Z. He, A. Deng and J. Zhu, Anal. Chem., 2013,
85, 3385.
Paper Nanoscale
16360 |Nanoscale,2015,7, 1635416360 This journal is © The Royal Society of Chemistry 2015
... Most importantly, it can differentiate CTCs from common cells, reflecting the hopeful programs of this technique for clinical detection and medicinal monitoring of cancers. [77]. ...
... In developing areas Scheme 10. Schematic illustration of the united fabrication of three-dimensional microchip arrays based on PDMS [77]. and disaster scenes where only very limited resources are readily accessible, POC system is an attractive tool to diagnose patients for proper clinical management [86]. ...
Article
Full-text available
Cancer metastasis is one of the foremost causes of cancer incidence and fatality in the whole of the world. Circulating tumor cells (CTC) have been confirmed to be among the most significant stimuli of metastasis in recent years and presently are the subject of extensive research aiming to be accurately identified by using biological and physical properties. Among the various studies conducted for isolation, identification, and characterization of CTCs, microfluidic systems have aroused great attention owing to their unique advantages such as low-cost, simplicity, reduction in reagent consumption, miniaturization, fast and precise control. The purpose of this review is to provide an overview of current state of the microfluidic biosensors for the screening of CTCs. Additionally, given the recent progress in this field, future outlook for the development of the microfluidics biosensing is briefly discussed.
... Aptamer/electroactive species-loaded AuNP probes 5 to 500 cells/mL 4 cells/mL for Ramos cells 3 cells/mL for CCRF-CEM cells [39] MCF-7/S1/Gr/AuNPs/GCE 50 to 1 × 10 4 cells/mL 27 cells/mL [134] MPA/Rh-NPs/rGONs/graphite electrode 5.0 to 1.0 × 10 5 cells/mL 1 cells/mL [133] Fe 3 O 4 NPs/rGO/MoS 2 15 to 45 cells/mL 6 cells/mL [135] FA-GAM-OA 5.0 to 1.0 × 10 6 cells/mL 5 cells/mL [84] A/Glu-GQD-Pd@Au 3.0 to 1.0 × 10 6 cells/mL 2 cells/mL [80] Herceptin-conjugated graphene 1 to 80 cells/mL - [136] Apta-PGO-LAPS' 5 to 5000 cells/mL - [137] MB-aptamers/dsDNA/AuNP-modified GE 1 × 10 2 to 1 × 10 6 cells/mL 23 cells/mL [138] HRP-Si/AuNPs-Ab2/MCF7/Ab1-SA/GO/microchip 1 to 10 5 cells/mL 10 cells/mL [139] ...
Article
Full-text available
Graphene is an emerging nanomaterial increasingly being used in electrochemical biosensing applications owing to its high surface area, excellent conductivity, ease of functionalization, and superior electrocatalytic properties compared to other carbon-based electrodes and nanomaterials, enabling faster electron transfer kinetics and higher sensitivity. Graphene electrochemical biosensors may have the potential to enable the rapid, sensitive, and low-cost detection of cancer biomarkers. This paper reviews early-stage research and proof-of-concept studies on the development of graphene electrochemical biosensors for potential future cancer diagnostic applications. Various graphene synthesis methods are outlined along with common functionalization approaches using polymers, biomolecules, nanomaterials, and synthetic chemistry to facilitate the immobilization of recognition elements and improve performance. Major sensor configurations including graphene field-effect transistors, graphene modified electrodes and nanocomposites, and 3D graphene networks are highlighted along with their principles of operation, advantages, and biosensing capabilities. Strategies for the immobilization of biorecognition elements like antibodies, aptamers, peptides, and DNA/RNA probes onto graphene platforms to impart target specificity are summarized. The use of nanomaterial labels, hybrid nanocomposites with graphene, and chemical modification for signal enhancement are also discussed. Examples are provided to illustrate applications for the sensitive electrochemical detection of a broad range of cancer biomarkers including proteins, circulating tumor cells, DNA mutations, non-coding RNAs like miRNA, metabolites, and glycoproteins. Current challenges and future opportunities are elucidated to guide ongoing efforts towards transitioning graphene biosensors from promising research lab tools into mainstream clinical practice. Continued research addressing issues with reproducibility, stability, selectivity, integration, clinical validation, and regulatory approval could enable wider adoption. Overall, graphene electrochemical biosensors present powerful and versatile platforms for cancer diagnosis at the point of care.
... CTCs have promising potential in monitoring the tumor prognosis, treatment effect, and metastasis condition [8][9][10]. However, due to the rare existence of CTCs in peripheral blood, the isolation and identification of variable CTCs is still challenging [11][12][13]. Epithelial to mesenchymal transition (EMT) plays a significant role during the CTCs' metastatic process. It results in a phenotypic change in CTCs, such as transforming of epithelial properties to mesenchymal properties, and poses hurdles in CTC isolation [14,15]. ...
Article
Full-text available
Ovarian cancer (OC) is a lethal disease occurring in women worldwide. Due to the lack of obvious clinical symptoms and sensitivity biomarkers, OC patients are often diagnosed in advanced stages and suffer a poor prognosis. Circulating tumor cells (CTCs), released from tumor sites into the peripheral blood, have been recognized as promising biomarkers in cancer prognosis, treatment monitoring, and metastasis diagnosis. However, the number of CTCs in peripheral blood is low, and it is a technical challenge to isolate, enrich, and identify CTCs from the blood samples of patients. This work develops a simple, effective, and inexpensive strategy to capture and identify CTCs from OC blood samples using the folic acid (FA) and antifouling-hydrogel-modified fluorescent-magnetic nanoparticles. The hydrogel showed a good antifouling property against peripheral blood mononuclear cells (PBMCs). The FA was coupled to the hydrogel surface as the targeting molecule for the CTC isolation, held a good capture efficiency for SK-OV-3 cells (95.58%), and successfully isolated 2-12 CTCs from 10 OC patients' blood samples. The FA-modified fluorescent-magnetic nanoparticles were successfully used for the capture and direct identification of CTCs from the blood samples of OC patients.
... Furthermore, the physical and chemical properties of interaction between cells and the extracellular matrix can be applicable to cell-capture platforms for the improvement of capture yield, purity and throughput through nanomaterials [17]. In particular, graphene based materials have been regarded as one of the promising materials as they can effectively serve as biointerfaces for water insoluble cancer drug carriers, photothermal cancer therapy and bacteria or DNA biosensors with biocompatible properties [18][19][20][21]. ...
Article
Full-text available
The analysis of circulating tumor cells (CTCs) in the peripheral blood of cancer patients is critical in clinical research for further investigation of tumor progression and metastasis. In this study, we present a novel surface-enhanced Raman scattering (SERS) substrate for the efficient capture and characterization of cancer cells using silver nanoparticles-reduced graphene oxide (AgNPs-rGO) composites. A pulsed laser reduction of silver nanowire-graphene oxide (AgNW-GO) mixture films induces hot-spot formations among AgNPs and artificial biointerfaces consisting of rGOs. We also use in situ electric field-assisted fabrication methods to enhance the roughness of the SERS substrate. The AgNW-GO mixture films, well suited for the proposed process due to its inherent electrophoretic motion, is adjusted between indium tin oxide (ITO) transparent electrodes and the nano-undulated surface is generated by applying direct-current (DC) electric fields during the laser process. As a result, MCF7 breast cancer cells are efficiently captured on the AgNPs-rGO substrates, about four times higher than the AgNWs-GO films, and the captured living cells are successfully analyzed by SERS spectroscopy. Our newly designed bifunctional substrate can be applied as an effective system for the capture and characterization of CTCs.
... The sensor integrates two functional components: (1) an anti-EpCAM coating on GO for recognizing/capturing EpCAM-expressing cells, and (2) horseradish peroxidase (HPR) and anti-CA15-3 (Ab2) loaded in Si/AuNPs to improve the selectivity of target cells and amplify the electrochemical detection signal. The performance was assessed on MCF7 breast cancer cells, showing high sensitivity with a wide range of 10 1 to 10 7 cells mL −1 and a detection limit of 10 cells mL −1 [64]. A CTC isolation platform based on GO functionalized polyester fabric sheets bearing anti-EpCAM antibodies has been proposed as a low-cost, easy-to-fit, and disposable platform, assuring high sensitivity. ...
Article
Full-text available
Graphene-based materials are intriguing nanomaterials with applications ranging from nanotechnology-related devices to drug delivery systems and biosensing. Multifunctional graphene platforms were proposed for the detection of several typical biomarkers (i.e., circulating tumor cells, exosomes, circulating nucleic acids, etc.) in liquid biopsy, and numerous methods, including optical, electrochemical, surface-enhanced Raman scattering (SERS), etc., have been developed for their detection. Due to the massive advancements in biology, material chemistry, and analytical technology, it is necessary to review the progress in this field from both medical and chemical sides. Liquid biopsy is considered a revolutionary technique that is opening unexpected perspectives in the early diagnosis and, in therapy monitoring, severe diseases, including cancer, metabolic syndrome, autoimmune, and neurodegenerative disorders. Although nanotechnology based on graphene has been poorly applied for the rapid diagnosis of viral diseases, the extraordinary properties of graphene (i.e., high electronic conductivity, large specific area, and surface functionalization) can be also exploited for the diagnosis of emerging viral diseases, such as the coronavirus disease 2019 (COVID-19). This review aimed to provide a comprehensive and in-depth summarization of the contribution of graphene-based nanomaterials in liquid biopsy, discussing the remaining challenges and the future trend; moreover, the paper gave the first look at the potentiality of graphene in COVID-19 diagnosis.
Article
Graphene has been rightly called the wonderous 2D material owing to its several unique properties. Despite continuous research efforts on the development of reduced graphene oxide (rGO), its optical properties remain largely unexplored. Herein we have explored the light irradiation effects on the optical and structural properties of rGO. Crystalline natured rGO was prepared by simple microwave exfoliation and ultra-sonication technique. Furthermore, rGO of hydrophobic (pho-rGO) and hydrophilic (phi-rGO) nature were derived from the same source. Transmission Electron Microscopy (TEM) and Dynamic Light Scattering (DLS) measurements revealed a reduction in the sheet size of phi-rGO upon light irradiation. Moreover, an increment in the light transmission was also observed in the light treated phi-rGO, which was verified for solar cell application. On the other hand, pho-rGO was used in creating a dust-repellent coating on glass surface without impacting light transmission.
Article
A three-dimensional (3D) graphene macroporous foam substrate with ZnO nanorod array was developed for specific and non-invasive detection of circulating tumor cells (CTCs). With the help of the epithelial cell adhesion molecule antibody (anti-EpCAM) and the array structure, the EpCAM-positive CTCs can adhere on the detection substrate (rGO-ZnO-antiEpCAM foam). Moreover, the resistance and the electrochemical impedance spectroscopy (EIS) of the rGO-ZnO-antiEpCAM foam was changing during the CTCs adhering process with high sensitivity. Compared to the EpCAM-negative cancer cell, the resistance of the substrate is rising with the amount of the EpCAM-positive CTCs. The possible reason is that the negative charges of CTCs affected the conductivity of the p-type reduced graphene oxide (rGO), and when CTCs were adhering to the rGO-ZnO-antiEpCAM foam, the resistance of the rGO-ZnO-antiEpCAM foam increased. In addition, the EIS measurement also shows the regular changes during the adhering process. From the equivalent circuit modeling of EIS, the charge transfer resistance (Rct) on the surface of the rGO-ZnO-antiEpCAM foam electrode had a linear growth. This may be due to the fixation of negatively charged Breast cancer cells (MCF-7) on the substrate, the charge transfer process was hindered, which caused the semicircle domain of the EIS curve to expand significantly with time. These results reveal that CTCs can not only be specifically adhered on the rGO-ZnO-antiEpCAM foam, but also the electrical microenvironment of the detection substrate was changed during CTCs adhering process. The construction of the detection substrate, the measurement and analysis methods of the electrical signals are of great significance in CTCs acquisition and sensing, which will provide an effective strategy for the real-time monitoring of CTCs at the clinic.
Article
Full-text available
Leukemia stem cells (LSCs) are suitable candidates to be deployed for the diagnosis and therapy of acute myeloid leukemia (AML) patients. In this study, a novel electrochemical cytosensor was designed for the sensitive detection and quantification of KG1a cells as a model of LSCs. The developed cytosensor was based on the overexpression of cell surface protein CD123 by leukemia KG1a cells. For this purpose, the glassy carbon electrode was modified by graphene quantum dots (GQDs), Au nanoparticles, streptavidin coated AuNPs, biotinylated CD123 antibody and target cells. The dense loading of CD123 antibody and electrical enhancement on the modified electrode were carried out using GQDs, this resulting in a sensitive detection of CD123 positive cells within KG1a cells. Step by step preparation of the nanomaterial-based cytosensor and its optimization steps were confirmed by different electrochemical techniques. The field emission scanning electron microscopy (FE-SEM) images also confirmed the proper attachment of the materials and the cells on the surface of the modified electrode. The linear detection range (LDR) and limit of detection (LOD) of the developed electrochemical biosensor were recorded as 1 cell/mL and 1-25 cells/mL, respectively, which is remarkable. Importantly, the present findings are precise and highly selective in the presence of other leukemia cells (NB4, HL60, and U937 cells). Further, the versatility and accuracy of the proposed cytosensor were evaluated using clinical samples. We believe that the cytosensor proposed in this study has the potential to serve as a next generation sensor for the early detection of leukemia stem cells.
Article
The biological compositions including cells, proteins/peptides, nucleic acids, extracellular vesicles (EVs) and metabolites are no doubt closely related to almost all physiological and pathological events in organism. Parts of them with specific characteristics can serve as therapeutic target or even disease indicator at early stage. However, all of them especially the specific portion associated with diseases are surrounded by complex environment where there are a large number of high-abundance proteins and salts and the like, leading to difficulties in direct analysis for them. Much efforts have been made to solve this problem, among which nanomaterials with outstanding flexibility in preparation and manipulation are attracting great attention. In this review, we provide a relatively comprehensive elaboration on recent advancement of nanomaterials as sample technique for bio-analysis, focusing on their preparation and application in all the above-mentioned biological compositions. Furthermore, the limits and development tendency of nanomaterials in this field are proposed.
Article
Full-text available
Fine gold nanorod@TiO2 yolk‐shell catalysts are synthesized by an improved silica template method. With a hollow TiO2 shell and a unique tunable cylindrical gold core, the catalyst exhibits a high surface area and a wide range of photoabsorption, from ultraviolet to near infrared. The remarkable photochemical activity is obtained when such catalyst is utilized to oxidize benzyl alcohol. image
Article
Full-text available
Detailed knowledge of the structures of gold nanoparticles on ceria surface is of fundamental importance to understand their extraordinary activities in catalysis. In this work, we employ density functional theory with the inclusion of the on-site Coulomb interaction (DFT+U) to investigate the structure evolution of small-sized gold (Au、Au4、Au8 and Au12) clusters on the four types of reduced CeO2-X(111) surfaces: SSV (single surface oxygen vacancy), LSVT (linear surface oxygen vacancy trimer), dLSVC (double linear surface oxygen vacancy with a surface vacancy dimer and a subsurface vacancy), and TSVT (triangular surface oxygen vacancy trimer). Our results indicate the relative stabilities of multilayer (3D) structures are strengthened gradually compared with the monolayer (2D) structures with the increasing number of gold atoms. In addition, the 2D-3D structure transition occurs in the size order of Au2D→3D@TSVT>Au2D→3D@dLSVC~Au2D→3D@LSVT>Au2D→3D@SSV, which is determined by the charge transfer magnitude between the CeO2 surfaces and gold clusters. Meanwhile, two competitive nucleation patterns are observed, fcc-like nucleation and hcp-like nucleation, which highly affect the morphology of supported gold clusters. Further site-by-site investigations indicate the coordination number and the charges of Au atoms are the dominant factors to influence the adsorption strength of CO and O2, and the interface plays a relative minor role. These findings not only enrich the knowledge of the relationship among the surface defects, the gold cluster structures and small molecules’ adsorptions, but provide a theoretical perspective to help design the optimal Au/CeO2 systems with high catalytic efficiency.
Article
Full-text available
We report a highly sensitive NO2 gas sensor based on multi-layer graphene (MLG) films synthesized by a chemical vapor deposition method on a microheater-embedded flexible substrate. The MLG could detect low-concentration NO2 even at sub-ppm (<200 ppb) levels. It also exhibited a high resistance change of similar to 6% when it was exposed to 1 ppm NO2 gas at room temperature for 1 min. The exceptionally high sensitivity could be attributed to the large number of NO2 molecule adsorption sites on the MLG due to its a large surface area and various defect-sites, and to the high mobility of carriers transferred between the MLG films and the adsorbed gas molecules. Although desorption of the NO2 molecules was slow, it could be enhanced by an additional annealing process using an embedded Au microheater. The outstanding mechanical flexibility of the graphene film ensures the stable sensing response of the device under extreme bending stress. Our large-scale and easily reproducible MLG films can provide a proof-of-concept for future flexible NO2 gas sensor devices.
Article
Here, we report non-invasive strategy for isolating cancer cells by autonomously propelled carbon nanotube (CNT) microrockets. H2O2-driven oxygen (O2) bubble-propelled microrockets were synthesized using CNT and Fe3O4 nanoparticles in the inner surface and covalently conjugating transferrin on the outer surface. Results show that self-propellant microrockets can specifically capture cancer cells.
Article
Circulating tumor cells (CTCs) are believed to play an important role in metastasis, a process responsible for the majority of cancer-related deaths. But their rarity in the bloodstream makes microfluidic isolation complex and time-consuming. Additionally the low processing speeds can be a hindrance to obtaining higher yields of CTCs, limiting their potential use as biomarkers for early diagnosis. Here, a high throughput microfluidic technology, the OncoBean Chip, is reported. It employs radial flow that introduces a varying shear profile across the device, enabling efficient cell capture by affinity at high flow rates. The recovery from whole blood is validated with cancer cell lines H1650 and MCF7, achieving a mean efficiency >80% at a throughput of 10 mL h−1 in contrast to a flow rate of 1 mL h−1 standardly reported with other microfluidic devices. Cells are recovered with a viability rate of 93% at these high speeds, increasing the ability to use captured CTCs for downstream analysis. Broad clinical application is demonstrated using comparable flow rates from blood specimens obtained from breast, pancreatic, and lung cancer patients. Comparable CTC numbers are recovered in all the samples at the two flow rates, demonstrating the ability of the technology to perform at high throughputs.
Article
Nearly all existing nanoelectronic sensors are based on charge detection, where molecular binding changes the charge density of the sensor and leads to sensing signal. However, intrinsically slow dynamics of interface-trapped charges and defect-mediated charge-transfer processes significantly limit those sensors' response to tens to hundreds of seconds, which has long been known as a bottleneck for studying the dynamics of molecule-nanomaterial interaction and for many applications requiring rapid and sensitive response. Here we report a fundamentally different sensing mechanism based on molecular dipole detection enabled by a pioneering graphene nanoelectronic heterodyne sensor. The dipole detection mechanism is confirmed by a plethora of experiments with vapour molecules of various dipole moments, particularly, with cis- and trans-isomers that have different polarities. Rapid (down to ~\n0.1 s) and sensitive (down to ~\n1 ppb) detection of a wide range of vapour analytes is achieved, representing orders of magnitude improvement over state-of-the-art nanoelectronics sensors.
Article
Smart material nanoassemblies that can simultaneously sense and shoot low level contaminants from air and water are important for overcoming the threat of hazardous chemicals. Graphene oxide (GO) sheets deposited on mesoscopic TiO2 films that underpin the deposition of Ag nanoparticles with UV-irradiation provide the foundation for the design of a smart material. The Ag particle size is readily controlled through precursor concentration and UV irradiation time. These semiconductor - graphene oxide - metal (SGM) films are SERS active and hence capable of sensing aromatic contaminants such as nitrobenzenethiol (NBT) in nanomolar range. Increased local concentration of organic molecule achieved through interaction with 2D carbon support (GO) facilitates low-level detection of contaminants. Upon UV irradiation of the NBT loaded SGM film, one can induce photocatalytic transformations. Thus, each component of the SGM film plays a pivotal role in aiding the detection and degradation of a contaminant dispersed in aqueous solutions. The advantage of using SGM films as multipurpose "detect and destroy" systems for nitroaromatic molecule is discussed.
Article
Rapid and efficient detection of cancer cells at their earliest stages is one of the central challenges in cancer diagnostics. We developed a simple, cost-effective, and highly sensitive colorimetric method for visually detecting rare cancer cells based on cell-triggered cyclic enzymatic signal amplification (CTCESA). In the absence of target cells, hairpin aptamer probes (HAPs) and linker DNAs stably coexist in solution, and the linker DNA assemble DNA-AuNPs, producing a purple solution. In the presence of target cells, the specific binding of HAPs to the target cells triggers a conformational switch that results in linker DNA hybridization and cleavage by nicking endonuclease-strand scission cycles. Consequently, the cleaved fragments of linker DNA can no longer assemble into DNA-AuNPs, resulting in a red color. UV-vis spectrometry and photograph analyses demonstrated that this CTCESA-based method exhibited selective and sensitive colorimetric responses to the presence of target CCRF-CEM cells, which could be detected by the naked eye. The linear response for CCRF-CEM cells in a concentration range from 102 to 104 cells was obtained with a detection limit of 40 cells, which is approximately 20 times lower than the detection limit of normal AuNP-based methods without amplification. Given the high specificity and sensitivity of CTCESA, this colorimetric method provides a sensitive label-free and cost-effective approach for early cancer diagnosis and point-to-care applications.
Article
Circulating tumor cells (CTCs) are low frequency cells found in the bloodstream after having been shed from a primary tumor. These cells are research targets because of the information they may potentially provide about both an individual cancer as well as the mechanisms through which cancer spreads in the process of metastasis. Established technologies exist for CTC isolation, but the recent progress and future of this field lie in nanomaterials. In this review, we provide perspective into historical CTC capture as well as current research being conducted, emphasizing the significance of the materials being used to fabricate these devices. The modern investigation into CTCs initially featured techniques that have since been commercialized. A major innovation in the field was the development of a microfluidic capture device, first fabricated in silicon and followed up with glass and thermopolymer devices. We then specifically highlight the technologies incorporating magnetic nanoparticles, carbon nanotubes, nanowires, nanopillars, nanofibers, and nanoroughened surfaces, graphene oxide and their fabrication methods. The nanoscale provides a new set of tools that has the potential to overcome current limitations associated with CTC capture and analysis. We believe the current trajectory of the field is in the direction of nanomaterials, allowing the improvements necessary to further CTC research.