Download as pdf or txt
Download as pdf or txt
You are on page 1of 14

Polymer Testing 115 (2022) 107746

Contents lists available at ScienceDirect

Polymer Testing
journal homepage: www.elsevier.com/locate/polytest

Application of fluorescent nano-biosensor for the detection of cancer


bio-macromolecular markers
Nan Ouyang a, 1, Lei Hong a, b, 1, Yuanshuai Zhou c, Jingzhong Zhang c, Shaheryar Shafi a,
Jinlin Pan a, Rongchuan Zhao a, Ying Yang d, **, Wenya Hou e, *
a
School of Biomedical Engineering, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
b
Suzhou Science & Technology Town Hospital, Suzhou, 215153, China
c
Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, 215163, China
d
The People’s Hospital of Suzhou New District, Suzhou, China
e
Shenzhen University General Hospital, Guangdong Key Laboratory for Genome Stability & Disease Prevention, Shenzhen University School of Medicine, Shenzhen,
Guangdong, 518060, China

A R T I C L E I N F O A B S T R A C T

Keywords: Fluorescent sensors developed based on nano-materials have penetrated into cancer medical sensing research
Cancer due to their unique properties, such as high sensitivity, high specificity. The number of cancer patients has
Nano-materials continued to rise in recent years, and the limitations of traditional cancer diagnosis methods can no longer meet
Cancer bio-macromolecular markers
the demand for accurate early cancer screening. Accurate identification of cancer bio-macromolecular markers
Fluorescent nano-biosensors
can significantly improve early diagnosis and follow-up treatment. To this end, we provide an overview of the
properties and uses of different nano-materials and detail the principles and functions of fluorescent nano-
biosensor detection for different cancer bio-macromolecular markers. In addition, the prospects and chal­
lenges of translating this convenient and visual diagnostic method into the clinical application are discussed.

1. Introduction bio-macromolecular markers. A wide variety of biosensors have been


developed based on different signaling methods, such as electrochemical
Cancer is abnormal cell division or uncontrolled growth in response [13], fluorescence [14], surface-enhanced Raman scattering (SERS)
to various tumorigenic factors. Cancer is a leading cause of death [15], surface plasmon resonance (SPR) [16]. Nanomaterial-based
worldwide, causing 10 million deaths in 2020 [1]. Despite the rapid biosensor technology relies on nano-materials with high specific sur­
development of modern cancer treatment technology, the survival rate face area, biocompatibility function, high stability, and other charac­
of patients is still low. Early diagnosis improves cancer outcomes, and teristics, showing high sensitivity, rapid response, accurate
early detection can significantly reduce cancer-related mortality [2,3]. identification, etc., which is widely used in biomarker sensing research
Currently, sample biopsy, tumor imaging, and cancer [17,18].
bio-macromolecular markers detection are the most common methods The fluorescent nano-biosensor takes the nano-material as a new
for cancer diagnostic [4–11]. However, current imaging and sample biosensing medium and uses the fluorescence signal as a detection
biopsies exist that are limited to identifying significant tissue areas with element to detect biomarkers. Cancer bio-macromolecular markers can
cancerous lesions. Cancer bio-macromolecular markers are molecules be proteins, glucose or nucleic acids secreted by the body or cancer cells
that change significantly during the cancer process (e.g., cancer in cancer development [19–23]. Recently, many novel nano-materials
cell-specific proteins, intracellular nucleic acids, enzymes and hor­ such as gold nano-particles (AuNPs), magnetic nano-particles, metal
mones). Therefore, precise and sensitive identification of cancer cells is nano-particles, quantum dots (QDs) and carbon nano-tubes (CTNs) have
essential for the early diagnosis of cancers [12]. Recently, with the de­ been used for cancer bio-macromolecular markers detection [24–26].
mand for low-cost, rapid, and sensitive detection of cancer Fluorescence-based nano-biosensors have many advantages in the

* Corresponding author.
** Corresponding author.
E-mail addresses: ou1020@mail.ustc.edu.cn, becsy123@163.com (N. Ouyang), huayu342@foxmail.com (Y. Yang), wenya.hou@szu.edu.cn (W. Hou).
1
These authors contributed equally to this work.

https://doi.org/10.1016/j.polymertesting.2022.107746
Received 13 April 2022; Received in revised form 8 August 2022; Accepted 12 August 2022
Available online 18 August 2022
0142-9418/© 2022 The Authors. Published by Elsevier Ltd. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-
nc-nd/4.0/).
N. Ouyang et al. Polymer Testing 115 (2022) 107746

Table 1
Comparison of biosensing properties of various nano-materials.
Materia Advantage Shortcoming Application References

Gold/Silver Nano- Variable optical properties, strong electrical High production cost, and low stability of Biosensing, Bioimaging [36,37]
particles conductivity, outstanding biocompatibility, biofunctionalized particles
silica nano-particles Large specific surface area, good biocompatibility, lack of autofluorescence Drug Delivery, and Biosensing [38]
high stability
CDs and GQDs High chemical stability, good electrical conductivity limited analytical options Bioimaging, Biosensing, and [39,40]
Genetic Diagnosis
Metal oxide nano- High catalytic efficiency, strong signal amplification, Toxic, poor analytical selectivity, poor Biosensing, Biomedical [41]
materials low cost biocompatibility Diagnosis and Therapy
Fluorescent Copper High efficiency, powerful molecular detection, low Fragile photostability, easy to oxidize, low Biosensing and Fluorescent [42,43]
Nano-materials biological toxicity biological tolerance Probes

detection of cancer bio-macromolecular markers (such as rapid response


Table 2
time and providing visual identification). In this review, we will sum­
Characteristics of an ideal fluorescent nano-biosensor.
marize the recent advances in fluorescent nano-biosensors targeting
Cancer bio-macromolecular markers. Due to the different features of Parameter Required characteristics
cancer bio-macromolecular markers such as PH, stability, concentration, Sensitivity Low detection limit (LOD) < 104 cfu mL- 1 or <10 nm
and the biological properties of nano-materials, detection strategies of Specificity Correctly distinguish between normal cells and cancer cells
Response time Quick Response Detection
various fluorescent nano-biosensors will be discussed in the following
Stability Stable in complex testing environments
sections. Different fluorescent nano-biosensors are highlighted in Biocompatibility Does not affect and adapts to the detection environment
sensitivity, stability, linear detection range, and detection limit. The
challenges and prospects of applying fluorescent nano-sensors will also
be discussed in this review. nano-materials (CuNMs), including copper nano-particles (CuNPs) and
copper nanoclusters (CuNCs), are widely used in biosensing and fluo­
2. Comparison of the potential of different nano-materials for rescent probe research owing to their abundant raw materials, easy
biosensing synthesis, low cost, and excellent biocompatibility. But the disadvan­
tages of CuNCs are short photostability and poor biotolerance [34]. In
Since the beginning of nanotechnology, nano-materials have recent years, 2D transition metal oxides materials attracted more and
received much attention in biology and medical research, including the more attention, which have many unique advantages, such as high
use of nano-materials for cancer bio-macromolecular markers detection. thermal stability and strong oxidation ability. 2D transition metal oxides
In general, the size of biomolecules (DNA or proteins) is usually in the materials have been widely used in the fields of electronics, energy
range of 2 – 100 nanometers, and biomolecules and nano-materials have technology, biosensing and biomedical diagnosis and treatment [35].
similar sizes. Furthermore, a critical advantage of nano-particles for Therefore, nano-materials have been developed for early cancer diag­
cancer detection is due to the high specific surface area of nano- nosis studies due to their good biosensing properties.
materials, which allows them to carry a wide range of biomolecules
with targeting and detection functions [27]. Stimulated by the demand 3. Fluorescent nano-biosensors developed for a variety of cancer
for early tumor diagnosis, various nano-particles with sensing potential bio-macromolecular markers
have been developed. As shown in Table 1, metal nano-particles, rep­
resented by gold or silver have the advantages of variable size, optical Early diagnosis of precancerous lesions can allow early cancer
properties and strong conductivity, among which the representative detection, reduce cancer mortality, and raise the chance of cure [12,44].
silver nanoclusters are widely used in biosensing, and the photo­ Nanotechnology offers high selectivity and sensitivity in cancer diag­
luminescence mechanism of silver nanoclusters is the coupling of sur­ nosis, and enables the simultaneous measurement of multiple targets.
face plasmon and emitter [28]. At the same time, Silica nano-particles Quantum dots (QDs) [45], gold nano-particles (AuNPs) [46] and poly­
have promising applications in the biomedical field owing to their large mer dots (PDs) are three standard nano-particle probes used to diagnose
specific surface area and high stability properties. Furthermore, multi­ cancer. Interestingly, recent studies have shown that nano-materials
functional silica nano-particles have been used in medical bioimaging combined with fluorescence sensing can accurately identify and target
and drug-targeted delivery studies [20]. In the past few years, cancer bio-macromolecular markers [47]. As shown in Table 2, bio­
carbon-based nano-materials have become the most widely used bio­ sensors combining fluorescence and nanotechnology have various
materials due to their unique optical, electronic, mechanical and properties, such as high stability, high specificity, strong sensitivity, fast
chemical properties [21]. For example, carbon dots (CDs), which are response speed and good biocompatibility.
quasi-spherical oxygen-containing carbon nanoparticles with a particle
size of less than 10 nm, which PL mechanisms are now generally 3.1. Protein detection
accepted: carbon core state luminescence from conjugated π-domain
with quantum confinement effect, surface state luminescence from the The FDA has approved many proteins for cancer detection, including
interaction of surface groups and carbon core and crosslink enhanced CEA (carcinoembryonic protein), PSA (prostate cancer), CA-125
emission effect, etc. [29,30]. Besides, graphene quantum dots (GQDs) (ovarian cancer) and others [48,49]. Furthermore, intracellular
are also car-bon-based nanoscale particles, which are emissive graphene reducing substances (IRS) have been used as a new indicator to differ­
nanosheets and less than 100 nm, and the main PL mechanisms of GQDs entiate cancer cells/normal cells [50]. In humans, GSH (a kind of IRS)
include the quantum confinement effect of conjugated π-domains, the concentration is about four times higher in cancer cells than in normal
surface/edge states of GQDs, and the synergistic effect of these two cells [51,52]. Therefore, the accurate and specific identification of
factors [31]. They exhibit the similar outstanding properties, including “signals” released by cancer cells is significant for cancer patients’ early
low toxicity, biocompatibility, photostability, water solubility, and diagnosis and treatment.
simplicity of preparation. However, carbon-based quantum dots’ low
quantum yield (QY) limits their pertinency [32,33]. Fluorescent copper

2
N. Ouyang et al. Polymer Testing 115 (2022) 107746

Fig. 1. Schematic diagram of PDAN@AgNC based fluorescent nano-biosensor for detection of CAE and AFP.
It is adapted from Ref. [57] with permission to reproduce from RSC.

Fig. 2. Schematic illustration of the paper assay device (PAD): CEA is recognized by the modified MSN, and a specific reaction occurs resulting in the release of
glucose, and GOD then decomposes the glucose to produce hydrogen peroxide which quenches the QDs fluorescence emission.
It is adapted from Ref. [59] with permission to reproduce from ACS.

3.1.1. CEA detection A study by Qiu et al. successfully developed an all-in-one paper-
CEA is a 180 kDa glycoprotein, which is considered as a cancer based analysis de vice (PAD) to detect CEA [59]. The assay was carried
marker for multiple cancers, such as breast cancer, lung cancer, colon out in a centrifuge tube using glucose-loaded mesoporous silica nano­
cancer and ovarian cancer. Thus, CEA is considered a valid indicator for containers (MSNs) with a CEA aptamer and a “Quantum
assessing response to cancer treatment [53–56]. Chen et al. designed a Dot-Enzyme-Impregnated Paper” attached to the lid. After adding CEA,
specific aptamer to detect CEA by binding to dsDNA-templated copper the analyte reacted specifically with aptamers immobilized on MSN, and
nano-particles (CuNPs) [57]. When the aptamer recognizes CEA, it then glucose was released. The immobilized Glucose oxidase (GOD)
prevents dsDNA from binding to Cu2+, which could quench the strong oxidized the released glucose on paper to produce hydrogen peroxide,
fluorescence emission of double-stranded DNA template copper which could quench the fluorescence of CdTe/CdSe QDs. The PAD-based
nano-particles. This sensor is susceptible to CEA detection, and the limit sensing system sensitively discriminates target CEA from other bio­
of detection (LOD) is 0.0065 ng mL− 1. In addition, jiang et al. proposed markers or proteins, and this device provides sensitive detection of CEA
an innovative device based on a polydopamine nanosphere@silver over a linear range of 0.05–20 ng mL− 1 with detection limits as low as
nanocluster (PDAN@AgNC) system for the accurate detection of CEA. 6.7 pg mL− 1 (Fig. 2).
Briefly, in the presence of CEA, AgNCs is released from the PDAN surface
as a complex, resulting in the recovery of previously quenched fluores­ 3.1.2. PSA detection
cence (Fig. 1). In addition, the device could also detect α-fetoprotei­ In prostate cancer progression, accurate assessment of prostate-
n-AFP (a liver cancer marker), and the detection limits for AFP and CEA specific antigen (PSA) levels is beneficial for cancer diagnosis [60].
were 2.4 nM and 5.6 nM, respectively [58]. This multi-target detection Early detection of prostate cancer can effectively improve a patient’s
technique shows great potential for the detection of cancer markers in chance of cure [61]. Therefore, Huang et al. designed a fluorescent
human serum samples. biosensor(Metal-organic frameworks(MOF)@ gold nano-particles

3
N. Ouyang et al. Polymer Testing 115 (2022) 107746

Fig. 3. (A) Synthesis method of MOF@AuNP@GO; (B) Schematic diagram of MOF@AuNP@GO fluorescent nano-sensor for detecting target proteins.
It is adapted from Ref. [62] with permission to reproduce from Theranostics.

(AuNP)@ graphene oxide(GO))based on a nanohybrid system [62]. The


designed nanohybrid exhibited an enhanced ssDNA binding affinity and
fluorescence quenching ability, and it used specific dye-labeled aptamer
probes to recognize prostate-specific antigens (Fig. 3). This nano-sensor
can sensitively measure PSA with a detection limit of 0.01 ng mL− 1,
which can quickly and sensitively measure cancer bio-macromolecular
markers. In another rapid detection research, Choi et al. proposed a
novel fluorescent nano-sensor based on “fluorescein isothiocyanate
(FITC)/peptide bound gold (Au) nano-particle complex (FPAN)", and it
relied on specific enzyme digestion reactions to detect PSA [63]. This
simple process of constructing the FPAN probe was superior to other
sensing systems because it reduced the possibility of probe defects in the
sensing process and diagnoses prostate cancer by measuring the fluo­
rescence signal. It could detect PSA concentrations ranging from 10 pM
to 100 nM.
Up to 30% of patients with prostate cancer have metastatic recur­
rence after treatment, so urokinase fibrinogen activator (uPA) as a
marker for metastatic prostate cancer, its effective diagnosis is particu­
larly important [64]. By using the unique fluorescent properties of
single-walled carbon nanotubes (SWCNTs), Williams et al. developed a
quantitative fluorescent nano-sensor to detect uPA in serum samples
[65]. The nano-sensor complexes were synthesized from single-walled
carbon nanotubes encapsulated with single-stranded DNA (TAT) 6
coupled with monoclonal anti-uPA antibodies (Fig. 4). The sensor could
quantitatively detect uPA concentrations in human blood products with
a detection limit of up to 50 nm.

3.1.3. GSH detection


Cancer cells have higher levels of reactive oxygen species and
therefore have higher concentrations of reducing substances to
Fig. 4. Synthesis scheme of Anti-uPA-DNA-SWCNT complexes.
neutralize reactive oxygen species, of which reducing glutathione (GSH)
It is adapted from Ref. [65] with permission to reproduce from ACS.
is one of the most abundant intracellular reducing substances, and GSH
is upregulated in many cancers such as gastric, colorectal and breast
cancers [66,67]. In order to perform cancer cell identification easily, Li

4
N. Ouyang et al. Polymer Testing 115 (2022) 107746

Fig. 5. Synthesis scheme of GQDs-MnO2 nanocomposites and the principle of GSH-stimulated fluorescence turn-on.
It is adapted from Ref. [69] with permission to reproduce from Elsevier.

Fig. 6. Schematic diagram of targeting TrxR identification and detection.


It is adapted from Ref. [74] with permission to reproduce from ACS.

et al. proposed a facile fluorescent biosensor based on silicon CPs@AgNPs [69,70]. The introduction of glutathione (GSH) decom­
nano-particles (SiNP) [68]. Chelating Fe3+ ions quenched the fluores­ posed MnO2 and caused the fluorescence recovery of GQDs. Therefore,
cence of SiNPs, and after redox reactions with reducing species, the GSH overexpressed in cancer cells could be accurately identified, and an
nano-sensors showed sensitive fluorescence recovery. The sensor relied acceptable linear detection range for GSH was 0.07–70 μM and the LOD
on efficient cellular uptake and could detect IRSs by fluorescence im­ was 48 nM (Fig. 5). However, a study by Wong et al. fabricated a novel
aging. The LOD was evaluated to be 352 nM and 16.7 μM. Under the fluorescent sensing platform based on BSA/AuNCs. Which showed
demand for high-precision identification of cancer cells, Wang et al. excellent sensing performance for GSH detection after its functionali­
constructed two novel sensors for intracellular GSH detection using zation with graphene oxide and folic acid [71]. And the specific per­
composite nano-materials composed of MnO2 and GQDs and formance as follows: its sensitive and extremetly rapid detection of GSH

5
N. Ouyang et al. Polymer Testing 115 (2022) 107746

Fig. 7. (A)The schematic of FRET-based MMP-2 detection method using QDs–peptide–dye nano-assembly; (B) Schematic diagram of the synthesis and detection
principle of QD-FRET nano-sensor.
It is adapted from Refs. [76,77] with permission to reproduce from Elsevier and ACS.

and with a detection limit down to 0.1 μM. [72,73]. Sidhu et al. proposed a ratiometric fluorescent nano-sensor,
“Biotin-CDs-Naphthalimide” to target TrxR in cancer cells [74]. The
3.1.4. TrxR detection schematic (Fig. 6) shows that Nano-sensors could precisely enter cancer
High expression of thioredoxin reductase (TrxR) has been observed cells via biotin receptor-mediated endocytosis. Because of the fluores­
in many cancers (e.g. colorectal cancer, lung cancer, prostate cancer). cence resonance energy transfer (FRET) between carbon dots (CDs) and
TrxR is an attractive pharmacological target for tumor radiosensitization naphthalimide, the fluorescence of the sensor was turned on once the

Fig. 8. Schematic design and activation mechanism of a fluorescence quenching biosensor based on sol-gel matrix film wrapped with Au nano-particles.
It is adapted from Ref. [79] with permission to reproduce from ACS.

6
N. Ouyang et al. Polymer Testing 115 (2022) 107746

Fig. 9. Schematic diagram of GQD-PEG-aptamer/MoS2-based FRET biosensor to detect EpCAM protein.


It is adapted from Ref. [82] with permission to reproduce from Elsevier.

Fig. 10. (A)Schematic diagram of synthesis of AgNCs (DNA/AgNCs) fluorescent probe sensor with DNA template; (B) Schematic diagram of fluorescent complex
nanoprobe to detect BRCA1 gene deletion.
It is adapted from Ref. [86]with permission to reproduce from Elsevier.

7
N. Ouyang et al. Polymer Testing 115 (2022) 107746

Fig. 11. Schematic diagram of logic gates based on AgNCs and HCR to recognize miRNA-21 and telomerase.
It is adapted from Ref. [88] with permission to reproduce from RSC.

overexpression of thioredoxin reductase (TrxR) was detected in cancer differentiate patient samples(Fig. 8).
cells, and this device could sensitively detect TrxR at concentrations
down to 7.2 × 10− 8 M. 3.1.7. EpCAM detection
The epithelial cell adhesion molecule (EpCAM) is a well-known
3.1.5. MMP-2 detection marker which is highly expressed in multiple cancers and implies a
Matrix metalloproteinase 2 (MMP-2) is a protease associated with poor prognosis [80]. Tao et al. designed an EpCAM antibody-dependent
tumor invasion and metastasis [75]. Li et al. constructed a novel fluo­ device to detect EpCAM overexpression in colon cancer cells [81], which
rescent sensing system utilizing the combination of quantum dots and presented imaging of colon cancer cells by combining anti-EpCAM an­
organic dyes to detect MMP-2 in vivo and in vitro [76]. When these probes tibodies with Rubpy-encapsulated core-shell silica nano-particles, and
are exposed to MMP-2, the selective cleavage of the peptide result in the showed distribution and abundance of EpCAM in cell membranes. In
recovery of fluorescence from QDs, by using the produced another meaningful work, Shi et al. reported a new GQD-PEG-Apta­
540QD–peptide–RB and 720QD–peptide–MPA probes, and the detection mer/MoS2 based FRET assay for the detection of EpCAM protein with
limit is reduced to 0.25 nM (Fig. 7A). In addition, Ochs et al. proposed a GQD and MoS2 nanosheets as a donor and quencher, respectively [82].
quantum dot-based fluorescent nano-sensor using the exact FRET The sensing platform immobilized the GQD-labeled EpCAM aptamer on
mechanism, and the device was used to detect matrix metalloproteinases the MoS2 surface. Once EpCAM exists, it could bind with the EpCAM
(MT1-MMP) on cell membranes [77]. As shown in the figure (Fig. 7B), aptamer properties so that the EpCAM aptamer could be detached from
the sensor could recognize activated MT1-MMP in cancer cells, and the the MoS2 nanosheet, which eventually led to the recovery of the fluo­
specific cleavage reaction interrupted the FRET between Cy3 peptide rescence intensity (Fig. 9). By monitoring the change in fluorescence
and QD. Thus, the fluorescence emission of QD was restored. Their signal, the target EpCAM protein could be detected sensitively and
studies can sensitively detect MMP-2 in vitro and in vivo, providing a selectively with a linear detection range from 3 nM to 54 nM, and the
diagnostic strategy for accurately identifying cancer cells. LOD was as low as 450 pM.

3.1.6. CA-125 detection


Cancer antigen 125 (CA-125) is an essential and significant 3.2. Nucleic acid detection
biomarker in ovarian malignancies [78]. Mona et al. designed a simple
fluorescence-quenched biosensor based on a gold-coated to detect 3.2.1. Genetic testing of BRCA1
oncomarker CA-125 [79]. The fabricated sensor consisted of a nano-gold Breast cancer susceptibility gene 1(BRCA1) is a tumor suppressor,
thin film doped into a sol-gel matrix. The quantification of CA-125 was and mutations in the BRCA1 gene predispose to breast and ovarian
based on the quenching ability of this fluorescence signal, with a cancer, and it can be a target for breast cancer prevention and treatment
sensitivity of 97.35%, a specificity of 94.29%, and the LOD was 1.45 U [83,84]. Borghei et al. developed a label-free detection strategy for
mL− 1. In particular, the developed biosensor could successfully significant deletion mutation in the BRCA1 gene [85]. According to the
principle of DNA-induced hybridization, the DNA-silver nanocluster

8
N. Ouyang et al. Polymer Testing 115 (2022) 107746

Fig. 12. (A) Synthesis and fluorescence emission principle of DNA/AgNCs; (B) Fluorescence detection strategy of miRNA-378.
It is adapted from Ref. [92] with permission to reproduce from Elsevier.

(NC) fluorescent probe could recognize the mutation DNA fragment of (AgNCs)”, a device that relies on fluorescent logic gates for signal
the BRCA1 target gene, resulting in higher fluorescence. Using this switching to directly reflect information about intracellular miRNA-21
nano-biosensor, the significant deletion mutation in the BRCA1 gene and telomerase (a ribonucleoprotein complex upregulated in most
could be detected with a LOD of 6.4 × 10 − 11 M (Fig. 10). Subsequently, human cancers) [90]. Briefly, in the presence of miRNA-21 and telo­
another new FRET biosensor based on the interaction of DNA and CdTe merase, disruption of the logic gate of this sensor device released DNA
quantum dots was developed [86]. Hybridization between deletional leading to the resumption of fluorescence from graphene oxide
BRCA1 targets and probed restores nanocluster and quantum dot fluo­ (GO)-quenched AgNCs, and the LOD for miRNA-21 was 2.8 pM (Fig. 11).
rescence emission. This method was sensitive to detecting the deletion of This device provides a simple, fast and reliable method for miRNA-21
the BRCA1 target gene, and its linear detection range was 0.5pM–1 nM and telomerase related research and diagnosis. These types of fluores­
(Fig. 10B). cent nanobiosensors are of great significance in human disease diagnosis
and anti-tumor therapy.
3.2.2. MiRNA testing MiRNA-378 in serum can be used as a new potential non-invasive
MiRNAs are short RNA sequences that regulate gene expression. biomarker in gastric cancer detection [91]. Liu et al. developed a sus­
MiRNA-21 is upregulated in almost all solid tumors of epithelial cell ceptible method for microRNA-378 detection, using DNA-templated
origin, and most of its reported targets are tumor suppressors [87]. silver nanoclusters as label-free fluorescent probes [92].
MiRNA-21 is an essential biomarker for early prediction and tumor MicroRNA-378 bonded to the cDNA complementary sequence that
metastasis, and its accurate detection often provides the best chance for triggers rolling circle amplification, thus realizing the “fluorescence
a cure [88]. Morteza et al. developed a fluorescent biosensor based on enhancement” phenomenon (Fig. 12). This sensor has low cost, high
DNA-AuNCs nano-materials, which was used to detect of miRNA-21 in specificity, and a detection limit of 1.07 fM in vitro serum samples.
human plasma samples, and the LOD of this method was 0.7 pM [89].
Gong et al. designed a multifunctional nanocomposite based on gold
nanorods (AuNR) for miRNA-21 detection and cancer therapy [88]. 3.3. Tumor-derived exosomes detection
Briefly, the device used functional DNA strands to modify the early
synthesized AuNRs, and carried doxorubicin (DOX) and folic acid. Once Exosomes are a class of small membrane vesicles of small molecular
microRNA-21 was detected to bind to double-stranded DNA, it would complexes that are commonly released into the tumor microenviron­
trigger drug release and cause fluorescence recovery. It also showed ment and can also be detected in body fluids. There is evidence that
high selectivity for the microRNA-21 assay with a LOD of 2.1 nM. Driven exosomes promote tumorigenesis by regulating angiogenesis, immunity.
by the need for multifunctional analysis, Jiang et al. proposed a novel Exosomes are viewed as a non-invasive biomarker for early clinical
fluorescent nano-sensor based on “DNA-templated silver nanoclusters cancer diagnosis [93,94]. Driven by the need for real-time in vitro cancer
diagnosis, Li et al. developed a homogenous magneto-fluorescent

9
N. Ouyang et al. Polymer Testing 115 (2022) 107746

Fig. 13. Schematic diagram of the hMFEX nano-sensor. (a) Tumor-derived exosomes are captured by magnetic beads with specific antibodies to form a magnetic
bead-exosome-adaptor complex. (b) Captured exosomes are detected by aptamer-triggered DNA three-way junctions cyclic assembly strategy along with TPE-TA and
the GO-based “turn-on” fluorescent system.
It is adapted from Ref. [95] with permission to reproduce from ACS.

exosome (hMFEX) nano-sensor for rapid and sensitive detection of surface-enhanced Raman spectroscopy (SERS) sensor using highly sen­
tumor-derived exosomes [95]. This hMFEX nano-sensor used specific sitive gold nanorods (AuNR) material [96]. Three specific aptamers
antibodies to recognize and bind to exosomes, followed by an modified on the surface of AuNRs were able to capture breast cancer
aptamer-triggered DNA three-way junctions cyclic assembly strategy to cell-derived exosomes into the sensing array, which will exhibit an
turn on the quenched fluorescence of the tert-amine tetraphenylethylene enhanced SERS signal. This sensor device is highly sensitive and ho­
(TPE-TA) and GO complexes (Fig. 13). This device detects exosomes of mogeneous, enabling a wide detection range (1.0 × 104–5.0 × 106
tumor origin with high specificity, and the limit of detection is 6.56 × particles/mL) and low detection limit (5.3 × 103 particles/mL). The
104 particles/μL. In addition, Wang et al. constructed a development of these types of highly sensitive fluorescent nano-sensors

10
N. Ouyang et al. Polymer Testing 115 (2022) 107746

Fig. 14. (A) Synthesis and detection strategy of AgNPs@GQDs-GOD fluorescent nano-sensor; (B) Schematic diagram of constructing a dual-function nano platform
for detection and treatment.
It is adapted from Ref. [99] with permission to reproduce from Elsevier.

has excellent prospects for clinical applications, enabling real-time particles, carbon dots, quantum dots, etc.) and also describes the prop­
diagnosis of early cancer-derived exosomes. erties, functions and fabrication methods of fluorescent nanobiosensors
for the detection of different cancer bio-macromolecular markers.
Currently, most fluorescent nanobiosensors are investigated for in vitro
3.4. Glucose detection cancer marker detection (e.g., blood and urine tests) and do not yet meet
the needs for in vivo biosensing and combination of drugs for targeted
Cancer cells have a higher glucose uptake rate than normal cells therapy. In addition, cancer cell-based biomarkers often involve multi­
[97]. Glucose is also an essential biomarker for cancer diagnosis [21, ple complex physiological processes. Therefore, another major chal­
98]. Targeting and identifying highly expressed glucose in cancer cells lenge is the need to design multi-targeted and more accurate fluorescent
can facilitate early diagnosis and promote the development of precision nano-sensors to identify cancer cells with high complexity.
therapy. Therefore, Hai et al. developed a dual-mode glucose fluorescent On the other hand, the optical imaging quality of most short-wave
biosensor based on AgNPs@GQDs-GOD nano-materials for cancer cell emitting nanomaterials is far from adequate for high-quality in vivo
identification [99]. This novel sensor targets cancer cell detection fluorescence sensing, while long-wave emission is ideal for applications
combined with targeted therapy. Due to high glucose levels in the tumor such as in vivo bioimaging, photothermal and photodynamic therapies
microenvironment, the nanocomposites undergo glucose-related redox [101]. Currently, there are a number of research teams working on the
reactions, ultimately leading to fluorescence recovery of GQDs. This emission red shift of nanomaterials (such as carbon dots). By choosing
device could be susceptible to identifying cancer cells with high glucose appropriate C source or reaction medium, changing reaction conditions
concentrations, and the detection range of glucose levels was from 10 or other treatments, high fluorescence CDs with long wavelength
μM to 4 mM, and its LOD was 4.78 μM. Furthermore, it could also induce emission can be obtained [102], however, its quantum yield (QY) needs
cancer cell apoptosis in combination with starvation-like therapy, metal to be improved. Therefore, suitable precursors, innovative synthesis
ion therapy, and Tannic acid (TA) (Fig. 14A and B). In another report on methods and appropriate purification methods or modeling with the
the diagnosis and treatment of cancer cells, Chen et al. developed a help of machine learning should be sought for the synthesis of CDs with
smart nanoprobe that can accurately identify cancer cells with high more desirable properties (e.g. deep tissue penetration, low photo­
glucose levels, and under NIR laser irradiation adjuvant therapy, it could damage and high-resolution fluorescence imaging properties). Facilitate
kill cancer cells well [100]. Therefore, these two sensors are promising the emission red shift and high QY of nanomaterials such as CDs, which
for potential practical diagnostic and therapeutic applications. will greatly promote the development of in vivo fluorescence sensing
[103–105]. At the same time, it is also worth paying attention to the
4. Conclusions and future prospects photoluminescence (PL) mechanisms of some of the most promising
nano-materials, such as carbon dots, Due to the structural and compo­
For decades, the total number of cancer patients and mortality rates sitional diversity and complexity of CDs, their photoluminescence
have continued to increase, despite some successes in treating cancer mechanism is still controversial and is likely to be highly dependent on
patients, and partially is due to the slow progress of new technologies for particle size, surface functional groups, degree of graphitization, het­
early cancer diagnosis. This review describes recent researches on eroatom doping, etc., and these photoluminescence origins should be
fluorescence sensing of cancer bio-macromolecular markers using nano- understood in depth, which will be important for the synthesis of
materials. It summarizes the development of fluorescent biosensors nano-materials with high biocompatibility, ultra-high resolution sensing
based on different nano-materials (including gold/silver/silica nano-

11
N. Ouyang et al. Polymer Testing 115 (2022) 107746

as well as imaging and application for early diagnosis of cancer bio­ [7] G. Mankaney, R.A. Sutton, C.A. Burke, Colorectal cancer screening: choosing the
right test, Cleve. Clin. J. Med. 86 (2019) 385–392.
molecules in complex in vivo environments.
[8] S.B. Fisher, D.A. Kooby, Laparoscopic pancreatectomy for malignancy, J. Surg.
In fact, for now, most of the fluorescent nano-sensors for accurate Oncol. 107 (2013) 39–50.
detection of cancer bio-macromolecular markers are still in the experi­ [9] C. Bohon, Cancer recognition and screening for common breast disorders and
mental stage, and the need for rigorous validation hampers the appli­ malignancy, Obstet. Gynecol. Clin. N. Am. 44 (2017) 257–270.
[10] A. Testa, E. Venturelli, M.F. Brizzi, Extracellular vesicles: new tools for early
cation of biosensor-based models to clinical application. There is a need diagnosis of breast and genitourinary cancers, Int. J. Mol. Sci. 22 (2021).
to consider better targeting effects, sensitivity, detection cost, and real- [11] Y. Peng, C. Shi, X. Wu, Y. Zhu, S. Zhuang, Terahertz imaging and spectroscopy in
time efficiency. Finally, we note that fluorescent nano-biosensors can cancer diagnostics: a technical review, BME Frontiers 2020 (2020) 1–11.
[12] J.D. Schiffman, P.G. Fisher, P. Gibbs, Early Detection of Cancer: Past, Present, and
precisely target and detect nucleic acid molecules. Which may have Future, Am Soc Clin Oncol Educ Book, 2015, pp. 57–65.
potential applications for the diagnosis of cancers that viral infections [13] R. Huang, L. He, Y. Xia, H. Xu, C. Liu, H. Xie, S. Wang, L. Peng, Y. Liu, Y. Liu,
may induce (e.g. hepatitis C virus (HCV) induced liver cancer, human N. He, Z. Li, A sensitive aptasensor based on a hemin/G-quadruplex-assisted
signal amplification strategy for electrochemical detection of gastric cancer
papillomavirus (HPV) induced cervical cancer, etc.) or nucleic acid exosomes, Small 15 (2019), e1900735.
molecules of the new coronavirus. [14] Y. Zhang, D. Hou, Z. Wang, N. Cai, C. Au, Nanomaterial-based dual-emission
ratiometric fluorescent sensors for biosensing and cell imaging, Polymers 13
(2021).
Author contributions [15] N. Choi, H. Dang, A. Das, M.S. Sim, I.Y. Chung, J. Choo, SERS biosensors for
ultrasensitive detection of multiple biomarkers expressed in cancer cells, Biosens.
Writing - original draft, data curation, Nan Ouyang; data collection Bioelectron. 164 (2020), 112326.
[16] M. Piliarik, H. Vaisocherova, J. Homola, Surface plasmon resonance biosensing,
and curation, Lei Hong; investigation, review, Yuanshuai Zhou; re­ Methods Mol. Biol. 503 (2009) 65–88.
sources, supervision, Jinzhong Zhang and Ying Yang; writing – review, [17] Y.J. Teh, A. Bahari Jambek, U. Hashim, A study of nano-biosensors and their
Shaheryar Shafi; investigation, Software, Jinlin Pan and Rongchuan output amplitude analysis algorithms, J. Med. Eng. Technol. 41 (2017) 72–80.
[18] C. Jianrong, M. Yuqing, H. Nongyue, W. Xiaohua, L. Sijiao, Nanotechnology and
Zhao; writing - review & editing, Wenya Hou. biosensors, Biotechnol. Adv. 22 (2004) 505–518.
[19] L. Korecka, K. Vytras, Z. Bilkova, Immunosensors in early cancer diagnostics:
Funding from individual to multiple biomarker assays, Curr. Med. Chem. 25 (2018)
3973–3987.
[20] C.A. Borrebaeck, Precision diagnostics: moving towards protein biomarker
This research was funded by the National Natural Science Founda­ signatures of clinical utility in cancer, Nat. Rev. Cancer 17 (2017) 199–204.
tion of China (Grant No. 32101039), Natural science foundation of [21] K. Ueda, Glycoproteomic strategies: from discovery to clinical application of
cancer carbohydrate biomarkers, Proteonomics Clin. Appl. 7 (2013) 607–617.
Guangdong province of China (2022A1515011208), the Natural Science [22] A.H. Ren, C.A. Fiala, E.P. Diamandis, V. Kulasingam, Pitfalls in cancer biomarker
Foundation of Tianjin (Grant No. 17JCYBJC43400), and the Major discovery and validation with emphasis on circulating tumor DNA, Cancer
Innovative Research Team of Suzhou (Grant No. ZXT2019007). Epidemiol. Biomarkers Prev. 29 (2020) 2568–2574.
[23] H. Ma, J. Liu, M.M. Ali, M.A. Mahmood, L. Labanieh, M. Lu, S.M. Iqbal, Q. Zhang,
W. Zhao, Y. Wan, Nucleic acid aptamers in cancer research, diagnosis and
Institutional review board statement therapy, Chem. Soc. Rev. 44 (2015) 1240–1256.
[24] S. Xiao, J. Lu, L. Sun, S. An, A simple and sensitive AuNPs-based colorimetric
aptasensor for specific detection of azlocillin, Spectrochim. Acta Mol. Biomol.
Not applicable. Spectrosc. 271 (2022), 120924.
[25] U.D. Kamaci, M. Kamaci, Selective and sensitive ZnO quantum dots based
fluorescent biosensor for detection of cysteine, J. Fluoresc. 31 (2021) 401–414.
Informed consent statement
[26] Y. Plekhanova, S. Tarasov, A. Bykov, N. Prisyazhnaya, V. Kolesov, V. Sigaev, M.
A. Signore, A. Reshetilov, Multiwalled carbon nanotubes and the electrocatalytic
Not applicable. activity of gluconobacter oxydans as the basis of a biosensor, Biosensors 9 (2019).
[27] S. Song, Y. Qin, Y. He, Q. Huang, C. Fan, H.Y. Chen, Functional nanoprobes for
ultrasensitive detection of biomolecules, Chem. Soc. Rev. 39 (2010) 4234–4243.
[28] X. Liu, R. Hu, Z. Gao, N. Shao, Photoluminescence mechanism of DNA-templated
Declaration of competing interest
silver nanoclusters: coupling between surface plasmon and emitter and sensing of
lysozyme, Langmuir 31 (2015) 5859–5867.
The authors declare that they have no known competing financial [29] C. Xia, S. Zhu, T. Feng, M. Yang, B. Yang, Evolution and synthesis of carbon dots:
from carbon dots to carbonized polymer dots, Adv. Sci. 6 (2019), 1901316.
interests or personal relationships that could have appeared to influence
[30] P. Miao, K. Han, Y. Tang, B. Wang, T. Lin, W. Cheng, Recent advances in carbon
the work reported in this paper. nanodots: synthesis, properties and biomedical applications, Nanoscale 7 (2015)
1586–1595.
Data availability [31] S. Zhu, Y. Song, J. Wang, H. Wan, Y. Zhang, Y. Ning, B. Yang, Photoluminescence
mechanism in graphene quantum dots: quantum confinement effect and surface/
edge state, Nano Today 13 (2017) 10–14.
Data will be made available on request. [32] L. Ansari, S. Hallaj, T. Hallaj, M. Amjadi, Doped-carbon dots: recent advances in
their biosensing, bioimaging and therapy applications, Colloids Surf. B
Biointerfaces 203 (2021), 111743.
Acknowledgments [33] S. Chung, R.A. Revia, M. Zhang, Graphene quantum dots and their applications in
bioimaging, biosensing, and therapy, Adv. Mater. 33 (2021), e1904362.
[34] Q. Cao, J. Li, E. Wang, Recent advances in the synthesis and application of copper
Thanks to Dr. Jiang for Figs. 1 and 11 citation permission.
nanomaterials based on various DNA scaffolds, Biosens. Bioelectron. 132 (2019)
333–342.
References [35] Y. Jia, X. Yi, Z. Li, L. Zhang, B. Yu, J. Zhang, X. Wang, X. Jia, Recent advance in
biosensing applications based on two-dimensional transition metal oxide
nanomaterials, Talanta 219 (2020), 121308.
[1] H. Sung, J. Ferlay, R.L. Siegel, M. Laversanne, I. Soerjomataram, A. Jemal,
[36] E. Boisselier, D. Astruc, Gold nanoparticles in nanomedicine: preparations,
F. Bray, Global cancer statistics 2020: GLOBOCAN estimates of incidence and
imaging, diagnostics, therapies and toxicity, Chem. Soc. Rev. 38 (2009)
mortality worldwide for 36 cancers in 185 countries, CA A Cancer J. Clin. 71
1759–1782.
(2021) 209–249.
[37] N. Ibrahim, N.D. Jamaluddin, L.L. Tan, N.Y. Mohd Yusof, A review on the
[2] R.M. Mann, R. Hooley, R.G. Barr, L. Moy, Novel approaches to screening for
development of gold and silver nanoparticles-based biosensor as a detection
breast cancer, Radiology 297 (2020) 266–285.
strategy of emerging and pathogenic RNA virus, Sensors (2021) 21.
[3] J. Matsuzaki, H. Tsugawa, H. Suzuki, Precision medicine approaches to prevent
[38] Y. Wang, Q. Zhao, N. Han, L. Bai, J. Li, J. Liu, E. Che, L. Hu, Q. Zhang, T. Jiang,
gastric cancer, Gut Liver 15 (2021) 3–12.
S. Wang, Mesoporous silica nanoparticles in drug delivery and biomedical
[4] M. Shetty, Imaging and differential diagnosis of ovarian cancer, Semin.
applications, Nanomedicine 11 (2015) 313–327.
Ultrasound CT MR 40 (2019) 302–318.
[39] A. Alaghmandfard, O. Sedighi, N. Tabatabaei Rezaei, A.A. Abedini, A. Malek
[5] A. Mahajan, A. Ahuja, N. Sable, H.E. Stambuk, Imaging in oral cancers: a
Khachatourian, M.S. Toprak, A. Seifalian, Recent advances in the modification of
comprehensive review, Oral Oncol. 104 (2020), 104658.
carbon-based quantum dots for biomedical applications, Mater Sci Eng C Mater
[6] E.D. Cook, A.C. Nelson, Prostate cancer screening, Curr. Oncol. Rep. 13 (2011)
Biol Appl 120 (2021), 111756.
57–62.

12
N. Ouyang et al. Polymer Testing 115 (2022) 107746

[40] W. Cheng, J. Xu, Z. Guo, D. Yang, X. Chen, W. Yan, P. Miao, Hydrothermal [70] Q. Wang, C. Wang, X. Wang, Y. Zhang, Y. Wu, C. Dong, S. Shuang, Construction of
synthesis of N,S co-doped carbon nanodots for highly selective detection of living CPs@MnO2-AgNPs as a multifunctional nanosensor for glutathione sensing and
cancer cells, J. Mater. Chem. B 6 (2018) 5775–5780. cancer theranostics, Nanoscale 11 (2019) 18845–18853.
[41] X. He, W.G. Aker, M.J. Huang, J.D. Watts, H.M. Hwang, Metal oxide [71] X.Y. Wong, D. Quesada-Gonzalez, S. Manickam, S.Y. New, K. Muthoosamy,
nanomaterials in nanomedicine: applications in photodynamic therapy and A. Merkoci, Integrating gold nanoclusters, folic acid and reduced graphene oxide
potential toxicity, Curr. Top. Med. Chem. 15 (2015) 1887–1900. for nanosensing of glutathione based on "turn-off" fluorescence, Sci. Rep. 11
[42] Z. Zhao, Y. Li, Developing fluorescent copper nanoclusters: synthesis, properties, (2021) 2375.
and applications, Colloids Surf. B Biointerfaces 195 (2020), 111244. [72] R.S. Patwardhan, D. Sharma, S.K. Sandur, Thioredoxin reductase: an emerging
[43] J. Mu, Y. Peng, Z. Shi, D. Zhang, Q. Jia, Copper nanocluster composites for pharmacologic target for radiosensitization of cancer, Transl Oncol 17 (2022),
analytical (bio)-sensing and imaging: a review, Mikrochim. Acta 188 (2021) 384. 101341.
[44] S. Abati, C. Bramati, S. Bondi, A. Lissoni, M. Trimarchi, Oral cancer and [73] F. Mohammadi, A. Soltani, A. Ghahremanloo, H. Javid, S.I. Hashemy, The
precancer: a narrative review on the relevance of early diagnosis, Int. J. Environ. thioredoxin system and cancer therapy: a review, Cancer Chemother. Pharmacol.
Res. Publ. Health 17 (2020). 84 (2019) 925–935.
[45] Z. Zhu, Y. Zhai, Z. Li, P. Zhu, S. Mao, C. Zhu, D. Du, L.A. Belfiore, J. Tang, Y. Lin, [74] J.S. Sidhu, A. Singh, N. Garg, N. Singh, Carbon dot based, naphthalimide coupled
Red carbon dots: optical property regulations and applications, Mater. Today 30 FRET pair for highly selective ratiometric detection of thioredoxin reductase and
(2019) 52–79. cancer screening, ACS Appl. Mater. Interfaces 9 (2017) 25847–25856.
[46] N. Bhatt, P.J. Huang, N. Dave, J. Liu, Dissociation and degradation of thiol- [75] A. Jezierska, T. Motyl, Matrix metalloproteinase-2 involvement in breast cancer
modified DNA on gold nanoparticles in aqueous and organic solvents, Langmuir progression: a mini-review, Med. Sci. Mon. Int. Med. J. Exp. Clin. Res. 15 (2009)
27 (2011) 6132–6137. RA32–40.
[47] U. Laraib, S. Sargazi, A. Rahdar, M. Khatami, S. Pandey, Nanotechnology-based [76] X. Li, D. Deng, J. Xue, L. Qu, S. Achilefu, Y. Gu, Quantum dots based molecular
approaches for effective detection of tumor markers: a comprehensive state-of- beacons for in vitro and in vivo detection of MMP-2 on tumor, Biosens.
the-art review, Int. J. Biol. Macromol. 195 (2022) 356–383. Bioelectron. 61 (2014) 512–518.
[48] A.A. Amayo, J.G. Kuria, Clinical application of tumour markers: a review, East [77] E.Y. Chung, C.J. Ochs, Y. Wang, L. Lei, Q. Qin, A.M. Smith, A.Y. Strongin,
Afr. Med. J. 86 (2009) S76–S83. R. Kamm, Y.X. Qi, S. Lu, Y. Wang, Activatable and cell-penetrable multiplex FRET
[49] R.M. Lahoud, A. O’Shea, C. El-Mouhayyar, I.D. Atre, K. Eurboonyanun, nanosensor for profiling MT1-MMP activity in single cancer cells, Nano Lett. 15
M. Harisinghani, Tumour markers and their utility in imaging of abdominal and (2015) 5025–5032.
pelvic malignancies, Clin. Radiol. 76 (2021) 99–107. [78] V.D.B. Bonifacio, Ovarian cancer biomarkers: moving forward in early detection,
[50] X. Zhang, F.G. Wu, P. Liu, N. Gu, Z. Chen, Enhanced fluorescence of gold Adv. Exp. Med. Biol. 1219 (2020) 355–363.
nanoclusters composed of HAuCl4 and histidine by glutathione: glutathione [79] M.N. Abou-Omar, M.S. Attia, H.G. Afify, M.A. Amin, R. Boukherroub, E.
detection and selective cancer cell imaging, Small 10 (2014) 5170–5177. H. Mohamed, Novel optical biosensor based on a nano-gold coated by schiff base
[51] H.J. Forman, H. Zhang, A. Rinna, Glutathione: overview of its protective roles, doped in sol/gel matrix for sensitive screening of oncomarker CA-125, ACS
measurement, and biosynthesis, Mol. Aspect. Med. 30 (2009) 1–12. Omega 6 (2021) 20812–20821.
[52] P. Kuppusamy, H. Li, G. Ilangovan, A.J. Cardounel, J.L. Zweier, K. Yamada, M. [80] S. Eyvazi, S. Farajnia, S. Dastmalchi, F. Kanipour, H. Zarredar, M. Bandehpour,
C. Krishna, J.B. Mitchell, Noninvasive imaging of tumor redox status and its Antibody based EpCAM targeted therapy of cancer, review and update, Curr.
modification by tissue glutathione levels, Cancer Res. 62 (2002) 307–312. Cancer Drug Targets 18 (2018) 857–868.
[53] G.F. Shi, J.T. Cao, J.J. Zhang, K.J. Huang, Y.M. Liu, Y.H. Chen, S.W. Ren, [81] L. Tao, K. Zhang, Y. Sun, B. Jin, Z. Zhang, K. Yang, Anti-epithelial cell adhesion
Aptasensor based on tripetalous cadmium sulfide-graphene molecule monoclonal antibody conjugated fluorescent nanoparticle biosensor for
electrochemiluminescence for the detection of carcinoembryonic antigen, Analyst sensitive detection of colon cancer cells, Biosens. Bioelectron. 35 (2012)
139 (2014) 5827–5834. 186–192.
[54] M.J. Duffy, R. Lamerz, C. Haglund, A. Nicolini, M. Kalousova, L. Holubec, [82] J. Shi, J. Lyu, F. Tian, M. Yang, A fluorescence turn-on biosensor based on
C. Sturgeon, Tumor markers in colorectal cancer, gastric cancer and graphene quantum dots (GQDs) and molybdenum disulfide (MoS2) nanosheets
gastrointestinal stromal cancers: European group on tumor markers 2014 for epithelial cell adhesion molecule (EpCAM) detection, Biosens. Bioelectron. 93
guidelines update, Int. J. Cancer 134 (2014) 2513–2522. (2017) 182–188.
[55] M. Grunnet, J.B. Sorensen, Carcinoembryonic antigen (CEA) as tumor marker in [83] E. Machackova, L. Foretova, M. Lukesova, P. Vasickova, M. Navratilova, I. Coene,
lung cancer, Lung Cancer 76 (2012) 138–143. H. Pavlu, V. Kosinova, J. Kuklova, K. Claes, Spectrum and characterisation of
[56] M. Turriziani, M. Fantini, M. Benvenuto, V. Izzi, L. Masuelli, P. Sacchetti, BRCA1 and BRCA2 deleterious mutations in high-risk Czech patients with breast
A. Modesti, R. Bei, Carcinoembryonic antigen (CEA)-based cancer vaccines: and/or ovarian cancer, BMC Cancer 8 (2008) 140.
recent patents and antitumor effects from experimental models to clinical trials, [84] A.P. Romagnolo, D.F. Romagnolo, O.I. Selmin, BRCA1 as target for breast cancer
Recent Pat. Anti-Cancer Drug Discov. 7 (2012) 265–296. prevention and therapy, Anti Cancer Agents Med. Chem. 15 (2015) 4–14.
[57] M. Chen, F. Yeasmin Khusbu, C. Ma, K. Wu, H. Zhao, H. Chen, K. Wang, [85] Y.S. Borghei, M. Hosseini, M.R. Ganjali, Detection of large deletion in human
A sensitive detection method of carcinoembryonic antigen based on dsDNA- BRCA1 gene in human breast carcinoma MCF-7 cells by using DNA-Silver
templated copper nanoparticles, New J. Chem. 42 (2018) 13702–13707. Nanoclusters, Methods Appl. Fluoresc. 6 (2017), 015001.
[58] Y. Jiang, Y. Tang, P. Miao, Polydopamine nanosphere@silver nanoclusters for [86] Y.S. Borghei, M. Hosseini, M.R. Ganjali, S. Hosseinkhani, A novel BRCA1 gene
fluorescence detection of multiplex tumor markers, Nanoscale 11 (2019) deletion detection in human breast carcinoma MCF-7 cells through FRET between
8119–8123. quantum dots and silver nanoclusters, J. Pharm. Biomed. Anal. 152 (2018)
[59] Z. Qiu, J. Shu, D. Tang, Bioresponsive release system for visual fluorescence 81–88.
detection of carcinoembryonic antigen from mesoporous silica nanocontainers [87] L.E. Buscaglia, Y. Li, Apoptosis and the target genes of microRNA-21, Chin. J.
mediated optical color on quantum dot-enzyme-impregnated paper, Anal. Chem. Cancer 30 (2011) 371–380.
89 (2017) 5152–5160. [88] Y. Gong, W. Yuan, X. Guo, Q. Zhang, P. Zhang, C. Ding, Fluorescent detection of
[60] F. Farshchi, M. Hasanzadeh, Nanomaterial based aptasensing of prostate specific microRNA-21 in MCF-7 cells based on multifunctional gold nanorods and the
antigen (PSA): recent progress and challenges in efficient diagnosis of prostate integration of chemotherapy and phototherapy, Mikrochim. Acta 188 (2021) 253.
cancer using biomedicine, Biomed. Pharmacother. 132 (2020), 110878. [89] M. Hosseini, E. Ahmadi, Y.S. Borghei, M. Reza Ganjali, A new fluorescence turn-
[61] M. Barani, F. Sabir, A. Rahdar, R. Arshad, G.Z. Kyzas, Nanotreatment and on nanobiosensor for the detection of micro-RNA-21 based on a DNA-gold
nanodiagnosis of prostate cancer: recent updates, Nanomaterials 10 (2020). nanocluster, Methods Appl. Fluoresc. 5 (2017), 015005.
[62] X. Huang, Z. He, D. Guo, Y. Liu, J. Song, B.C. Yung, L. Lin, G. Yu, J.J. Zhu, [90] Y. Jiang, Z. Guo, M. Wang, J. Cui, P. Miao, Construction of fluorescence logic
Y. Xiong, X. Chen, Three-in-one" nanohybrids as synergistic nanoquenchers to gates responding to telomerase and miRNA based on DNA-templated silver
enhance No-wash fluorescence biosensors for ratiometric detection of cancer nanoclusters and the hybridization chain reaction, Nanoscale 14 (2022) 612–616.
biomarkers, Theranostics 8 (2018) 3461–3473. [91] Z.Z. Li, L.F. Shen, Y.Y. Li, P. Chen, L.Z. Chen, Clinical utility of microRNA-378 as
[63] J.H. Choi, H.S. Kim, J.W. Choi, J.W. Hong, Y.K. Kim, B.K. Oh, A novel Au- early diagnostic biomarker of human cancers: a meta-analysis of diagnostic test,
nanoparticle biosensor for the rapid and simple detection of PSA using a Oncotarget 7 (2016) 58569–58578.
sequence-specific peptide cleavage reaction, Biosens. Bioelectron. 49 (2013) [92] Z. Liu, Y. Wang, J. Li, Y. Yuan, X. Wu, W. Liu, Y. Liu, A label-free fluorescent
415–419. enhancement nanosensor for ultrasensitive and highly selective detection of
[64] Y. Li, P.J. Cozzi, Targeting uPA/uPAR in prostate cancer, Cancer Treat Rev. 33 miRNA-378 through signal synergy amplification, Anal. Chim. Acta 1087 (2019)
(2007) 521–527. 86–92.
[65] R.M. Williams, C. Lee, D.A. Heller, A fluorescent carbon nanotube sensor detects [93] V.C. Kok, C.C. Yu, Cancer-derived exosomes: their role in cancer biology and
the metastatic prostate cancer biomarker uPA, ACS Sens. 3 (2018) 1838–1845. biomarker development, Int. J. Nanomed. 15 (2020) 8019–8036.
[66] X. Cheng, H.D. Xu, H.H. Ran, G. Liang, F.G. Wu, Glutathione-depleting [94] R. Kalluri, The biology and function of exosomes in cancer, J. Clin. Invest. 126
nanomedicines for synergistic cancer therapy, ACS Nano 15 (2021) 8039–8068. (2016) 1208–1215.
[67] A. Bansal, M.C. Simon, Glutathione metabolism in cancer progression and [95] B. Li, W. Pan, C. Liu, J. Guo, J. Shen, J. Feng, T. Luo, B. Situ, Y. Zhang, T. An,
treatment resistance, J. Cell Biol. 217 (2018) 2291–2298. C. Xu, W. Zheng, L. Zheng, Homogenous magneto-fluorescent nanosensor for
[68] C. Li, Y. Xu, S.Y. Liu, Y. Zhang, W. Yin, Z. Dai, X. Zou, Cancer cell identification by tumor-derived exosome isolation and analysis, ACS Sens. 5 (2020) 2052–2060.
facile imaging of intracellular reductive substances with fluorescent nanosensor, [96] J. Wang, H. Xie, C. Ding, Designed Co-DNA-Locker and ratiometric SERS sensing
Talanta 234 (2021), 122650. for accurate detection of exosomes based on gold nanorod arrays, ACS Appl.
[69] Q. Wang, L. Li, X. Wang, C. Dong, S. Shuang, Graphene quantum dots wrapped Mater. Interfaces 13 (2021) 32837–32844.
square-plate-like MnO2 nanocomposite as a fluorescent turn-on sensor for [97] P. Dell’ Antone, Energy metabolism in cancer cells: how to explain the Warburg
glutathione, Talanta 219 (2020), 121180. and Crabtree effects? Med. Hypotheses 79 (2012) 388–392.

13
N. Ouyang et al. Polymer Testing 115 (2022) 107746

[98] J.C. Melvin, H. Garmo, L. Holmberg, N. Hammar, G. Walldius, I. Jungner, [102] H. Ding, X.-X. Zhou, J.-S. Wei, X.-B. Li, B.-T. Qin, X.-B. Chen, H.-M. Xiong, Carbon
M. Lambe, M. Van Hemelrijck, Glucose and lipoprotein biomarkers and breast dots with red/near-infrared emissions and their intrinsic merits for biomedical
cancer severity using data from the Swedish AMORIS cohort, BMC Cancer 17 applications, Carbon 167 (2020) 322–344.
(2017) 246. [103] D. Li, E.V. Ushakova, A.L. Rogach, S. Qu, Optical properties of carbon dots in the
[99] X. Hai, X. Zhu, K. Yu, S. Yue, W. Song, S. Bi, Dual-mode glucose nanosensor as an deep-red to near-infrared region are attractive for biomedical applications, Small
activatable theranostic platform for cancer cell recognition and cascades- 17 (2021), e2102325.
enhanced synergetic therapy, Biosens. Bioelectron. 192 (2021), 113544. [104] W. Tahir, S. Kura, J. Zhu, X. Cheng, R. Damseh, F. Tadesse, A. Seibel, B.S. Lee,
[100] L. Chen, H. Li, H. He, H. Wu, Y. Jin, Smart plasmonic glucose nanosensors as F. Lesage, S. Sakadžic, D.A. Boas, L. Tian, Anatomical modeling of brain
generic theranostic agents for targeting-free cancer cell screening and killing, vasculature in two-photon microscopy by generalizable deep learning, BME
Anal. Chem. 87 (2015) 6868–6874. Frontiers 2021 (2021) 1–12.
[101] R. Yan, Z. Guo, X. Chen, L. Tang, M. Wang, P. Miao, Red-emissive carbon [105] Y. Han, B. Tang, L. Wang, H. Bao, Y. Lu, C. Guan, L. Zhang, M. Le, Z. Liu, M. Wu,
nanodots for highly sensitive ferric(III) ion sensing and intracellular imaging, Machine-learning-driven synthesis of carbon dots with enhanced quantum yields,
Analyst 146 (2021) 6450–6454. ACS Nano 14 (2020) 14761–14768.

14

You might also like