ArticlePDF Available

Graphene Transistors for In Vitro Detection of Health Biomarkers

Wiley
Advanced Functional Materials
Authors:

Abstract and Figures

Biomarkers are primary indicators for precise diagnosis and treatment. The early identification of health biomarkers has been sustained by the evolutionary success in sensor technologies. Among them, graphene field‐effect transistor (GFET) biosensors have exhibited major advantages such as an ultrashort response time, high sensitivity, easy operation, capability of integration, and label‐free detection. Owing to the atomic thickness, graphene restricts charge carrier flow merely at the material surface and responds to foreign stimuli directly, leading to effective signal acquisition and transmission. Here, this review summarizes the latest advances in GFET biosensors in a comprehensive manner that contains the device design, working principle, surface functionalization, and proof‐of‐concept applications. It provides a comprehensive survey of GFET biosensors with regard to biomarker analysis at the single‐device level and integrated prototypes that include wearable sensors, biomimetic systems, healthcare electronics, and diagnostic platforms. Moreover, there is discussion on the long‐standing research efforts and outlook for the future development of GFET sensor systems from lab to fab.
This content is subject to copyright. Terms and conditions apply.
REVIEW
www.afm-journal.de
Graphene Transistors for In Vitro Detection of Health
Biomarkers
Changhao Dai, Derong Kong, Chang Chen, Yunqi Liu, and Dacheng Wei*
Biomarkers are primary indicators for precise diagnosis and treatment. The
early identification of health biomarkers has been sustained by the
evolutionary success in sensor technologies. Among them, graphene
field-effect transistor (GFET) biosensors have exhibited major advantages
such as an ultrashort response time, high sensitivity, easy operation,
capability of integration, and label-free detection. Owing to the atomic
thickness, graphene restricts charge carrier flow merely at the material surface
and responds to foreign stimuli directly, leading to effective signal acquisition
and transmission. Here, this review summarizes the latest advances in GFET
biosensors in a comprehensive manner that contains the device design,
working principle, surface functionalization, and proof-of-concept
applications. It provides a comprehensive survey of GFET biosensors with
regard to biomarker analysis at the single-device level and integrated
prototypes that include wearable sensors, biomimetic systems, healthcare
electronics, and diagnostic platforms. Moreover, there is discussion on the
long-standing research efforts and outlook for the future development of
GFET sensor systems from lab to fab.
1. Introduction
Health biomarkers are essential biological indicators in clini-
cal diagnostics.[, ] They are associated with human tissue bio-
logical characteristics and can provide valuable information for
early diagnosis, treatment, and prevention of diseases.[– ] Thus,
identifying a health-related biomarker at an early stage is of vi-
tal importance in healthcare applications. Conventional analyt-
ical techniques suer from temporal heterogeneities, invasive
procedures, insucient sensitivity, and time-consuming and so-
phisticated operation.[– ] By comparison, electrical sensing tech-
nology has attracted extensive interest due to its noninvasive,
C. Dai, D. Kong, C. Chen, D. Wei
State Key Laboratory of Molecular Engineering of Polymers
Department of Macromolecular Science
Fudan University
Shanghai , China
E-mail: weidc@fudan.edu.cn
C. Dai, D. Kong, C. Chen, Y. Liu, D. Wei
Laboratory of Molecular Materials and Devices
Fudan Un iver sity
Shanghai , China
The ORCID identification number(s) for the author(s) of this article
can be found under https://doi.org/./adfm.
DOI: 10.1002/adfm.202301948
label-free, real-time, and easily-operated de-
tection capability.[,] Over the past few
decades, research eorts have been devoted
to developing advanced electric sensors
such as lab-on-a-chip platforms,[] lateral
flow immune assays,[] electrochemilu-
minescence sensors,[] field-eect transis-
tor (FET) sensors,[, ] enzyme-linked im-
munosorbent assays,[] surface plasmon
resonance,[, ] and others.[,, ] Among
them, FET-based sensors exhibit major ad-
vantages such as high integrability, inher-
ent signal amplification ability, low energy
expenditure, and ultrashort turn-around
time.[, ] However, traditional FET sensors
are limited by low signal conversion e-
ciency and poor sensor performance be-
cause their bulk channel (nm) re-
stricts the carrier modulation at the solid-
electrolyte surface.[, ] To address this is-
sue, advances in FET sensors have been
made in replacing the bulk channel by D
materials with atomic thickness (< nm), which in-
clude graphene family materials,[,, ] transition metal
dichalcogenide,[] black phosphorus,[,] and D transition-
metal carbides (MXenes),[–] just to name a few.[– ]
D material-based FETs, especially graphene-based FETs
(GFETs), have demonstrated unique potential in biomarker anal-
ysis and healthcare applications.[,,, ] Due to an ideal van der
Waals surface without dangling bonds, all carriers of graphene
sheets flow merely on the surface and are directly exposed to
foreign stimuli/perturbations, leading to their ultrasensitive re-
sponse to target analytes.[– ] Additionally, the large surface-to-
volume ratio endows graphene with abundant modification sites,
which is primary to building integrated and multifunctional sen-
sor platforms.[, ] From these standpoints, GFET-based sensor
technology is quite attractive for sensitive biomarker detection
in healthcare fields.[– ] Recently, substantial research eorts in
GFET biosensors have been devoted to the following aspects: i)
how to build high-performance sensor units with standardized
methods; ii) how to develop integrated and multifunctional pro-
totypes with the balance of cost, user interface and device reli-
ability; and iii) how to understand the relevancy between sensor
information and clinical characteristics in human testing.[,,, ]
In this review, we systematically discuss recent progress in
GFET biosensors, from fundamental investigation at the single-
device level to integrated systems for health monitoring appli-
cations. This review provides a brief description of the device
configuration, working principle, surface functionalization, and
performance characterization. A comprehensive report of GFET
Adv. Funct. Mater. 2023,  ©  Wiley-VCH GmbH
2301948 (1 of 26)
www.advancedsciencenews.com www.afm-journal.de
Figure 1. Working principles of GFET biosensors. a) Schematic illustration of a liquid-gate GFET sensor. b) The analytes include proteins, nucleic acids,
viruses, bacteria, etc. The probes include aptamers, antibodies, enzymes, CRISPR/Cas, etc. c) Typical ambipolar transfer characteristics of graphene. d)
Sensing principle on the graphene surface: the binding of negatively (positively) charged analytes induced negative (positive) shifts of VCNP. e) Potential
diagram of the electrical double layer model. The arrow in the dipole shows the electric field generated by the positive charge of the protein on the
gate surface. f) Relative change in the normalized signal at dierent analyte concentrations. (a) Adapted with permission. [] Copyright , American
Chemical Society. (e) Reproduced under the terms of CC BY-NC license.[] Copyright , The Authors, published by the American Association for the
Advancement of Science.
biosensors with regard to their conceptual applications in non-
invasive biomarker detection is provided. The following sec-
tion summarizes sensing prototypes, including wearable sen-
sors, biomimetic systems, healthcare electronics, and diagnostic
platforms. Finally, we discuss future challenges and put forward
suggestions to build sensor systems that are suitable for home-
care monitoring and healthcare applications.
2. Design and Operation
2.1. Working Principle
A GFET sensor works by monitoring the channel current modu-
lation at the interface, where current modulation can be induced
by external perturbations such as charged adsorption, biological
binding, and chemical reactions.[, ] Under an external gate bias
(Vgs), the gate electrode and the semiconductor act as either plat
of a capacitor. Accordingly, an induced electric field perpendic-
ular to the gate electrode forms a conducting channel, in which
the carriers flow from the source to drain.[, ] Figure 1a schemat-
ically shows a liquid-gate GFET sensor with a reference electrode
providing gate voltage through the electrolyte. Composed of all
sp-hybridized carbon atoms, graphene can promote in-plane car-
rier transport at the surface, which makes it extremely sensitive
to the external environment.[, ] To develop GFET biosensors,
biological probes are immobilized on the channel via dierent
linkers to capture target analytes and generate measurable sig-
nals (Figure b). Although many factors, such as the operational
mode, electrode encapsulation and choice of channel materials,
aect the sensing performance, the heart of GFET biosensors
Adv. Funct. Mater. 2023,  ©  Wiley-VCH GmbH
2301948 (2 of 26)
16163028, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/adfm.202301948 by Fudan University, Wiley Online Library on [11/05/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.afm-journal.de
depends largely on the interface between the graphene chan-
nel and the electrolyte.[,, ] For example, the Debye length (𝜆D)
is one of the key parameters that demonstrates the charge car-
rier’s net electrostatic eect in electrolyte solutions.[, ] Within
𝜆D, the charged analytes will introduce mobility modulation in
the graphene channel. If the distance between the charged ana-
lytes and the graphene channel is larger than 𝜆D, a notable de-
terioration of the sensor response is expected due to the Debye
screening layer.[,] For physiological solutions at room temper-
ature, the Debye length can be calculated via Equation :[]
𝜆D=𝜀𝜀rkBT
NAeI()
where 𝜖is the permittivity of free space; 𝜖ris the dielectric con-
stant; kBis Boltzmann’s constant; Tis the temperature; NAis
Avogadro’s number; eis the elementary charge; and Iis the ionic
strength of the solution.
Three characteristic curves are widely used for operating GFET
biosensors, which include output curves, transfer curves and
time-dependent response curves. First, Figure c indicates a rep-
resentative transfer curve (Ids versus Vgs) of GFETs. The trans-
fer curve is obtained by sweeping Vgs and a constant drain volt-
age, which oers FET performance parameters such as the field-
eect mobility, threshold voltage, and charge neutrality point
(CNP).[, ] VCNP (also known as VDirac) is the gate voltage at the
minimum current, where the electron transport almost equals
the hole transport.[,, ] After recognizing positively charged
analytes, VCNP shifted toward a positive potential (right panel
in Figure d), and the device exhibited a p-doping eect in the
electrolyte, and vice versa (left panel in Figure d). In such dop-
ing eects, induced charges move from charged analytes to the
graphene surface directly, thus modulating the Fermi level of
graphene as well as the carrier mobility of GFETs.[] Second, the
output curves (Ids versus Vds) demonstrate the change in drain-
to-source current under sweeping drain voltage at certain Vgs.
The current shifts correspond to the analyte concentrations, and
the linear behavior of the output curves indicates the contact
quality of the electrodes. Third, the time-dependent tests moni-
tor the drain-to-source current Ids, which is aected by the sur-
face potential of the gate electrode.[, ] More specifically, the
charged analytes induce a potential barrier and an electric dipole
across the electric double layer at the electrolyte/gate interface
(Figure e).[] When |Vds ||VpVg,e|, the channel current Ids
of GFETs is given by Equation :[,, ]
Ids =𝜇W
LCiVpVg,e +Vds
Vds N()
where μis the hole mobility; Wand Lare the channel width and
length, respectively; Ciis the eective capacitance per unit area
of the GFET; Vpis the pinch-o voltage; Vg,e is the eective gate
voltage; and Nis the number of charged analytes. Accordingly,
the change in Ids is proportional to the number of charged an-
alytes adsorbing on the graphene channel, contributing to the
quantitative detection of targets. However, the FET testing quan-
tity is nontrivial because challenges remain in two aspects: i) the
diculty in obtaining the precise charge number of each binding
process; and ii) the inherent staturation state (Figure f) when all
bioreceptors are occupied by target analytes.[,,, ] Experimen-
tally, the anity between probes and analytes can be obtained via
the Hill-Langmuir equation:[, ]
ΔVDirac
ΔVDirac,max
=AcsensingKDn
+csensingKDn()
where Ais the saturation response coecient of GFET sensors;
ΔVDirac,max is the largest ΔVDirac, denoted ΔVDirac,max =VDirac,max
VDirac, (VDirac,max and VDirac, relate to the analyte solution with
maximum and no analytes, respectively); nis the Hill coecient
corresponding to the binding cooperativity; and KDof bioreceptor
probes is extracted from the fitted curve (see the linear region in
Figure f).
2.2. Device Fabrication
A GFET is a three-terminal electronic device that includes source,
drain and gate electrodes, with a semiconductor channel bridg-
ing the source and drain.[] The typical fabrication process of
GFET biosensors includes four steps: i) electrode fabrication; ii)
graphene synthesis, transfer and patterning; iii) surface func-
tionalization; and iv) electrode packaging/encapsulation.[,, ]
For GFET biosensors, the key to fabrication is to construct an
active layer that consists of high-quality graphene and specific
recognition probes.[, ] Generally, current approaches for syn-
thesizing monolayer graphene can be categorized by top-down
fabrication (e.g., mechanical/liquid exfoliation), bottom-up fab-
rication (e.g., chemical/physical vapor deposition), and organic
synthesis.[,, ] Due to the low eciency of exfoliation and
poor quality of organic synthesis, the chemical vapor deposition
method has been widely applied in producing high-quality, large-
area, and highly uniform graphene sheets for various sensing
applications.[, ]
2.2.1. Surface Functionalization
Surface functionalization is of vital importance to build high-
performance GFET biosensors.[, ] To date, a large number of
publications have reported various functionalization strategies,
including channel functionalization (e.g., bifunctional linkers via
N-hydroxysuccinimide chemistry,[,] carbon quantum dots,[]
drop-casting strategy,[,] nanoparticles,[ ,] etc.[])andgate
functionalization (e.g., Staphylococcus aureus protein A,[]
sulfhydryl group or gold-thiol binding,[] etc.[,] ). Figure 2a,b
demonstrates two representative modification approaches on
the graphene channel. As one of the most widely-used bifunc-
tional linkers, -pyrenebutyric acid N-hydroxysuccinimide ester
(PASE) contains a pyrene group absorbed on graphene via 𝜋-𝜋
stacking and a carbonyl group activated with biological probes
(Figure a).[,,,] Noticeably, the excess PASE molecules on the
sensing area will lead to nonspecific absorption and thereby false
diagnosis in clinical tests. Thus, research eorts have been de-
voted to precise functionalization to achieve controllable implace-
ment of recognition probes on graphene.[,, ] In , Kwon
et al. proposed a linker-free functionalization strategy based on
the edge eects of patterned graphene, indicating a – fold
Adv. Funct. Mater. 2023,  ©  Wiley-VCH GmbH
2301948 (3 of 26)
16163028, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/adfm.202301948 by Fudan University, Wiley Online Library on [11/05/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.afm-journal.de
Figure 2. Functionalization strategies of GFET biosensors. a,b) Channel functionalization via a) bifunctional linkers such as PASE and b) edge defects
on patterned graphene. c–e) Gate functionalization via c) gold-thiol binding, d) tetrahedral DNA nanostructures, and e) extended gate modification.
(a) Reproduced under the terms of CC BY license. [] Copyright , The Authors, published by American Chemical Society. (b) Reproduced under
the terms of CC BY license. [] Copyright , The Authors, published by MDPI. (c) Reproduced under the terms of CC BY-NC license. [] Copyright
, The Authors, published by the American Association for the Advancement of Science. (d) Adapted with permission. [] Copyright , American
Chemical Society. (e) Reproduced under the terms of CC BY license. [] Copyright , The Authors, published by Springer Nature.
enhanced response compared to pristine GFETs modified with
succinimidyl ester (Figure b).[] More recently, Wang and
coworkers applied tetrahedral DNA nanostructures to build
artificial electromechanical devices with higher precision and
more functionalities on the graphene channel.[] They also
found that DNA nanostructures helped control the distribution
of probes in the sensing area, thus leading to enhanced per-
formance of GFET biosensors and other bioelectronics. Apart
from channel modification methods, valuable progress has also
been made in gate functionalization to develop advanced FET
sensors.[,, ] Liu et al. linked mercaptoacetic acid-modified
SARS-CoV- antibodies onto a Au gate electrode via gold-thiol
binding, which enables gate voltage pulses and minute-level
detection (Figure c).[] By comparison, Wang et al. used a
DNA-functionalized graphene gate to enrich the detection signal
caused by target analytes, which achieved .-fold improved sen-
sitivity compared with conventional channel functionalization
methods (Figure d).[] Recently, Sheibani et al. developed a
wearable sensory electronic chip based on platinum/graphene
aptamer extended gate GFETs (Figure e).[] In this configura-
tion, the gate electrode is attached through metal vias above the
aptamer-modified graphene, which allows the complementary
metal–oxide–semiconductor (CMOS) process for high-level inte-
gration. Furthermore, the extended-gate structure demonstrates
major advantages such as less drift and temperature dependence
in sensing applications.[, ]
2.2.2. Antifouling Strategies
Antifouling strategies are also crucial for maintaining high sen-
sor performance.[, ] Generally, electrode fouling means non-
specific binding that blocks the active sites of the sensing sur-
face (Figure 3a).[ ,,] The issue of fouling causes rapid loss of
sensitivity and specificity, especially when GFETs are exposed to
complex detection solutions such as saliva, serum, blood, and
plasma.[, ] To address this issue, research eorts have been
devoted to immobilizing block layers on graphene sensing sur-
faces to prevent unwanted nonspecific adsorption, which in-
cludes polyethylene glycol (PEG),[– ] bovine serum albumin
(BSA),[] DNA nanostructures,[] ethanolamine,[] etc.[,] As
an example, Wang et al. compared two antifouling strategies
by using BSA and a DNA tetrahedral (DNA-TDN) layer as the
block layer of the sensing area (Figure b,c).[] Both of these
Adv. Funct. Mater. 2023,  ©  Wiley-VCH GmbH
2301948 (4 of 26)
16163028, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/adfm.202301948 by Fudan University, Wiley Online Library on [11/05/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.afm-journal.de
Figure 3. Antifouling strategies of GFET biosensors. a–c) Schematic illustration of nonspecific binding on a) bare graphene, b) graphene modified with
a BSA layer, and c) a DNA-TDN layer. d) Antifouling strategy in the COF/graphene FET biosensors. The red circle shows the chelating eect at the
sensing surface. e) Scheme illustrating the hydrogel-based encapsulation of sensor electrodes. (a–c) Reproduced under the terms of CC BY license.[]
Copyright , The Authors, published by Springer Nature Limited. (d) Adapted with permission.[] Copyright , Wiley-VCH Verlag GmbH & Co.
KGaA, Weinheim. (e) Reproduced under the terms of CC BY license.[] Copyright , published by Springer Nature Limited.
block layers eciently avoid nonspecific binding and enhance
the long-term stability in solutions. By comparison, Yang et al.
prevented potential foiling by combining covalent organic frame-
works (COFs) with GFET biosensors (Figure d).[] In this con-
figuration, the mesoporous crystalline structure of COFs not only
avoids nonspecific adsorption but also leads to a chelating eect
for an ultrashort response time ( ms). Most recently, Atkin-
son et al. demonstrated an electrode packaging strategy based
on the combination of nanoparticles and hydrogels, leading to
enhanced sensor sensitivity and high stability at the same time
(Figure e).[] Noticeably, this encapsulation strategy facilitates
ecient electron transfer at the analyte-electrode interface, which
provides design rules to build miniature biosensors with high re-
liability and accuracy in real-world tests.
2.3. Performance Assessment
An ultimate goal of GFET biosensors is to enable home-
care monitoring applications in the digital health era.[– ]
However, variations in material quality, device configuration,
surface functionalization, testing conditions, operation proce-
dures, and signal evaluation lead to uncertainty during prac-
tical applications.[,, ] To better quantify the sensor per-
formance, several characterization metrics are defined as
follows:
Limit of Detection: The limit of detection (LoD) means the
ability to measure the smallest amount or concentration level
of analytes. A common LoD calculation approach recommended
by the IUPAC utilizes the exposure of a sensing device to
known analyte concentrations to generate a calibration curve
(Figure f).[,,] Experimentally, most analytical specifications
depend on the three-deviation approach (i.e., the analyte concen-
tration at the signal that is three times the background noise level)
to obtain credible output signals (Equation ).[– ]
LoD =𝜎
S()
where 𝜎is the standard deviation of the testing results (i.e.,
𝜎·root-mean-square noise); and Sis the slope of the calibration
curves.
Range of Detection: The range of detection (RoD) is the linear
region in which the detection signal is proportional to the num-
ber/amount of analytes. Notably, the RoD can be obtained from
the smallest to the largest point in the calibration curves rather
than the analytical measuring interval.
Response: Response in this Review is determined by the rela-
tive change of testing signals, including current, resistance, volt-
age, etc. To set the current change as an example, the sensor re-
sponse can be calculated as follows:
response =Isen I
I
×%()
where Iis the initial current of the sensor and Isen is the mea-
sured current after testing. According to Equation , the response
time is determined as the detection time when the signal arrives
at % of the maximum detection value.
Accuracy: Testing accuracy reflects measure deviation with
the true value, denoted as “accuracy =(average true value)/true
value”. Practically, accuracy is often demonstrated by the area un-
der receiver operating characteristic (ROC) curves.[] In typical
ROC curves, the cross-validation prediction performance is com-
posed of two parts:
Sensitivity =TP
TP +FN ()
Specificity =TN
TN +FP ()
Adv. Funct. Mater. 2023,  ©  Wiley-VCH GmbH
2301948 (5 of 26)
16163028, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/adfm.202301948 by Fudan University, Wiley Online Library on [11/05/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.afm-journal.de
where TP represents true positives, TN represents true negatives,
FP represents false positives, and FN represents false negatives.
Thus, specificity demonstrates the sensor capability to provide
a statistically significant signal to target analytes and a negligi-
ble signal to nonspecific analytes. As a comparison, sensitivity
reflects the response to target analytes at a tiny concentration.
Durability: Sensor durability refers to the long-standing qual-
ity or maintenance capability of the initial response under
variable external conditions (e.g. temperature, pressure, time,
moisture).[, ] Equation gives a calculation approach to obtain
the temperature durability:
responsetemp =responsetemp response
response
()
where responseis the initial sensor response and responsetemp
is the response temperature measured after storage.
Relative Standard Deviation: The relative standard deviation
(RSD) reflects the deviated degree of a set of data. In other words,
the RSD value indicates how precise the average of the experi-
mental results is, which can be calculated as follows:
RSD =SD
X×%()
where SD is the standard deviation of the observations and
Xis
the mean value of the data.
3. Biomarker Analysis
There is an everlasting demand for sensitive detection of health-
related biomarkers, including nucleic acids,[] proteins,[] small
biomolecules,[, ] and cell-related products.[, ] This section
summarizes the recent progress in GFET biosensors for bio-
chemical sensing and related fitness applications over the past
few decades.
3.1. Nucleic Acid
Nucleic acids are the carriers of genetic information that plays
a crucial role in biological processes.[– ] Early advances in
GFET nucleic acid sensors have been made in the sensitive de-
tection of DNA/RNA in unprocessed biological fluids.[,,,]
For example, Ganguli et al. demonstrated zeptomolar-level de-
tection of enzymatic DNA amplification using crumpled GFETs
(Figure 4a).[] The authors took advantage of the crumpled
graphene and Bst polymerase in loop-mediated isothermal am-
plification to electrically detect the reduction in the primer
molecules.[] Due to the increased Debye length at the crumpled
graphene surface, the GFET sensor exhibited larger VDirac shifts
than flat graphene-based sensors (Figure b), leading to an LoD
as low as × ssDNA in buer. Similarly, Huang et al.
applied a reverse transcription-loop-mediated isothermal ampli-
fication to fabricate a GFET nucleic acid sensor with an LoD of
 copies mLRNA in a sample.[]
Research progress has also been demonstrated in the rapid nu-
cleic acid detection of unamplified samples.[– ] In , Ha-
jian et al. combined GFETs with clustered regularly interspaced
short palindromic repeat (CRISPR)-associated nuclease (Cas)-
based technologies to enable the digital detection of a target
gene sequence.[] The as-prepared platform achieved -minute
detection of unamplified DNA samples with a sensitivity of
. × .[] Shortly after, the same research group modi-
fied the GFET sensor by using dierent Cas variants and ortho-
logues to improve single-nucleotide polymorphisms (SNP) dis-
crimination, to fabricate a single-nucleotide polymorphism chip
and to enable multiplex electrical measurements at the same time
(Termed CRISPR–SNP-Chip, Figure c).[] The CRISPR-SNP-
Chip achieved the digital testing of a target gene with SNP speci-
ficity. As a demonstration, the chip was able to discriminate ge-
nomic DNA from wild-type hiPSCs (termed WTC) and hiPSCs
(termed CS) in the fALS disease model with a statistically sig-
nificant dierence (Figure d). Apart from CRISPR technologies,
researchers have designed other DNA aptamers for ultrasensi-
tive sensing applications.[, ] For example, Kong et al. demon-
strated a GFET nucleic acid sensor based on Y-shaped DNA dual
probes (Y-dual probes, Figure e).[] Owing to the modified Y-
dual probes that simultaneously bind with ORFab and N genes
of SARS-CoV- nucleic acid, the GFET sensor exhibited a high
recognition ratio and an LoD down to . copy μL(Figure f),
which is orders of magnitude lower than existing nucleic
acid assays.
This section summarizes representative applications of GFET
nucleic acid sensors, including quantitative detection, genome
mutation monitoring, and disease diagnosis. Table 1 presents
and compares their sensing performance by categorizing the tar-
get analytes.
3.2. Protein
Proteins consisting of one or more amino acid chains are unique
components in all living organisms and are of vital impor-
tance in physiological systems.[, ] This section summarizes
the latest advances in GFET protein sensors for healthcare ap-
plications such as monitoring enzymatic activity,[] protein-
to-protein interactions,[ ] early disease diagnosis,[ ] and epi-
demic prevention and control,[,, ] and so on.[, ]
Research progress has been made in aptamer-conjugated
strategies for sensitively detecting proteins at dierent
concentrations.[, ] For example, Yu et al. functionalized
solution-gate GFETs with a -base DNA aptamer on the gate
electrode to detect thrombin molecules as low as  .[ ]
After recognizing the heparin-binding site of thrombin, the
DNA aptamer formed a stable G-quadruplex structure and
caused measurable current changes within  s.[ ] Most
recently, Ban et al. developed a DNA aptamer-conjugated GFET
sensor platform to rapidly (< min) detect nucleocapsid, spike,
and receptor-binding domain (RBD) proteins of SARS-CoV-
in saliva samples.[] The hand-held wireless sensor device
exhibited LoDs of . and . PFU mLfor spike and
nucleocapsid proteins, respectively. Furthermore, the sensor
platform was able to distinguish COVID--positive samples
(e.g., Omicron-B.., NY, DG, and YF virtual
variants) from negative controls (i.e., Middle East Respiratory
Syndrome samples) with statistical significance, indicating its
Adv. Funct. Mater. 2023,  ©  Wiley-VCH GmbH
2301948 (6 of 26)
16163028, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/adfm.202301948 by Fudan University, Wiley Online Library on [11/05/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.afm-journal.de
Figure 4. GFET nucleic acid sensors. a,b) Crumpled GFET nucleic sensor: a) Schematic illustration of detecting target and nontarget dsDNA near the
crumpled graphene surface. b) Response contrast for amplified and nonamplified primer DNA. c,d) CRISPR-based GFET nucleic sensor: c) Working
principle of the Cas-CS enzyme in the presence of CS (left panel) or WTC genomic DNA (right panel). d) Response contrast for DNA samples
with a single-base mismatch. e,f) DNA-based Y dual-probe GFET sensor: e) Device configuration and sensing mechanism at the sensing interface of
GFETs. f ) Signal response with dierent probes at dierent SARS-CoV- cDNA concentrations. (a, b) Adapted with permission. [] Copyright ,
WILEY-VCH Verlag GmbH & Co. KGa. (c, d) Reproduced under the terms of CC BY license. [] Copyright , The Authors, published Springer Nature.
(e, f) Adapted with permission. [] Copyright , American Chemical Society.
potential for wide-range public health applications and real-time
environmental monitoring.[]
Advances in GFET protein sensors have also focused on im-
proving sensor performance with emerging technologies.[, ]
For example, Dai et al. developed a multiantibody functionaliza-
tion strategy to enhance the sensitivity and accuracy of GFET
biosensors. In this configuration, three dierential antibodies
were immobilized on the graphene surface to bind not only RBD
domains but also adjacent sites of SARS-CoV- (Figure 5a).[]
Owing to the cooperative recognition eect, the sensor demon-
strated an antigen-binding anity of . × and an LoD
down to . × gmL
spike protein in artificial saliva.
Furthermore, the multiantibody GFET sensor was also able to
detect diluted COVID- clinical samples, while conventional
single-antibody sensors exhibited negligible response changes
(Figure b). By comparison, Hwang et al. introduced a special
functionalization strategy based on crumpled graphene to im-
prove sensor performance.[ ] The authors found that the cav-
ernous planar graphene surface can decrease the Debye length,
leading to almost tenfold enhanced sensitivity in .×PBS so-
lution (Figure c). Moreover, the as-prepared GFET with vari-
able crumpled ratios (–%) is a powerful tool to investigate
protein-to-protein interactions.
Although considerable eorts have been made to enhance the
sensitivity of GFET biosensors, several challenges remain before
their successful translation to the medical market. One of the key
challenges in practice is how to avoid nonspecific adsorption in
unprocessed biological fluids, which would block sensing active
sites, generate background noise, and damage the long-term sta-
bility of GFETs.[, ] To address this issue, Wang et al. designed
a self-assembled sti tetrahedral double-stranded DNA structure
to avoid nonspecific adsorption from background biomolecules
(Figure d).[] The individual DNA tetrahedron consists of a .-
nm-high base and a flexible probe, where the rigid bases serve
as an anti-fouling layer that keeps nontarget analytes away from
the graphene channel. The developed GFET sensor exhibited
Adv. Funct. Mater. 2023,  ©  Wiley-VCH GmbH
2301948 (7 of 26)
16163028, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/adfm.202301948 by Fudan University, Wiley Online Library on [11/05/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.afm-journal.de
Tabl e 1 . Sensing Performance of GFET Nucleic Acid Sensors.
Analyte Receptor LoD Detection time [s] Condition Ref.
DNA CRISPR–dCas . × < MgClbuer []
DNA CRISPR–dCas . × < Buer []
DNA DNA . ×  PB []
DNA DNA ×  PBS []
DNA DNA . ×  .×PBS []
DNA DNA ×  ×PBS []
DNA DNA ×  .×PBS []
DNA DNA × > .×PBS []
DNA DNA × < PBS []
DNA DNA × N/A DI water []
DNA DNA × < DI water []
DNA DNA Tweezer × > Tris buer []
DNA G-quadruplex DNAzymes . ×  PBS, serum []
DNA ssDNA ×  ×SCC []
miRNA DNA .× N/A PBS []
miRNA- DNA ×  PBS []
miRNA- ssDNA ×  .×PBS []
miRNA- DNA ×  ×PBS []
RNA CRISPR-Casa . copies μL .×PBS []
RNA CRISPR-Casa ×  nuclease-free water []
RNA CRISPR-Casa . ×  ×PBS []
RNA DNA ×  serum []
RNA DNA ×  serum []
RNA DNA . fg mL throat swab []
RNA DNA . copies μL throat swab []
RNA DNA . × < PBS []
RNA DNA – copies μL ×TRB []
RNA DNA .–. copies μL full artificial saliva []
RNA DNA .–. copies μL full artificial saliva []
RNA DNA  copies μL full artificial saliva []
RNA PMO . × . ×  serumthroat swab []
Note: N/A, not available. DI, deionized. PB, phosphate buer. PBS, phosphate-buered saline. PMO, phosphorodiamidate morpholino oligos. PNA, peptide nucleic acid.
SCC, saline-sodium citrate.
almost % accuracy with an LoD down to .–. copies μL
unamplified SARS-CoV- nucleic acids in biofluids.[] Because
this anti-fouling strategy is not a trade-o between sensitivity and
accuracy, it enables the GFET to be a comprehensive tool for accu-
rate detection of ions ( × Hg
+), proteins (e.g., ×
thrombin), and small biomolecules (e.g., × adeno-
sine triphosphate) in complex biological fluids.[]
To date, rigorous progress has been made in developing GFET
protein sensors based on dierent configurations and functional-
ization strategies. Table 2 summarizes the sensing performance
of selective GFET protein sensors.
3.3. Small Biomolecules
Small biomolecules are key biomarkers in early disease di-
agnosis, including glucose,[] dopamine,[] adenosine
triphosphate,[ ] urea,[ ] and metabolites with an average
molecular weight of < kDa.[,, ] This section analyzes
notable progress in GFET small biomolecular sensors by
categorizing their recognition probes.
Early advances in GFET small biomolecular sensors focused
on enzyme-based functionalization for detecting a vast number
of analytes.[, ] These GFET biosensors work by monitoring
relative pH changes during enzyme-catalyzed hydrolysis.[, ]
For instance, Fenoy et al. developed an acetylcholine sensor based
on an acetylcholinesterase (AchE)-modified strategy and the elec-
trosynthesis of an amino gmoiety-bearing polymer layer on the
graphene channel (Figure 6a).[ ] In this device, the copoly-
mer poly(-amino-benzylamine-co-aniline) film can not only pro-
vide a non-denaturing environment for enzyme immobilization,
but also enhance the pH sensitivity of GFETs (from . to
. μApH
). Thus, the as-prepared GFET sensor responded
to pH changes induced by enzyme-catalyzed hydrolysis of tar-
get analytes. The LoD reaches . × with a sensitivity of
(. ±.) μAAch
decade and an average response time of
Adv. Funct. Mater. 2023,  ©  Wiley-VCH GmbH
2301948 (8 of 26)
16163028, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/adfm.202301948 by Fudan University, Wiley Online Library on [11/05/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.afm-journal.de
Figure 5. GFET protein sensors. a,b) Multiantibody GFET sensor: a) Schematic illustration of the sensing principle on the graphene surface, where the
three antibodies bind to dierent sites of the SARS-CoV- virus. b) Comparison of the detection of the diluted clinical sample using multiantibodies
and a single antibody. c) Crumpled GFET sensor with a schematic illustration of the binding process. d) Anti-foiling mechanism of the GFET sensor:
Electrostatic actuation enables probes to capture target analytes, while rigid DNA bases prevent nonspecific absorption on the graphene. (a, b) Adapted
with permission. [] Copyright  American Chemical Society. (c) Adapted with permission. [ ] Copyright , Wiley-VCH GmbH. (d) Reproduced
under the terms of CC BY license. [] Copyright , The Authors, published by Springer Nature Limited.
 s (Figure b). Moreover, the enzyme-modified device exhib-
ited a long-term stability of .% and good device-to-device re-
producibility with an RSD of .%, indicating its high potential
for practical applications. Nevertheless, the covalent attachment
of enzymatic bioreceptors on the graphene surface exhibits sev-
eral drawbacks, including disrupted folding of bioreceptors, ex-
cess active sites, damage to the sensing materials, and so on.[, ]
To address this issue, research eorts have been devoted to de-
veloping nonenzymatic GFET biosensors for the sensitive detec-
tion of small biomolecules.[– ] For example, Danielson et al.
demonstrated a GFET lactose sensor based on gold nanoparti-
cles and specific protein receptors, which was able to detect as
low as × analyte over a dynamic range from  to
 mol L.[ ] Similarly, Ku et al. fabricated a GFET sen-
sor by immobilizing the cortisol monoclonal antibody (C-Mab)
onto the surface of graphene.[ ] In this design, graphene acted
as a transducer that converted antibody-analyte binding interac-
tions into electrical signals. Owing to the high stability of C-Mab
(-hour storage at room temperature), the as-prepared GFET
sensor can be integrated into a prototypical system that enables
continuous and noninvasive cortisol monitoring of the human
body (Figure c). The sensor exhibited an LoD of  pg mLcor-
tisol in various biological fluids, which is low enough to measure
the cortisol concentration in human tears (Figure d).[ ]
3.4. Cell
Cellular living microenvironments contain a wide range of
biomarkers, including enzymes, nucleic acids, proteins, ions,
small organic molecules, etc.[– ] The monitoring of cellu-
lar activity and products provides key information associated
with cancer, biological reactions, and genetic processes.[,,]
This section summarizes advances in GFET biosensors for
achieving in vitro detection of neural potential and cellular
products.
Research progress in GFET cell sensors has been made for
monitoring extracellular or intracellular potential signals.[, ]
For example, Dupuit et al. combined solution-gate GFETs with a
multicompartment microfluidic chip for multimodal recording
of neuron electrical activity (Figure 7a).[ ] The GFETs provided
a highly sensitive and transparent sensing area, while the fluidic
microchannels, synaptic, and somatic chambers helped demon-
strate the spontaneous activity of neuron networks. The spike
signals acquired by GFETs and the customized TiN MEA exhibit
several dierences (lower panels in Figure a). Although the
MEA can resolve the first spikes, it had diculty in extracting
secondary spikes from the background noise due to the low
amplitude ability. By comparison, the GFETs realized threefold
higher detection eciency to enable continuous mapping of
neural network topology.[] In addition, Yang et al. detected
weak neural potential signals directly in a complex bioenviron-
ment by combining GFETs with covalent organic frameworks
(COFs).[] In this study, murine neurons were cultured di-
rectly on the graphene channel in medium for long-standing
measurements (Figure b). Due to the COF’s mesoporous
structure, the culture solution can permeate the COF, generate
synaptic potential signals, and finally arouse rhythmic transient
current changes. Moreover, the device exhibited impressive anti-
fouling capability with a high signal-to-noise ratio of . when
Adv. Funct. Mater. 2023,  ©  Wiley-VCH GmbH
2301948 (9 of 26)
16163028, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/adfm.202301948 by Fudan University, Wiley Online Library on [11/05/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.afm-journal.de
Tabl e 2 . Sensing Performance of GFET Protein Sensors.
Analyte Receptor LoD Detection time [s] Condition Ref.
A𝛽protein antibody pg mL> . m PBS []
A𝛽protein antibody . pg mL PBS/Human plasma/aCSF []
Anti-diuretic hormone aptamer . ag mL  m PBS, serum []
Anti-p p antigen × – ×PBS []
BGP-C antibody  fg mL fg mL pg mL>  m PB .% serum% serum []
CD antibody  particles μL .×PBS []
cTnI aptamer . × (. pg mL) N/A PBS, serum []
Cytokine aptamer . ×  ×PBS, sweat []
Ferritin antibody × – .×PBS []
GFAP antibody  fg mL fg mL< ×PBSHuman plasma []
HBsAg antibody ×  serum []
HBsAg antibody × < PBS, serum, saliva []
Hemin catalyst × beef []
Hemoglobin aptamer . × . × . × – ×PBSSerumurine []
Hepatitis C virus core protein aptamer . × N/A reaction buer []
HPV- E aptamer . × PBS, saliva []
IFN-𝛾aptamer . ×  ×PBS, human sweat []
IFN-𝛾aptamer . × N/A biofluids []
IgE aptamer . ×  .×PBS []
IL- aptamer . × < ×PBS []
IL- aptamer . × N/A×PBS, / serum []
IL- aptamer . × N/A biofluids []
IL- aptamer . × < ×PBS, human saliva []
Insulin aptamer . × N/A×PBS, / diluted serum []
Insulin antibody ×  buer []
Ligand odorant receptor ×  ×PBS []
MMP- polypeptide × . ×< buerplasma []
Neuropeptide Y aptamer ×  artificial sweat []
Neuropeptide Y aptamer × < .×PBS []
NT-proBNP antibody pg mL PBS []
PSA antibody pg mLN/A serum []
PSA aptamer ×< PBS []
SARS-CoV- spike antibody antigen . fg mL(. × )  serum []
SARS-CoV- spike antibody antigen . × seconds UTM []
SARS-CoV- antigen antibody × N/A PBS []
SARS-CoV- antigen antibody fg mL ×PBS []
SARS-CoV- antigen aptamer . PFU mL< ×PBS []
SARS-CoV- N protein antibody  ag mL< buer []
SARS-CoV- spike protein antibody fg mL(. × ) seconds PBSUTM []
SARS-CoV- spike protein antibody × seconds UTM []
SARS-CoV- spike protein antibody  pg mL(. ×– ) < artificial saliva []
SARS-CoV- spike protein antibody × N/A.×PBS []
SARS-CoV- spike protein antibody × N/A PBS []
SARS-CoV- spike protein antibody ag mL. ×PBS []
SARS-CoV- spike protein Antibody fg mL. PBS []
SARS-CoV- spike protein Antibody fg mLN/A PBS []
SARS-CoV- spike protein Antibody fg mLseconds ×PBS, artificial saliva []
streptavidin pyrene–PEG ×< Trizma buer []
Thrombin Aptamer . ×  .×PBS []
(Continued)
Adv. Funct. Mater. 2023,  ©  Wiley-VCH GmbH
2301948 (10 of 26)
16163028, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/adfm.202301948 by Fudan University, Wiley Online Library on [11/05/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.afm-journal.de
Tabl e 2 . (Continued).
Analyte Receptor LoD Detection time [s] Condition Ref.
Thrombin Aptamer ×  .×PBS []
Thrombin Aptamer × < ×TRB []
TNF-𝛼Aptamer . × N/A biofluids []
TNF-𝛼Aptamer . ×  ×PBS []
t-Tau Antibody . pg mL PBS/Human plasma/aCSF []
t-Tau Antibody  fg mLN/A ×PBS []
Zika protein Antibody . ×  ×PBS% serum []
Note: N/A, not available. aCSF, artificial cerebrospinal fluid. BGP-C, one type of the anti-bone Gla protein. cTnI, cardiac troponin I. GFAP, Glial fibrillary acidic protein. HBsAg,
hepatitis B surface antigen. IgE, immunoglobulin E. IL-, Interleukin . PB, phosphate buer. PBS, phosphate-buered saline. PSA, prostate specific antigen. UTM, universal
transport medium. TNF, tumor necrosis factor. TRB, thorbim-binding buer.
Figure 6. GFET small biomolecular sensors. a,b) GFET acetylcholine sensor: a) Schematic illustration of the surface functionalization and the
acetylcholinesterase-catalyzed hydrolysis of acetylcholine. AchE, acetylcholinesterase. PABA, poly(-amino-benzylamine-co-aniline) film. b) Flow re-
sponse of the sensor at dierent analyte concentrations ( μLmin
,Vgs =−. V, Vds =. V). c,d) GFET cortisol sensor system: c) A prototype
integrated with GFET, resistor, capacitor, D printed interconnect, and rigid island for signal acquisition and transmission. d) Sensor response to cortisol
concentrations in the buer and artificial tear solvent. (a, b) Adapted with permission. [ ] Copyright , Elsevier B.V. (c, d) AReproduced under the
terms of CC BY-NC license. [ ], Copyright , The Authors, published by the American Association for the Advancement of Science.
operating in  ppm PEDOT:PSS or complex fouling bioenvi-
ronments (Figure c).[]
Advantages in GFET cell sensors have also been demonstrated
in the sensitive detection of cellular products.[, ] For instance,
Zhang et al. developed an electrolyte-gate GFET managed us-
ing ultrahigh frequencies (UHF) to enable direct detection of
biomarkers in high-salt solutions (Figure d,e).[] To o vercom e
the universal Debye screening eect, the developed GFETs were
operated in reflectometry mode (UHF, GHz) and achieved
an LoD of × streptavidin (– orders of magnitude
lower than conventional dielectric-modulated Si-based FET sen-
sors). Thus, the UHF GFET sensor was able to record the ac-
tion of  cells mmcardiomyocytes cells cultured on the
graphene channel (Figure e). Notably, the UHF reflectometry
detection scheme provides a solution to fully overcome the high-
ionic screening eect (i.e., Debye screening eect) in GFET-
based biomarker detection applications.[, ] Most recently, Lei
et al. demonstrated a photocatalysis-induced renewable reduced
graphene oxide nanosheet FET (rGON-FET) biosensor for real-
time recording of extracellular activity.[] The sensing chan-
nel comprised a sandwich structure of rGON, TiO-rGO, and
Fluo-AM molecules that are specific to Ca+(Figure f). Be-
cause it exports one Ca+from the cell at the expense of im-
porting three Na+, the as-prepared rGON-FET achieved second-
level detection of Na+concentrations with a remarkable response
of .% (Figure g).[] Moreover, the photo-responsive self-
cleaning TiOenabled regenerative detection of such exocytotic
Ca+in living cells.[ ]
In short, GFET biosensors have exhibited major advantages
in wide-range biomarker detection applications. Noticeably, the
Adv. Funct. Mater. 2023,  ©  Wiley-VCH GmbH
2301948 (11 of 26)
16163028, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/adfm.202301948 by Fudan University, Wiley Online Library on [11/05/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.afm-journal.de
Figure 7. GFET cell sensors. a) Schematics of the neuron-gated GFET and GMEA, showing the sensor results of neuronal spikes acquired from the
GFET and the GMEA, respectively. b,c) GFET neural sensor: b) Optical microscope image of nerve cells cultured on a graphene channel. c) Sensor
response upon neural electrical signals and  ppm PEDOT:PSS. d,e) Ultrahigh frequency GFET sensor for the detection of cell action: d) Schematic
of the binding process on the graphene surface, where a pyrene linker group was attached to peptide E (i.e., (EIAALEK)). e) Detection of the unique
contractility information of cardiomyocyte cells. Inset: Illustration of a cardiomyocyte cell on GFETs. f,g) The rGON-FET biosensor for cellular Ca+
detection: f) Schematics of the sensing principle, in which high Na+solution stimulates, the Na+/Ca+exchanger opens and Ca+euxes. g) Real-time
detection of Na+released from living cells. Inset: Optical image of human umbilical vein endothelial cells (HUVECs) cultured on the device. (a) Adapted
with permission. [ ] Copyright , Wiley-VCH GmbH. (b, c) Adapted with permission. [] Copyright , WILEY-VCH Verlag GmbH & Co. KGaA,
Weinheim. (d, e) Adapted with permission. [] Copyright , Wiley-VCH GmbH. (f, g) Adapted with permission. [ ] Copyright , Elsevier Ltd.
sensing performance depends largely on the interfacial design
and surface chemistry because most recognition processes and
signal transduction occur at the graphene surface. Thus, further
studies should focus on probe selection and surface functional-
ization strategies to improve the above-mentioned performance
metrics of GFET biosensors, which will accelerate their practical
applications from lab to fab.
4. Prototypical Applications
The healthcare market has witnessed notable progress in GFET-
based sensor systems, including wearable, biomimetic, and
point-of-care electronics.[,, ] In this section, we summarize
the latest advances in fabrication techniques and prototypical
applications of GFET sensor systems. The challenges of these
sensor systems and the possible technical solutions are also
analyzed.
4.1. Wearable Sensor Systems
Wearable sensors attached to the human body can mea-
sure physiological biomarkers for continuous healthcare
monitoring.[,,, ] Among existing wearable sensor sys-
tems, GFETs have demonstrated major advantages such as fast
response speed, reliable material quality, high compatibility
with flexible substrates, and large deformations on human
skin.[,, ] Furthermore, they are capable of detecting various
disease indicators in complex biological fluids and monitoring
human activity in real time.
Attempts in wearable sensors have also been made in health
monitoring by the detection of wide-range biomarkers.[, ] For
example, Gao et al. demonstrated a skin-based flexible gel elec-
trolyte graphene transistor for human sweat monitoring directly
on the skin (Figure 8a).[ ] The constructed GFET epidermal
sensor realized noninvasive and continuous detection of glucose
with a dynamic range from ×to . ×.[ ] In addi-
tion, Sheibani et al. reported a wearable sensory electronic chip
based on a platinum/graphene aptamer extended gate FET for
the identification of cortisol in biological buers.[] The device
has a LoD of × in physiological fluids with cortisol
concentrations of ×to ×. Although these GFET
biosensors enable sensitive detection of various biomarkers, their
single-analyte detection ability is not sucient for practical appli-
cations.
Adv. Funct. Mater. 2023,  ©  Wiley-VCH GmbH
2301948 (12 of 26)
16163028, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/adfm.202301948 by Fudan University, Wiley Online Library on [11/05/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.afm-journal.de
Figure 8. GFET-based wearable sensor systems. a) A wearable GFET sensor incorporated with gel electrolyte for noninvasive glucose monitoring. b)
Ultraflexible GFET biosensors with a schematic illustration of aptamer functionalization on the graphene surface (upper panel). Photographs of the
flexible device conformably attached onto the finger and artificial eyeball (lower panel). c,d) Intelligent aptameric DGTFET wireless biosensing device: c)
Image of the DGTFET device worn on the participant’s forehead. The four wires in the left panel are the ) sensing channel signal, ) ground operation,
) gate voltage, and ) reference channel signal. d) Time-resolved measurement of TNF-𝛼in human sweat. e–g) GFET-based eyeball sensor systems:
e) The sensor fits the finger entirely. f) Image of the GFET sensor attached to an artificial eye. g) ΔVDirac as a function of the L-cysteine concentration in
artificial tears. Error bars are determined by the standard deviation of measurements. (a) Adapted fwith permission. [ ] Copyright , Elsevier B.V.
(b) Reproduced under the terms of CC BY license.[ ] Copyright , The Authors, published by MDPI. (c, d) Adapted with permission. [ ] Copyright
, Wiley-VCH GmbH. (e–g) Adapted with permission.[ ] Copyright , Wiley-VCH GmbH.
To address this issue, Hao et al. developed a wearable GFET
sensor based on a polyethylene terephthalate substrate for
minute-level detection of multiplexed cytokine biomarkers, in-
cluding IFN-𝛾,TNF-𝛼and IL-.[ ,] The sensor maintains
its sensitivity and response after withstanding large deforma-
tions on the human finger or an artificial eye (Figure b). Inte-
grated with an Android platform, the GFET sensor enables re-
liable signal acquisition and transmission in on-site detection
of clinical samples from asymptomatic or mild COVID- pa-
tients (Figure c). The sensor platform enabled multiplexed de-
tection of IFN-𝛾,TNF-𝛼and IL- with LODs of . × ,
. ×, and . × , respectively (Figure d). No-
tably, the testing accuracy, device reliability, and operation cost
of such sensor systems require further evaluation before appli-
cations for population-wide tests.[, ] More recently, Huang et al.
developed ultraflexible and transparent GFET sensor platforms
attached to artificial eyes for continuous L-cysteine monitoring in
the human body (Figure e,f).[ ] After -cycle deformations of
fold (°), bend (radii  μm), and shrink (%) recovery, this
portable sensor system still enables consistent and reliable detec-
tion of . × L-cysteine in artificial tears (Figure g).[ ] In
addition, the sensor platforms based on the WO/Au/WOelec-
trode exhibit negligible influence on human eyesight, compared
with devices covered by conventional Au electrodes.
Advantages have also been demonstrated in GFET
biosensors to monitor human activity and control robotic
movement.[,, ] In , Park et al. reported a wearable
GFET pressure sensor for breath and heart rate detection fab-
ricated by using vertically aligned, position- and size-controlled
arrays of ZnO nanotubes grown on a graphene layer.[] The
Adv. Funct. Mater. 2023,  ©  Wiley-VCH GmbH
2301948 (13 of 26)
16163028, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/adfm.202301948 by Fudan University, Wiley Online Library on [11/05/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.afm-journal.de
graphene layer acts as a substrate for the catalyst-free growth
of ZnO nanotubes and serves as a flexible conduction channel
connecting the ZnO nanotubes to the metal electrodes. This
wearable pressure sensor based on the hybridization of D ZnO
nanotubes with D graphene films shows a high-pressure sensi-
tivity (. kPa) and can detect weak flows of inert gases. The
nanotube sensor can be used to monitor respiration and heart
rate when attached to the volunteer’s mid-chamber and wrist.[ ]
Similarly, Paul et al. developed a wearable GFET pressure sensor
based on crumpled graphene flake network channels for robotic
hand-free control.[ ] The device was fixed to the temple area of
the face and controlled by the respective temporal muscles.[ ]
4.2. Biomimetic Sensor Systems
Biomimetic and bioinspired sensing devices have attracted ex-
tensive attention over the past few decades.[– ] They are the
key component for intelligent healthcare systems in our everyday
life.[,, ] Here, we summarize the latest advances in GFET-
based biomimetic sensor systems, including bioelectronic eyes,
noses, tongues, etc.
Attempts have been made in GFET sensor systems for devel-
oping electronic noses or tongues.[– ] Electronic noses can
convert interactions with odor molecules into electrical signals,
such as potential changes and surface charge modulation. Al-
though the concept of electronic noses (e-noses) dates back to
the s, their applications were limited by misbinding infor-
mation from odor changes in mixtures.[ ] To address this issue,
Hayasaka et al. developed a GFET-based e-nose to obtain high se-
lectivity under complex conditions (Figure 9a). This GFET elec-
tronic nose recorded the gas-sensitive conductivity profile and
decoupled it into four unique physical parameters. These pa-
rameters were further projected onto a feature space as four-
dimensional output vectors, which were associated with dierent
target gases by machine learning analysis. As a demonstration,
the combination of this GFET e-nose with trained pattern recog-
nition algorithms enables almost % accuracy when testing
water, methanol and ethanol vapors individually (Figure b).[ ]
By comparison, electronic tongues generally detect taste-related
biomarkers in biological fluids (Figure c).[– ] For exam-
ple, Ahn et al. demonstrated a duplex bioelectronic tongue
based on GFETs that were functionalized with specific recep-
tor nanovesicles (Figure d).[, ] The bioelectronic tongue
was able to detect as low as  aM monosodium glutamate
within  s (Figure e), paving the way to identify emerging
artificial tastants in food. Very recently, Lee et al. developed a
solution-gated GFET sensor functionalized with Nafion films or
gold nanoparticles.[ ] These gold nanoparticles allow glucose
molecules to attach to the graphene surface more eectively than
other substances acting as receptors. Thus, the sensor can detect
glucose molecules as low as  with a response/recovery
time of / s. Owing to its sensitive and rapid detection abil-
ity, this GFET electronic tongue was used to identify four real
beverages (orange juice, original coke, Sprite and Zero Coke), in-
dicating its promising potential in practical applications.[ ]
Advances have also been reported in developing bioelectronic
eyes based on GFET sensor systems.[– ] The human eye
perceives vision by converting light intensity into action poten-
tials through photosensitive receptors.[, ] Thus, artificial elec-
tronic eyes require high-performance bioengineered light re-
sponse elements for visual signal acquisition and conversion. For
example, Yang et al. demonstrated a phototransistor sensing sys-
tem for visual perception imaging by coupling ontogenetically
engineered living cells on a graphene channel.[ ] Engineered
living cells have photosensitive ion channels that convert light
intensity into bioelectrical signals. This process is similar to the
light pattern-to-action potential conversion process triggered by
the photoreceptors of retinal cells, making the phototransistor
imaging process similar to the human visual process. As an ap-
plication, the system can record the photocurrent changes at dif-
ferent locations of the original image and then recover a  ×
pixel image.[ ]
Apart from bioelectronic organs, mapping brain activity is
equally important for fitness monitoring fields.[, ] Neural
mapping is generally based on noninvasive techniques such
as magnetoencephalography and electroencephalography for
recording or stimulation.[ ] However, these noninvasive tech-
niques do not provide suciently accurate and interpretable neu-
ral signals due to the filtering of the human brain skull, prompt-
ing eorts to develop the next generation of electronic brains.
In addition, conventional noninvasive techniques are severely
hampered by current microelectrode materials when recording
ultralow or sublow brain activity (<. Hz).[ ] To address this
issue, Graaido’s group developed extra and intracortical arrays
for ultralow neural recording based on graphene solution-gated
field-eect transistors (gSGFETs), which are capable of record-
ing neural signals from infralow (<. Hz) frequencies to typ-
ical local field potential bandwidths (Figure f).[] Similar to
solution-filled glass microtubes, such gSGFETs can work with-
out the constraints of spatial sampling, thus oering the possi-
bility of spatially resolved mapping.[] Shortly after, this group
further advanced gSGFET technology by integrating a single
gSGFET to build a -channel sensing array.[,] The sens-
ing array was used to monitor the local field potential of the
cortex in a freely moving rat model while monitoring its D
position over a long period of time up to  h (Figure g).
This work demonstrates the maturity of the technology in terms
of its long-term and broadband recording capabilities in freely
moving animals.[ ] Most recently, the group developed im-
plantable graphene depth neural probes (gDNPs) based on sg-
GFETs and applied them to epilepsy research.[ ] The gDNPs
consist of a linear array of graphene microtransistors. To en-
able them to penetrate the mouse cortex and reach the hip-
pocampus without flexion, silk fibroin was used to temporar-
ily harden the flexible gDNPs. The gDNPs could reliably record
and map high spatial resolution seizures, preictal DC shifts and
seizure-related diusion depolarisations, as well as high fre-
quencies through the cortical layers to the hippocampus.[] In
short, these studies demonstrate the potential of graphene tran-
sistor technology to reveal seizure-related mechanisms in the
awake brain in vivo, advance the application of invasive tech-
niques for mapping brain activity, provide a reliable tool for
future applications in neuroscience and biomedical engineer-
ing, and pave the way for the development of the electronic
brain.
Adv. Funct. Mater. 2023,  ©  Wiley-VCH GmbH
2301948 (14 of 26)
16163028, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/adfm.202301948 by Fudan University, Wiley Online Library on [11/05/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.afm-journal.de
Figure 9. GFET-based biomimetic sensor systems. a,b) GFET bioelectronic nose: a) Schematic of the smell sensor system comprising a cap chamber, a
GFET test chip, an IC socket, a casing, and BNC connector ports. b) Confusion matrix of multiclass classification using the as-prepared sensor systems.
c–e) GFET bioelectronic tongue: c) Schematic of an artificial tongue and d) the device configuration. e) Real-time response toward umami and sweet
tastants using the sensor systems. MSG, monosodium glutamate. f,g) GFET neural sensor systems: f) Device configuration of gSGFETs enabling ultralow
frequency neural recording. g) Illustration of a rat implanted with an untethered recording system. The neural signals transmitted by the gSGFET array
sensors are digitized and transmitted wirelessly to a signal receiver, which is connected to a computer for signal recording. The right lower panel shows
a photograph of the gSGFET array placed on the rat cortex. (a, b) Reproduced under the terms of CC BY license. [ ] Copyright , The Authors,
published by MDPI. (c) Reproduced under the terms of CC BY-NC license. [ ] Copyright , The Authors, published by American Association for
the Advancement of Science. (d, e) Adapted with permission. [ ] Copyright , American Chemical Society. (f ) Reproduced with permission. []
Copyright , The Authors, under exclusive license to Springer Nature Limited. (g) Reproduced under the terms of CC BY license. [ ] Copyrights
, The Authors, published by Springer Nature.
4.3. Point-of-Care Diagnostics
The past few decades have witnessed a remarkable tech-
nical revolution in point-of-care (POC) diagnostic tools for
home-care health monitoring applications.[,, ] Among these
emerging POC technologies, GFET biosensors have exhib-
ited major advantages such as high sensitivity, biocompatibil-
ity, multifunctionality, ultrashort response time, and integrated
capabilities.[,, ] Corporated with electrical readouts and dig-
ital chips, GFET-based POC sensor systems hold great potential
for next-generation field-deployable sensing tools in resource-
poor settings.[,, ] This section summarizes the latest ad-
vances in GFET POC sensor systems for the detection of cancer
biomarkers, pathogenic bacteria, and infectious viruses.
4.3.1. Cancer Biomarker
Cancer screening requires the sensitive detection of low-
abundance biomarkers at an early stage.[,, ] Research eorts
have been made in GFET biosensors for rapid, low-cost, multi-
plexed detection of cancer-related small biomolecules.[, ] In
, Hao et. al. demonstrated aptamer-functionalized GFETs
based on a HfOburied gate for cytokine detection.[ ] With
Adv. Funct. Mater. 2023,  ©  Wiley-VCH GmbH
2301948 (15 of 26)
16163028, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/adfm.202301948 by Fudan University, Wiley Online Library on [11/05/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.afm-journal.de
Figure 10. GFET-based cancer and pathogenic sensor systems. a,b) GFET-based portable cytokine sensor systems: a) Photograph of the portable sensor
system consisting of signal transduction circuits, sample well, signal processing circuits, amplifier and regulate circuits, WiFi module, and LCD screen.
b) Detection of interleukin- in human saliva solutions from dierent individuals using the GFET portable system. c) Schematic of GFET EV-Chip (left
panel) and the detection of blood-derived exosomes from plasma samples (right panel). d–f) An ABX-GMFET potable sensor system from the d) front
view and e) top view. f) The extended photograph demonstrates an ABX-GMFET microfluidic on an electrode, in which the target bacteria flow. (a, b)
Adapted with permission. [ ]. Copyright , Elsevier B.V. (c) Adapted with permission. [] Copyright , Wiley-VCH GmbH. (d–f ) Adapted with
permission. [ ] Copyright , Elsevier B.V.
the aid of customized printed circuit boards, they developed
POC GFET sensor systems that visualize cytokine concentra-
tion changes via a smartphone or cloud server (Figure 10a).
This nanosensing system can detect as few as . ×
interleukin- molecules within  s (Figure b), indicating its
unique potential for noninvasive early diagnosis and long-term
healthcare applications. Similarly, the same group developed an-
other polyethyleneimine-based aptamer GFET for minute-level
detection (– min) of hemoglobin in undiluted biological flu-
ids, with detection limits down to . × (in serum) and
. × (in urine).[ ] In , Mandal et al. demonstrated
a portable GFET sensor platform with a computationally opti-
mized coplanar gate electrode for PSA detection.[ ] Integrated
with customized readout modules and a rotating disk microflu-
idic device, the GFET sensor platform achieved an LoD of ×
 mLPSA in serum with a dynamic range up to ×
gmL
.[ ] Moreover, the platform exhibited sucient speci-
ficity to nontarget interferers induced by BSA and IgG molecules,
which is critical to medical diagnosis in future home-care tests.
Research progress has also been made in exosome detec-
tion via GFET biosensors.[,, ] Exosomes released from can-
cer cells contain specific information that is of vital importance
for precise diagnosis.[ ] In , Hajian et al. demonstrated
an exosome specific GFET termed EV-chip based on antibody-
Adv. Funct. Mater. 2023,  ©  Wiley-VCH GmbH
2301948 (16 of 26)
16163028, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/adfm.202301948 by Fudan University, Wiley Online Library on [11/05/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.afm-journal.de
functionalized graphene (Figure c).[] The EV-chip works by
combining the specificity of antigen–antibody interactions and
the high electrophoretic mobility of exosomes, which allows real-
time exosome analyte quantification in blood. The device LoD
reached ×particles mLand the quantification limit was
as low as ×particles mL. Notably, GFET sensor systems
consisting of a picoammeter, potentiostat, multiplexer, micropro-
cessor controller, and serial interface weigh less than kg, mak-
ing them a potential POC tool for clinical tests.[ ] Although the
integrated EV-chip exhibited several advantages in exosome de-
tection, the insucient sensitivity might lead to poor diagnostic
accuracy in population-wide applications. To address this issue,
Ramadan et al. developed a GFET exosome sensor system by dop-
ing carbon dots on the graphene surface to enhance sensitivity.[]
The incorporated carbon dots allowed the formation of a compat-
ible heterogeneous structure on sp-hybridized graphene, thus
leading to improved recognition eciency. As a demonstration,
the carbon-dot-enhanced GFET biosensors successfully detected
exosomes down to  particles μL, approximately orders of
magnitude higher than conventional exosome sensors.[, ] No-
tably, research eorts should be devoted to simplifying the op-
eration procedure and shortening the sample-to-answer time for
ideal user-friendliness.
4.3.2. Pathogenic Bacteria
Human pathogenic bacteria are disease-causing bacteria
that can lead to many serious diseases, epidemics, and
pandemics.[,,, ] There is an ever-lasting demand to
eectively detect pathogenic bacteria in various environmental
conditions such as water, food, soil, and air.[ ,] Unfortunately,
conventional assays with sophisticated operation depend largely
on specialist sta and expensive instruments, which take several
hours or days to identify target bacteria in unprocessed biological
samples.
Research eorts have been devoted to developing POC tools
that enable on-site detection of antibiotic-resistant pathogenic
bacteria in complex samples.[, ] For instance, Kim et al. devel-
oped a two-channel GFET-based portable device (ABX-GMFETs)
for the real-time detection of gram bacteria with an LoD down
to CFUmL
(Figure d).[] The antibiotic-modified ABX-
GMFET was able to distinguish gram-positive/negative bacte-
ria as a result of the chemical moiety interaction between the
bioprobes and target bacteria. Furthermore, the dual microflu-
idics incorporated with the platform minimized human inter-
vention and then facilitated automation in practical applications
(Figure e,f).[] Although the ABX-GMFET can identify Gram
bacteria, it is not sucient for multianalyte detection on a self-
contained chip.[, ] To this end, Kumar et al. demonstrated a
peptide-modified GFET sensor platform for -minute and multi-
analyte detection of antibiotic-resistant bacteria.[ ] The authors
dramatically reduced the LoD to cells mLby employing di-
electrophoresis in GFETs. Additionally, two separate sample wells
in this sensor platform allowed the simultaneous detection of two
dierent bacteria, Staphylococcus aureus and antibiotic-resistant
Acinetobacter baumannii.[ ]
Research in GFET POC systems has also realized the sensitive
detection of bacterial products.[, ] In , Lin et al. achieved
-ppb-level detection of indole gas molecules, a metabolite prod-
uct of Escherichia coli (E. coli).[] This strategy introduced an-
other way to develop bacterial sensor systems by monitoring tar-
get gas products, which would make sense in food safety moni-
toring fields. By comparison, Tan et al. proposed a GFET-based
sensor system for continuous and real-time monitoring of E.
coli.[ ] In this configuration, graphene was functionalized us-
ing two-terminal tail spike proteins, of which the N-terminal
domain can be anchored by -ethyl--(-dimethylaminopropyl)-
carbodiimide/N-hydroxy-succinimide and the C-terminal do-
main can bind to E. coli cellular lipopolysaccharide. Thus, the
as-prepared GFET sensor system was able to detect E. coli
with a sensitivity of . mV per bacteria.[ ] Most recently,
Burch and coworkers reported an aptamer-based GFET platform
that enables simultaneous detection of three dierent opioid
metabolites (i.e., noroxycodone, norfentanyl, and -ethylidene-
,-dimethyl-,-diphenylpyrrolidine).[ ] The authors placed
four isolated sets of GFETs with a Pt reference electrode on a
single chip (. cm ×. cm) to functionalize separately and re-
alize multianalyte detection simultaneously. In addition, the de-
vice LoD reaches .–. × gmL
for these opioid metabo-
lites, which is orders of magnitude lower than previous costly
laboratory-based techniques.
4.3.3. Neurodegenerative Disease
Neurodegenerative diseases refer to a heterogeneous group of
disorders in the central or peripheral nervous system, which
commonly include Alzheimer’s disease (AD), Parkinson’s dis-
ease, schizophrenia, and others.[,– ] Among these diseases,
AD is a widely investigated neurodegenerative disease that ac-
counts for % of all dementia and aects the quality of human
life.[– ] Early diagnosis of AD is of vital importance because
there is no certain treatment after the lesion has progressed in
the late stage.[, ] In , Park et al. demonstrated a dual-
antibody-modified GFET biosensor for multiplex detection of two
AD biomarkers (Figure 11a).[] In this configuration, a cus-
tomized jig was combined with GFETs and an Ag/AgCl refer-
ence electrode to monitor the output signals of each biomarker
(Figure b). The as-prepared platform realized sensitive detec-
tion down to . pg mLtarget proteins in biofluids (human
plasma and artificial cerebrospinal fluid). Owing to the dierent
isoelectric points and surface charges of AD biomarkers (A𝛽-
and t-Tau proteins), the GFET biosensor exhibited high selectiv-
ity with a distinctive output signal for each analyte (Figure c).
More recently, Kwon et al. demonstrated a simplified, linker-
free, anti-tau antibody immobilization process to build GFET AD
biosensors (see Figure b).[] The linker-free antibody immobi-
lized on the patterned graphene achieved an LoD of  mLfg
Tau proteins in plasma. Apart from AD diagnostics, research ef-
forts have also been made in developing on-chip GFET biosen-
sors to detect neurological biomarkers in blood.[,, ] In ,
Xu et al. reported sensitive and ultrafast detection of glial fib-
rillary acidic proteins in patient plasma by using GFET biosen-
sor chips.[ ] Compared with commercial ELISA kits and single-
molecule array (Simoa) technology, the proposed GFET approach
exhibited competitive LoD down to  fg mLand a short
Adv. Funct. Mater. 2023,  ©  Wiley-VCH GmbH
2301948 (17 of 26)
16163028, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/adfm.202301948 by Fudan University, Wiley Online Library on [11/05/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.afm-journal.de
Figure 11. GFET-based Alzheimer sensor systems. a) Schematic illustration of the GFET platform for multiplex detection of Alzheimer’s biomarkers
(A𝛽- and t-Tau). b) Device configuration in front and side views. c) Selectivity of the GFET platform toward pivotal Alzheimer biomarkers. (a–c)
Adapted with permission. [ ] Copyright , Elsevier B.V.
sample-to-answer time of < min, indicating its great promise
for POC diagnosis applications.[, ]
4.3.4. Infectious Virus
Epidemics caused by infectious viruses are often characterized by
rapid transmission, wide dispersal and high public risk.[– ]
The primary step for epidemic prevention and control is rapid
and accurate diagnosis of presymptomatic and asymptomatic in-
dividuals who have low viral loads.[– ] This section summa-
rizes the notable progress in GFET-based POC systems for sen-
sitively detecting viruses such as Zika,[ ] Ebola,[ ] SARS-CoV-
,[ ] hepatitis C,[ ] tuberculosis,[] and others.[]
Early advances have been made in antibody-modified GFET
biosensors for amplification-free detection of infectious viruses
in unprocessed samples.[, ] At the very beginning of the
COVID- outbreak, Seo et al. invented antibody-modified
GFETs enabling successful detection of SARS-CoV- in universal
transport medium (LoD: . ×pfu mL) and clinical sam-
ples (. ×copies mL).[ ] Additionally, Piccinini et al.
developed a portable GFET sensor based on monoclonal an-
tibodies for the detection of SARS-CoV- spike protein (LoD:
. × ).[ ] Although these single antibody-modified sen-
sor platforms realized rapid SARS-CoV- detection within min-
utes, the testing accuracy (<%) was limited because of poten-
tial antigen escape from a single captured binding site.[, ] To
prevent potential viral escape mutants, Dai et al. demonstrated
a multiantibody-modified strategy to achieve highly precise anti-
gen pool testing (Figure 12a).[] In this strategy, the SARS-CoV-
antigen was locked in a tight “antigenantibody complex” cap-
tured by three dierent antibodies, thus leading to an LoD down
to . copy μLin clinical samples and a diagnostic accu-
racy up to %. Owing to such satisfactory sensor perfor-
mance, the multiantibody-modified GFET sensor systems enable
-in- pooled screening tests with high detection throughput
(Figure b), which is key to understanding the clinical relevancy
between sensor information and population-wide tests.[, ]
Research attempts have also been made in aptamer-based
GFET sensor systems for on-site detection of SARS-CoV- nu-
cleic acids. In , Park et al. combined the sensitive end-point
detection ability of crumped graphene with the amplification
capability of reverse transcriptase loop-mediated isothermal
amplification technology to develop a portable GFET sensor
system (Figure c). This method took advantage of crumped
graphene to adsorb SARS-CoV- nucleic acids, thus leading to
an LoD of  μLSARS-CoV- in universal transport medium.
Furthermore, the GFET sensor system can dierentiate COVID-
-positive/negative samples with % accuracy, demonstrating
its potential in large-scale tests (Figure d,e). Most recently,
Wang et al. developed molecular electromechanical sys-
tems powered by LG-GFETs (MoIEMS-gFETs) for the direct
Adv. Funct. Mater. 2023,  ©  Wiley-VCH GmbH
2301948 (18 of 26)
16163028, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/adfm.202301948 by Fudan University, Wiley Online Library on [11/05/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.afm-journal.de
Figure 12. GFET-based infectious sensor systems. a,b) A portable GFET sensor prototype: a) Schematic of sample collection, treatment, and image of
the integrated platform. b) ΔIds/Ids versus t curve upon the addition of COVID- -in- pooled samples. c–e) GFET-based COVID- sensor systems:
c) Device configuration and surface functionalization of the GFET sensor systems. d) Responses of positive and negative clinical SARS-CoV- samples
on the crumpled GFETs. e) ROC curve analysis for positive and negative SARS-CoV- clinical samples. f–h) A MoIEMS-gFET prototype: f) Schematic
illustration of the device configuration and working principle. g) A probe is conjugated at the tip of the ss-DNA cantilever for specific recognition. Note:
E indicates an electric field. h) A prototype of the main system and the Mole-EMS GFET testing module. i) Ids/Ids versus time curve for the MoIEMS-
gFET when adding COVID--positive clinical samples. Inset: The average diagnosis time in clinical tests. (a, b) Adapted with permission. [] Copyright
, American Chemical Society. (c–e) Adapted with permission. [] Copyright , American Chemical Society. (f–h) Adapted with permission.[]
Copyright , The Authors, under exclusive license to Springer Nature Limited.
Adv. Funct. Mater. 2023,  ©  Wiley-VCH GmbH
2301948 (19 of 26)
16163028, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/adfm.202301948 by Fudan University, Wiley Online Library on [11/05/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.afm-journal.de
detection of SARS-CoV- RNA in nasopharyngeal swab samples
(Figure f–i).[] The MoIEMS was self-assembled from four
DNA strands consisting of a flexible arm and a rigid base. By
immobilizing the MoIEMS onto the GFET, its rigid base helped
to avoid nonspecific adsorption of contaminants, while the
flexible arm could precisely modulate the molecular recognition
and signal conversion processes driven by an external electric
field, thus significantly enhancing the detection sensitivity
(Figure f,g). Critically, the MoIEMS-gFETs eliminated the
need for complex and time-consuming nucleic acid extraction
and amplification processes in neo-coronavirus nucleic acid
samples. As a demonstration, the system realized an LoD down
to  copies mLSARS-CoV- RNA and an average response
time of less than min when testing nasopharyngeal samples
from  patients with COVID- and  COVID--negative
controls (Figure i).[]
As shown above, impressive advances in GFET biosensors
have been demonstrated in prototypical applications, including
wearable devices, biomimetic electronics, and diagnostic plat-
forms. Nevertheless, the commercialization of such sensing sys-
tems is restricted by their clinical consistency with population-
wide human tests. Further investigation and enormous research
eorts are desired to make GFET biosensors suitable for the clin-
ical market, which will pave the way for future healthcare moni-
toring technologies.
5. Conclusion and Outlook
Emerging GFET biosensors with unique properties and ap-
pealing electronic applications have promising potential to pro-
mote future innovation in healthcare domains.[,, ] In Sec-
tions . and ., we reviewed that GFET biosensors could suf-
fer from Debye screening eects and foiling eects under com-
plex physiological conditions, leading to diculty in quantita-
tive detection applications. Accordingly, we summarize recent
progress in GFET biosensors, with discussions on materials,
bioreceptor design, fabrication techniques, surface functional-
ization, antifouling approaches, and proof-of-concept applica-
tions at the integrated level. Over the past decade, GFET-based
sensor systems have demonstrated wide-ranging applications
across wearable electronics, biomimetic devices, health monitor-
ing, and medical diagnostics. Despite rapid development, some
major concerns still exist before the commercialization of GFET
biosensors.[,, ] Here, we list several key translation challenges
and discuss potential research directions:
5.1. Standardization
Ensuring that GFET biosensors are both accurate and reliable is
of vital importance for their acceptance in clinical tests.[, ] This
goal requires standardization in fabricating individual sensors
with high-quality graphene materials and minimized device-to-
device variations.[, ] Great eorts have been made to develop
GFET sensor systems with uniform fabrication, functionaliza-
tion approaches and testing conditions. Few technologies, how-
ever, can meet medical standards, which include biosafety, time
eciency, high-throughput requirements, and easy-to-operate
demands.[, ] Additionally, the analytical operation procedures
of GFET biosensors dier from research groups, hampering the
process of their launch in the real world.[,, ] Thus, standardiza-
tion is essential for GFET biosensors to maintain performance
and qualify home tests in healthcare fields.
5.2. System Integration
A commercial sensor system requires the integration of a power
supply, sensing components, digital processing, and data com-
munication modules.[,, ] Such stand-alone systems enable
home-care health monitoring at the molecular level. Neverthe-
less, most GFET-based sensor prototypes in this Review focused
primarily on single measurements, and research eorts should
continue toward developing multifunctional platforms that can
capture unique information of diverse health biomarkers.[, ]
In combination with other technologies such as optogenetics and
gene-editing techniques, integrated GFET sensor systems will be
a comprehensive tool not only for fitness applications but also for
widespread biodefense applications.[, ]
5.3. Translation to the Healthcare Market
Recently, the global healthcare market has witnessed continuous
growth in biomarker monitoring applications.[, ] The success-
ful translation of GFET biosensors and prototypical demonstra-
tions to the medical community still faces several hurdles re-
lated to clinical operation. Foremost, fitness monitoring appli-
cations promise rich sensor information, which requires exten-
sive correlation studies and successful validation before market
acceptance.[,, ] To this end, advanced technologies such as big
data, cloud computing, artificial intelligence, and machine learn-
ing algorithms are necessary to take full advantage of diagnos-
tic information obtained by GFET biosensors. Another key con-
cern is to control environmental conditions that aect the sen-
sor performance in real-world tests. For example, external tem-
perature and moisture should be considered when constructing
and operating GFET-based sensor systems to ensure diagnos-
tic accuracy.[,,] Third, there is an ever-lasting need for user-
friendly healthcare systems that are accurate, cost-eective, sim-
ple to perform with minimal training, and require fewer testing
reagents at minimal risk of harm.[, ] These concerns indicate
the outstanding need for research eorts before clinical transla-
tion and commercial realization.
Overall, GFET-based sensor systems facilitate their practical
use, especially in healthcare fields, due to their ultrahigh sensi-
tivity and facile integration. They will be able to monitor a wide
range of health biomarkers with high quality, ultimately enabling
home-care fitness applications and personal diagnostics. Notably,
the successful lab-to-fab translation of these sensors requires
massive validation in understanding the clinical relevancy of sen-
sor information and developing universal assessment metrics in
large-scale human tests. Given the competitive and tremendous
research eorts in this field, we anticipate exciting developments
in GFET biosensors that will change the medical community and
improve people’s lives in the near future.
Adv. Funct. Mater. 2023,  ©  Wiley-VCH GmbH
2301948 (20 of 26)
16163028, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/adfm.202301948 by Fudan University, Wiley Online Library on [11/05/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.afm-journal.de
Acknowledgements
C.D., D.K., and C.C. contributed equally to this work. The authors
acknowledge financial support from the National Key R&D Program
of China (YFC), National Natural Science Foundation of
China (), Strategic Priority Research Program of the Chinese
Academy of Sciences (XDB), Chongqing Bayu Scholar Program
(DP) and Fudan University.
Conflict of Interest
The authors declare no conflict of interest.
Keywords
biomarkers, biosensors, diagnostics, field-eect transistors, graphene
Received: February , 
Revised: March , 
Published online:
[] J. R. Sempionatto, J. A. Lasalde-Ramirez, K. Mahato, J. Wang, W.
Gao, Nat. Rev. Chem. 2022,6, .
[] L. Gardner, K. Kostarelos, P. Mallick, C. Dive, M. Hadjidemetriou,
Nat. Rev. Clin. Oncol. 2022,19, .
[] E. MacLean, T. Broger, S. Yerlikaya, B. L. Fernandez-Carballo, M. Pai,
C. M. Denkinger, Nat. Microbiol. 2019,4, .
[] D. C. Mohr, M. Zhang, S. M. Schueller, Annu. Rev. Clin. Psychol.
2017,13, .
[] X. Shen, R. Kellogg, D. J. Panyard, N. Bararpour, K. E. Castillo, B.
Lee-McMullen, A. Delfarah, J. Ubellacker, S. Ahadi, Y. Rosenberg-
Hasson, A. Ganz, K. Contrepois, B. Michael, I. Simms, C. Wang, D.
Hornburg, M. P. Snyder, Nat. Biomed. Eng. 2023, https://doi.org/
./s---.
[] J. Kim, A. S. Campbell, B. E. de Avila, J. Wang, Nat. Biotechnol. 2019,
37, .
[] C.Dai,Y.Liu,D.Wei,Chem. Rev. 2022,122, .
[] J. Tu, R. M. Torrente-Rodríguez, M. Wang, W. Gao, Adv. Funct. Mater.
2019,30, .
[] Q. Fu, L. Yu, Y. Wang, P. Li, J. Song, Adv. Funct. Mater. 2022,33,
.
[] D. Tyagi, H. Wang, W. Huang, L. Hu, Y. Tang, Z. Guo, Z. Ouyang, H.
Zhang, Nanoscale 2020,12, .
[] N. Wongkaew, M. Simsek, C. Griesche, A. J. Baeumner, Chem. Rev.
2019,119, .
[] J.Wang,C.Jiang,J.Jin,L.Huang,W.Yu,B.Su,J.Hu,Angew. Chem.,
Int. Ed. 2021,60, .
[] H. Xiong, J. Gao, Y. Wang, Z. Chen, M. M. Chen, X. Zhang, S. Wang,
Analyst 2019,144, .
[] L. Wang, X. Wang, Y. Wu, M. Guo, C. Gu, C. Dai, D. Kong, Y. Wang,
C.Zhang,D.Qu,C.Fan,Y.Xie,Z.Zhu,Y.Liu,D.Wei,Nat. Biomed.
Eng. 2022,6, .
[] C. Dai, M. Guo, Y. Wu, B. P. Cao, X. Wang, Y. Wu, H. Kang, D. Kong,
Z. Zhu, T. Ying, Y. Liu, D. Wei, J. Am. Chem. Soc. 2021,143, .
[] H.Youse,A.Mahmud,D.Chang,J.Das,S.Gomis,J.B.Chen,H.
Wang, T. Been, L. Yip, E. Coomes, Z. Li, S. Mubareka, A. McGeer, N.
Christie, S. Gray-Owen, A. Cochrane, J. M. Rini, E. H. Sargent, S. O.
Kelley, J. Am. Chem. Soc. 2021,143, .
[] H. Xiong, Z. Huang, Q. Lin, B. Yang, F. Yan, B. Liu, H. Chen, J. Kong,
Anal. Chem. 2022,94, .
[] M. Kitta, K. Murai, K. Yoshii, H. Sano, J. Am. Chem. Soc. 2021,143,
.
[] M. Mahmoudi, H. Hofmann, B. Rothen-Rutishauser, A. Petri-Fink,
Chem. Rev. 2012,112, .
[] A. Hassibi, A. Manickam, R. Singh, S. Bolouki, R. Sinha, K. B. Jirage,
M. W. McDermott, B. Hassibi, H. Vikalo, G. Mazarei, L. Pei, L.
Bousse, M. Miller, M. Heshami, M. P. Savage, M. T. Taylor, N.
Gamini, N. Wood, P. Mantina, P. Grogan, P. Kuimelis, P. Savalia,
S. Conradson, Y. Li, R. B. Meyer, E. Ku, J. Ebert, B. A. Pinsky, G.
Dolganov, et al., Nat. Biotechnol. 2018,36, .
[] J. Sabate Del Rio, O. Y. F. Henry, P. Jolly, D. E. Ingber, Nat. Nanotech-
nol. 2019,14, .
[] H. L. Chia, C. C. Mayorga-Martinez, M. Pumera, Adv. Funct. Mater.
2021,31, .
[] T. Rodrigues, V. Mishyn, Y. R. Leroux, L. Butruille, E. Woitrain, A.
Barras, P. Aspermair, H. Happy, C. Kleber, R. Boukherroub, D.
Montaigne, W. Knoll, S. Szunerits, Nano Today 2022,43, .
[] L. W. Hui, R. B. Bai, H. T. Liu, Adv. Funct. Mater. 2022,32, .
[] T. A. Chen, C. P. Chuu, C. C. Tseng, C. K. Wen, H. P. Wong, S. Pan,
R. Li, T. A. Chao, W. C. Chueh, Y. Zhang, Q. Fu, B. I. Yakobson, W. H.
Chang, L. J. Li, Nature 2020,579, .
[] N. R. Glavin, R. Rao, V. Varshney, E. Bianco, A. Apte, A. Roy, E. Ringe,
P. M. Ajayan, Adv. Mater. 2020,32, .
[] M. S. Long, P. Wang, H. H. Fang, W. D. Hu, Adv. Funct. Mater. 2019,
29, .
[] B.L.Wang,Y.F.Sun,H.Y.Ding,X.Zhao,L.Zhang,J.W.Bai,K.Liu,
Adv. Funct. Mater. 2020,30, .
[] R. Kempt, A. Kuc, T. Heine, Angew. Chem., Int. Ed. 2020,59, .
[] Y. Li, Z. Peng, N. J. Holl, M. R. Hassan, J. M. Pappas, C. Wei, O. H.
Izadi,Y.Wang,X.Dong,C.Wang,Y.W.Huang,D.Kim,C.Wu,ACS
Omega 2021,6, .
[] T. Li, H. Peng, Acc. Mater. Res. 2021,2, .
[] K. Yi, D. Liu, X. Chen, J. Yang, D. Wei, Y. Liu, D. Wei, Acc. Chem. Res.
2021,54, .
[] X. Wang, Y. Zhang, J. Wu, Z. Zhang, Q. Liao, Z. Kang, Y. Zhang,
Chem. Rev. 2022,122, .
[] V. Orts Mercadillo, K. C. Chan, M. Caironi, A. Athanassiou, I. A.
Kinloch, M. Bissett, P. Cataldi, Adv. Funct. Mater. 2022,32, .
[] X. Zhang, Q. Jing, S. Ao, G. F. Schneider, D. Kireev, Z. Zhang, W. Fu,
Small 2020,16, .
[] W. Fu, L. Jiang, E. P. van Geest, L. M. Lima, G. F. Schneider, Adv.
Mater. 2017,29, .
[] E. Danielson, V. A. Sontakke, A. J. Porkovich, Z. W. Wang, P. Kumar,
Z. Ziadi, Y. Yokobayashi, M. Sowwan, Sens. Actuators B-Chem. 2020,
320, .
[] S. Ramadan, R. Lobo, Y. Zhang, L. Xu, O. Shaforost, D. Kwong Hong
Tsang, J. Feng, T. Yin, M. Qiao, A. Rajeshirke, L. R. Jiao, P. K. Petrov, I.
E. Dunlop, M. M. Titirici, N. Klein, ACS Appl. Mater. Interfaces 2021,
13, .
[] X. Zhang, T. Liu, A. Boyle, A. Bahreman, L. Bao, Q. Jing, H. Xue,
R. Kieltyka, A. Kros, G. F. Schneider, W. Fu, Adv. Mater. 2022,34,
.
[] A. Ganguli, V. Faramarzi, A. Mostafa, M. T. Hwang, S. You, R. Bashir,
Adv. Funct. Mater. 2020,30, .
[] M. T. Hwang, M. Heiranian, Y. Kim, S. You, J. Leem, A. Taqieddin, V.
Faramarzi, Y. Jing, I. Park, A. M. van der Zande, S. Nam, N. R. Aluru,
R. Bashir, Nat. Commun. 2020,11, .
[] D. K. Ban, Y. Liu, Z. Wang, S. Ramachandran, N. Sarkar, Z. Shi, W.
Liu, A. G. Karkisaval, E. Martinez-Loran, F. Zhang, G. Glinsky, P. R.
Bandaru, C. Fan, R. Lal, ACS Nano 2020,14, .
[] Y.Luo,M.Wang,C.Wan,P.Cai,X.J.Loh,X.Chen,Adv. Mater. 2020,
32, .
[] L. Portilla, K. Loganathan, H. Faber, A. Eid, J. G. D. Hester, M. M.
Tentzeris, M. Fattori, E. Cantatore, C. Jiang, A. Nathan, G. Fiori, T.
Ibn-Mohammed, T. D. Anthopoulos, V. Pecunia, Nat. Electron. 2023,
6, .
Adv. Funct. Mater. 2023,  ©  Wiley-VCH GmbH
2301948 (21 of 26)
16163028, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/adfm.202301948 by Fudan University, Wiley Online Library on [11/05/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.afm-journal.de
[] F. Chen, Q. Qing, J. Xia, J. Li, N. Tao, J. Am. Chem. Soc. 2009,131,
.
[] H. Liu, A. Yang, J. Song, N. Wang, P. Lam, Y. Li, H. K. Law, F. Yan,
Sci. Adv. 2021,7, eabg.
[] W. Fu, L. Feng, G. Panaitov, D. Kireev, D. Mayer, A. Oenhausser,
H. J. Krause, Sci. Adv. 2017,3, .
[] D. Akinwande, C. Huyghebaert, C. H. Wang, M. I. Serna, S.
Goossens,L.J.Li,H.P.Wong,F.H.L.Koppens,Nature 2019,573,
.
[] Z. Wang, K. Yi, Q. Lin, L. Yang, X. Chen, H. Chen, Y. Liu, D. Wei, Nat.
Commun. 2019,10, .
[] D. K. Ban, T. Bodily, A. G. Karkisaval, Y. Dong, S. Natani, A.
Ramanathan, A. Ramil, S. Srivastava, P. Bandaru, G. Glinsky, R. Lal,
Proc. Natl. Acad. Sci. U.S.A. 2022,119, .
[] Y. Ohno, K. Maehashi, K. Matsumoto, J. Am. Chem. Soc. 2010,132,
.
[] J.Zhang,S.Shen,R.Lin,J.Huang,C.Pu,P.Chen,Q.Duan,X.You,
C. Xu, B. Yan, X. Gao, Z. Shen, L. Cai, X. Qiu, H. Hou, Adv. Mater.
2023,35, .
[] F. Chen, Q. Tang, T. Ma, B. H. Zhu, L. Y. Wang, C. He, X. L. Luo, S. J.
Cao, L. Ma, C. Cheng, Infomat 2022,4, .
[] L. Zhang, J. Dong, F. Ding, Chem. Rev. 2021,121, .
[] Q. Fan, J. Li, Y. Zhu, Z. Yang, T. Shen, Y. Guo, L. Wang, T. Mei, J.
Wan g, X . Wang, ACS Appl. Mater. Interfaces 2020,12, .
[] H. Li, Y. Shi, G. Han, J. Liu, J. Zhang, C. Li, J. Liu, Y. Yi, T. Li, X. Gao, C.
Di, J. Huang, Y. Che, D. Wang, W. Hu, Y. Liu, L. Jiang, Angew. Chem.,
Int. Ed. 2020,59, .
[] L. Yao, S. Gao, S. Liu, Y. Bi, R. Wang, H. Qu, Y. Wu, Y. Mao, L. Zheng,
ACS Appl. Mater. Interfaces 2020,12, .
[] S.He,A.M.Evans,E.Meirzadeh,S.Y.Han,J.C.Russell,R.A.
Wiscons, A. K. Bartholomew, D. A. Reed, A. Zangiabadi, M. L.
Steigerwald, C. Nuckolls, X. Roy, J. Am. Chem. Soc. 2022,144, .
[] X. Zhang, Y. Feng, S. Duan, L. Su, J. Zhang, F. He, ACS Sens. 2019,
4, .
[] S. Sheibani, L. Capua, S. Kamaei, S. S. A. Akbari, J. Zhang, H. Guerin,
A. M. Ionescu, Commun. Mater. 2021,2, .
[] Z. Meng, R. M. Stolz, L. Mendecki, K. A. Mirica, Chem. Rev. 2019,
119, .
[] S. S. Kwon, D. Kim, M. Yun, J. G. Son, S. H. Lee, Biosens. Bioelectron.
2021,192, .
[] X. Wang, D. Kong, M. Guo, L. Wang, C. Gu, C. Dai, Y. Wang, Q. Jiang,
Z. Ai, C. Zhang, D. Qu, Y. Xie, Z. Zhu, Y. Liu, D. Wei, Nano Lett. 2021,
21, .
[] H. Li, W. Shi, J. Song, H. Jang, J. Dailey, J. Yu, H. Katz, Chem. Rev.
2019,119,.
[] H. J. Jang, X. Sui, W. Zhuang, X. Huang, M. Chen, X. Cai, Y. Wang, B.
Ryu, H. Pu, N. Ankenbruck, K. Beavis, J. Huang, J. Chen, ACS Appl.
Mater. Interfaces 2022,14, .
[] Z. Gao, H. Xia, J. Zauberman, M. Tomaiuolo, J. Ping, Q. Zhang, P.
Ducos,H.Ye,S.Wang,X.Yang,F.Lubna,Z.Luo,L.Ren,A.T.C.
Johnson, Nano Lett. 2018,18, .
[] N. Gao, T. Gao, X. Yang, X. Dai, W. Zhou, A. Zhang, C. M. Lieber,
Proc. Natl. Acad. Sci. U.S.A. 2016,113, .
[] Z.Wang,W.Dai,S.Yu,Z.Hao,R.Pei,C.G.DeMoraes,L.H.Suh,
X. Zhao, Q. Lin, Tala n ta 2022,250, .
[] J. Deng, F. Tian, C. Liu, Y. Liu, S. Zhao, T. Fu, J. Sun, W. Tan, J. Am.
Chem. Soc. 2021,143, .
[] D. Najjar, J. Rainbow, S. Sharma Timilsina, P. Jolly, H. de Puig, M.
Yafia, N. Durr, H. Sallum, G. Alter, J. Z. Li, X. G. Yu, D. R. Walt, J.
A. Paradiso, P. Estrela, J. J. Collins, D. E. Ingber, Nat. Biomed. Eng.
2022,6, .
[] J. T. Atkinson, L. Su, X. Zhang, G. N. Bennett, J. J. Silberg, C. M.
Ajo-Franklin, Nature 2022,611, .
[] L.Yang,Y.Q.Jin,X.J.Wang,B.Yu,R.Z.Chen,C.Zhang,Y.Zhao,Y.
G.Yu,Y.Q.Liu,D.C.Wei,Adv. Electron. Mater. 2020,6, .
[] S. Nedelcu, K. Thodkar, C. Hierold, Microsyst. Nanoeng. 2022,8, .
[] H. Chen, P. Chen, J. Huang, R. Selegard, M. Platt, A. Palaniappan,
D. Aili, A. I. Tok, B. Liedberg, Anal. Chem. 2016,88, .
[] G. L. Long, J. D. Winefordner, Anal. Chem. 1983,55, A.
[] R. Hajian, S. Balderston, T. Tran, T. deBoer, J. Etienne, M. Sandhu, N.
A. Wauford, J. Y. Chung, J. Nokes, M. Athaiya, J. Paredes, R. Peytavi,
B. Goldsmith, N. Murthy, I. M. Conboy, K. Aran, Nat. Biomed. Eng.
2019,3, .
[] W. Zheng, S. M. LaCourse, B. Song, D. K. Singh, M. Khanna, J. Olivo,
J. Stern, J. N. Escudero, C. Vergara, F. Zhang, S. Li, S. Wang, L.
M. Cranmer, Z. Huang, C. M. Bojanowski, D. Bao, I. Njuguna, Y.
Xiao, D. C. Wamalwa, D. T. Nguyen, L. Yang, E. Maleche-Obimbo,
N. Nguyen, L. Zhang, H. Phan, J. Fan, B. Ning, C. Li, C. J. Lyon, E. A.
Graviss, et al., Nat. Biomed. Eng. 2022,6, .
[] K. Mensah, I. Cisse, A. Pierret, M. Rosticher, J. Palomo, P. Morfin, B.
Placais, U. Bockelmann, Adv. Healthcare Mater. 2020,9, .
[] V. Mishyn, T. Rodrigues, Y. R. Leroux, L. Butruille, E. Woitrain, D.
Montaigne, P. Aspermair, H. Happy, W. Knoll, R. Boukherroub, S.
Szunerits, Anal. Bioanal. Chem. 2022,414, .
[] M. Ma, Y. Zhou, J. Li, Z. Ge, H. He, T. Tao, Z. Cai, X. Wang, G. Chang,
Y. H e , Analyst 2020,145, .
[] H. Gao, F. Yang, K. Sattari, X. Du, T. Fu, S. Fu, X. Liu, J. Lin, Y. Sun,
J. Yao, Sci. Adv. 2022,8, eabn.
[] K. M. Cheung, J. M. Abendroth, N. Nakatsuka, B. Zhu, Y. Yang, A.
M. Andrews, P. S. Weiss, Nano Lett. 2020,20, .
[] K. Shoorideh, C. O. Chui, Proc. Natl. Acad. Sci. U.S.A. 2014,111,
.
[] D. Ji, M. Guo, Y. Wu, W. Liu, S. Luo, X. Wang, H. Kang, Y. Chen, C.
Dai, D. Kong, H. Ma, Y. Liu, D. Wei, J. Am. Chem. Soc. 2022,144,
.
[] S. Balderston, J. J. Taulbee, E. Celaya, K. Fung, A. Jiao, K. Smith,
R. Hajian, G. Gasiunas, S. Kutanovas, D. Kim, J. Parkinson, K.
Dickerson, J. J. Ripoll, R. Peytavi, H. W. Lu, F. Barron, B. R.
Goldsmith, P. G. Collins, I. M. Conboy, V. Siksnys, K. Aran, Nat.
Biomed. Eng. 2021,5, .
[] W. E. Huang, B. Lim, C. C. Hsu, D. Xiong, W. Wu, Y. Yu, H. Jia, Y.
Wang, Y. Zeng, M. Ji, H. Chang, X. Zhang, H. Wang, Z. Cui, Microb.
Biotechnol. 2020,13, .
[] Y. Sun, C. Yang, X. Jiang, P. Zhang, S. Chen, F. Su, H. Wang, W. Liu, X.
He, L. Chen, B. Man, Z. Li, Biosens. Bioelectron. 2023,222, .
[] H. Li, J. Yang, G. Wu, Z. Weng, Y. Song, Y. Zhang, J. A. Vanegas, L.
Avery, Z. Gao, H. Sun, Y. Chen, K. D. Dieckhaus, X. Gao, Y. Zhang,
Angew. Chem., Int. Ed. 2022,61, .
[] Y. Wu, D. Ji, C. Dai, D. Kong, Y. Chen, L. Wang, M. Guo, Y. Liu, D.
Wei, Nano Lett. 2022,22, .
[] H. Yu, H. Zhang, J. Li, Z. Zhao, M. Deng, Z. Ren, Z. Li, C. Xue, M.
G. Li, Z. Chen, ACS Sens. 2022,7, .
[] D. Kong, X. Wang, C. Gu, M. Guo, Y. Wang, Z. Ai, S. Zhang, Y. Chen,
W. Liu, Y. Wu, C. Dai, Q. Guo, D. Qu, Z. Zhu, Y. Xie, Y. Liu, D. Wei, J.
Am. Chem. Soc. 2021,143, .
[] R. Campos, J. Borme, J. R. Guerreiro, G. Machado, Jr., M. F.
Cerqueira, D. Y. Petrovykh, P. Alpuim, ACS Sens. 2019,4, .
[] S.Li,K.Huang,Q.Fan,S.Yang,T.Shen,T.Mei,J.Wang,X.Wang,
G. Chang, J. Li, Biosens. Bioelectron. 2019,136, .
[] S.C.Xu,S.Z.Jiang,C.Zhang,W.W.Yue,Y.Zou,G.Y.Wang,H.
L. Liu, X. M. Zhang, M. Z. Li, Z. S. Zhu, J. H. Wang, Appl. Surf. Sci.
2018,427, .
[] J. Sun, X. Xie, K. Xie, S. Xu, S. Jiang, J. Ren, Y. Zhao, H. Xu, J. Wang,
W. Yu e, Nanoscale Res. Lett. 2019,14, .
[] R. Vishnubhotla, A. Sriram, O. O. Dickens, S. V. Mandyam, J. L. Ping,
E. Adu-Beng, A. T. C. Johnson, IEEE Sens. J. 2020,20, .
Adv. Funct. Mater. 2023,  ©  Wiley-VCH GmbH
2301948 (22 of 26)
16163028, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/adfm.202301948 by Fudan University, Wiley Online Library on [11/05/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.afm-journal.de
[] M. Azize, D. J. Bishop, N. E. Fuhr, ACS Appl. Nano Mater. 2022,5,
.
[] Z. Ge, M. Ma, G. Chang, M. Chen, H. He, X. Zhang, S. Wang, Analyst
2020,145, .
[] K. Li, J. Tu, Y. Zhang, D. Jin,T. Li, J. Li, W. Ni, M. M. Xiao, Z. Y. Zhang,
G. J. Zhang, iScience 2022,25, .
[] C. H. Huang, W. T. Huang, T. T. Huang, S. H. Ciou, C. F. Kuo, A. H.
Hsieh, Y. S. Hsiao, Y. J. Lee, ACS Appl. Electron. Mater. 2021,3, .
[] M. Deng, Z. Ren, H. Zhang, Z. Li, C. Xue, J. Wang, D. Zhang, H.
Yang, X. Wang, J. Li, Adv. Sci. 2023,10, .
[] J. Gao, Y. Gao, Y. Han, J. Pang, C. Wang, Y. Wang, H. Liu, Y. Zhang,
L. Han, ACS Appl. Electron. Mater. 2020,2, .
[] M. Tian, M. Qiao, C. C. Shen, F. L. Meng, L. A. Frank, V. V.
Krasitskaya, T. J. Wang, X. M. Zhang, R. H. Song, Y. X. Li, J. J. Liu, S.
C. Xu, J. H. Wang, Appl. Surf. Sci. 2020,527, .
[] G. Ke, D. Su, Y. Li, Y. Zhao, H. Wang, W. Liu, M. Li, Z. Yang, F. Xiao,
Y.Yuan,F.Huang,F.Mo,P.Wang,X.Guo,Sci. China Mater. 2021,
64, .
[] J. Gao, C. Wang, C. Wang, Y. Chu, S. Wang, M. Y. Sun, H. Ji, Y. Gao, Y.
Wang, Y. Han, F. Song, H. Liu, Y. Zhang, L. Han, Anal. Chem. 2022,
94, .
[] J. Li, D. Wu, Y. Yu, T. Li, K. Li, M. M. Xiao, Y. Li, Z. Y. Zhang, G. J.
Zhang, Biosens. Bioelectron. 2021,183, .
[] I. M. Bhattacharyya, S. Cohen, A. Shalabny, M. Bashouti, B.
Akabayov, G. Shalev, Biosens. Bioelectron. 2019,132, .
[] M. S. Chae, Y. K. Yoo, J. Kim, T. G. Kim, K. S. Hwang, Sens. Actuators
B-Chem. 2018,272, .
[] O. Oshin, D. Kireev, H. Hlukhova, F. Idachaba, D. Akinwande, A.
Atayero, Sensors 2020,20, .
[] H. Yu, Z. Zhao, B. Xiao, M. Deng, Z. Wang, Z. Li, H. Zhang, L. Zhang,
J. Qian, J. Li, Anal. Chem. 2021,93, .
[] P. Fathi-Hafshejani, N. Azam, L. Wang, M. A. Kuroda, M. C.
Hamilton, S. Hasim, M. Mahjouri-Samani, ACS Nano 2021,15,
.
[] Z. Hao, Y. Luo, C. Huang, Z. Wang, G. Song, Y. Pan, X. Zhao, S. Liu,
Small 2021,17, .
[] C. Choi, Y. Lee, K. W. Cho, J. H. Koo, D. H. Kim, Acc. Chem. Res. 2019,
52, .
[] I. Palacio, M. Moreno, A. Nanez, A. Purwidyantri, T. Domingues,
P. D. Cabral, J. Borme, M. Marciello, J. I. Mendieta-Moreno, B.
Torres-Vazquez, J. I. Martinez, M. F. Lopez, M. Garcia-Hernandez, L.
Vazquez, P. Jelinek, P. Alpuim, C. Briones, J. A. Martin-Gago, Biosens.
Bioelectron. 2023,222, .
[] M. T. Hwang, I. Park, M. Heiranian, A. Taqieddin, S. You, V.
Faramarzi, A. A. Pak, A. M. van der Zande, N. R. Aluru, R. Bashir,
Adv.Mater. Technol. 2021,6, .
[] D. Park, J. H. Kim, H. J. Kim, D. Lee, D. S. Lee, D. S. Yoon, K. S.
Hwang, Biosens. Bioelectron. 2020,167, .
[] R. S. Selvarajan, R. A. Rahim, B. Y. Majlis, S. C. B. Gopinath, A. A.
Hamzah, Sensors 2020,20, .
[] M. Hinnemo, A. Makaraviciute, P. Ahlberg, J. Olsson, Z. Zhang, S.
L. Zhang, Z. B. Zhang, IEEE Sens. J. 2018,18, .
[] Y. Kanai, Y. Ohmuro-Matsuyama, M. Tanioku, S. Ushiba, T. Ono, K.
Inoue, T. Kitaguchi, M. Kimura, H. Ueda, K. Matsumoto, ACS Sens.
2020,5, .
[] Z. Wang, Z. Hao, X. Wang, C. Huang, Q. Lin, X. Zhao, Y. Pan, Adv.
Funct. Mater. 2020,31, .
[] L. Xu, S. Ramadan, O. E. Akingbade, Y. Zhang, S. Alodan, N.
Graham, K. A. Zimmerman, E. Torres, A. Heslegrave, P. K. Petrov,
H. Zetterberg, D. J. Sharp, N. Klein, B. Li, ACS Sens. 2022,7, .
[] J. A. Basu, C. RoyChaudhuri, IEEE Sens. J. 2018,18, .
[] K. H. Cho, D. H. Shin, J. Oh, J. H. An, J. S. Lee, J. Jang, ACS Appl.
Mater. Interfaces 2018,10, .
[] S. J. Gao, R. R. Wang, Y. L. Bi, H. Qu, Y. Chen, L. Zheng, Sens. Actu-
ators B-Chem. 2020,305, .
[] Z. Hao, C. Huang, C. Zhao, A. Kospan, Z. Wang, F. Li, H. Wang, X.
Zhao, Y. Pan, S. Liu, ACS Appl. Bio. Mater. 2022,5, .
[] P. Aspermair, V. Mishyn, J. Bintinger, H. Happy, K. Bagga, P.
Subramanian, W. Knoll, R. Boukherroub, S. Szunerits, Anal. Bioanal.
Chem. 2021,413, .
[] Z. Wang, Z. Hao, S. Yu, C. Huang, Y. Pan, X. Zhao, Nanomaterials
2020,10, .
[] X. Wang, Y. Zhu, T. R. Olsen, N. Sun, W. Zhang, R. Pei, Q. Lin, Elec-
trochim. Acta 2018,290, .
[] Z. Hao, Y. Pan, C. Huang, Z. Wang, X. Zhao, Biomed. Microdevices
2019,21, .
[] Z. Hao, Y. Pan, C. Huang, Z. Wang, Q. Lin, X. Zhao, S. Liu, ACS Sens.
2020,5, .
[] M. B. Lerner, D. Pan, Y. N. Gao, L. E. Locascio, K. Y. Lee, J. Nokes, S.
Afsahi, J. D. Lerner, A. Walker, P. G. Collins, K. Oegema, F. Barron,
B. R. Goldsmith, Sens. Actuators B-Chem. 2017,239, .
[] T. Murugathas, C. Hamiaux, D. Colbert, A. V. Kralicek, N. O. V. Plank,
C. Carraher, ACS Appl. Electron. Mater. 2020,2, .
[] A. E. Islam, R. Martineau, C. M. Crasto, H. Kim, R. S. Rao, B.
Maruyama, S. S. Kim, L. F. Drummy, ACS Appl. Nano Mater. 2020,
3, .
[] Y. Zhou, B. Liu, Y. Lei, L. Tang, T. Li, S. Yu, G. J. Zhang, Y. T. Li, Small
2022,18, e.
[] W. M. Munief, X. Lu, T. Teucke, J. Wilhelm, A. Britz, F. Hempel, R.
Lanche, M. Schwartz, J. K. Y. Law, S. Grandthyll, F. Muller, J. U.
Neurohr, K. Jacobs, M. Schmitt, V. Pachauri, R. Hempelmann, S.
Ingebrandt, Biosens. Bioelectron. 2019,126, .
[] N. Mandal, V. Pakira, N. Samanta, N. Das, S. Chakraborty, B.
Pramanick, C. RoyChaudhuri, Tala n ta 2021,222, .
[] H. Kang, X. Wang, M. Guo, C. Dai, R. Chen, L. Yang, Y. Wu, T. Ying,
Z. Zhu, D. Wei, Y. Liu, D. Wei, Nano Lett. 2021,21, .
[] L. Xu, S. Ramadan, B. G. Rosa, Y. Zhang, T. Yin, E. Torres, O.
Shaforost, A. Panagiotopoulos, B. Li, G. Kerherve, D. K. Kim, C.
Mattevi, L. R. Jiao, P. K. Petrov, N. Klein, Sens. Diagn. 2022,1,
.
[] D. Shahdeo, N. Chauhan, A. Majumdar, A. Ghosh, S. Gandhi, ACS
Appl. Bio. Mater. 2022,5, .
[] J. Gao, C. Wang, Y. Chu, Y. Han, Y. Gao, Y. Wang, C. Wang, H. Liu, L.
Han, Y. Zhang, Tala n t a 2022,240, .
[] I. Novodchuk, M. Kayaharman, I. Prassas, A. Soosaipillai, R. Karimi,
I. A. Goldthorpe, E. Abdel-Rahman, J. Sanderson, E. P. Diamandis,
M. Bajcsy, M. Yavuz, Biosens. Bioelectron. 2022,210, .
[] G. Seo, G. Lee, M. J. Kim, S. H. Baek, M. Choi, K. B. Ku, C. S. Lee, S.
Jun, D. Park, H. G. Kim, S. J. Kim, J. O. Lee, B. T. Kim, E. C. Park, S.
I. Kim, ACS Nano 2020,14, .
[] B. V. Krsihna, S. Ahmadsaidulu, S. S. T. Teja, D. Jayanthi, A.
Navaneetha, P. R. Reddy, M. D. Prakash, Silicon 2022,14, .
[] S. Kim, H. Ryu, S. Tai, M. Pedowitz, J. R. Rzasa, D. J. Pennachio,
J. R. Hajzus, D. K. Milton, R. Myers-Ward, K. M. Daniels, Biosens.
Bioelectron. 2022,197, .
[] A. Wasfi, F. Awwad, N. Qamhieh, B. Al Murshidi, A. R. Palakkott, J.
G. Gelovani, Sci. Rep. 2022,12, .
[] N. I. Khan, M. Mousazadehkasin, S. Ghosh, J. G. Tsavalas, E. Song,
Analyst 2020,145, .
[] Z. Hao, Z. Wang, Y. Li, Y. Zhu, X. Wang, C. G. De Moraes, Y. Pan, X.
Zhao, Q. Lin, Nanoscale 2018,10, .
[] S. Afsahi, M. B. Lerner, J. M. Goldstein, J. Lee, X. Tang, D. A.
Bagarozzi Jr., D. Pan, L. Locascio, A. Walker, F. Barron, B. R.
Goldsmith, Biosens. Bioelectron. 2018,100, .
[] W. Ji, D. Q. Wu, W. Tang, X. Xi, Y. Z. Su, X. J. Guo, R. L. Liu, Sens.
Actuators B-Chem. 2020,304, .
Adv. Funct. Mater. 2023,  ©  Wiley-VCH GmbH
2301948 (23 of 26)
16163028, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/adfm.202301948 by Fudan University, Wiley Online Library on [11/05/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.afm-journal.de
[] S. C. Xu, C. Zhang, S. Z. Jiang, G. D. Hu, X. Y. Li, Y. Zou, H. P. Liu, J.
Li,Z.H.Li,X.X.Wang,M.Z.Li,J.H.Wang,Sens. Actuators B-Chem.
2019,284, .
[] E. Piccinini, C. Bliem, C. Reiner-Rozman, F. Battaglini, O. Azzaroni,
W. Knoll, Biosens. Bioelectron. 2017,92, .
[] A. Beraud, M. Sauvage, C. M. Bazan, M. Tie, A. Bencherif, D. Bouilly,
Analyst 2021,146, .
[] A. Silvestri, J. Zayas-Arrabal, M. Vera-Hidalgo, D. Di Silvio, C. Wetzl,
M. Martinez-Moro, A. Zurutuza, E. Torres, A. Centeno, A. Maestre,
J. M. Gomez, M. Arrastua, M. Elicegui, N. Ontoso, M. Prato, I.
Coluzza, A. Criado, Nanoscale 2023,15, .
[] G. E. Fenoy, W. A. Marmisolle, O. Azzaroni, W. Knoll, Biosens. Bio-
electron. 2020,148, .
[] S. Wang, M. Z. Hossain, K. Shinozuka, N. Shimizu, S. Kitada, T.
Suzuki, R. Ichige, A. Kuwana, H. Kobayashi, Biosens. Bioelectron.
2020,165, .
[] R. Zhang, Y. Jia, ACS Sens. 2021,6, .
[] E. Danielson, M. Dindo, A. J. Porkovich, P. Kumar, Z. Wang, P. Jain,
T. Mete, Z. Ziadi, R. Kikkeri, P. Laurino, M. Sowwan, Biosens. Bioelec-
tron. 2020,165, .
[] M. Ku, J. Kim, J. E. Won, W. Kang, Y. G. Park, J. Park, J. H. Lee, J.
Cheon, H. H. Lee, J. U. Park, Sci. Adv. 2020,6, eabb.
[] R. Kalluri, V. S. LeBleu, Science 2020,367, .
[] M. A. Unal, F. Bayrakdar, H. Nazir, O. Besbinar, C. Gurcan, N.
Lozano, L. M. Arellano, S. Yalcin, O. Panatli, D. Celik, D. Alkaya,
A. Agan, L. Fusco, S. Suzuk Yildiz, L. G. Delogu, K. C. Akcali, K.
Kostarelos, A. Yilmazer, Small 2021,17, .
[] Y. Lei, R. Zeng, Y.-T. Li, M.-M. Xiao, Z.-Y. Zhang, G.-J. Zhang, Carbon
2023,201, .
[] V. Dupuit, O. Terral, G. Bres, A. Claudel, B. Fernandez, A. Briançon-
Marjollet, C. Delacour, Adv. Funct. Mater. 2022,32, .
[] W. Kong, H. Kum, S. H. Bae, J. Shim, H. Kim, L. Kong, Y. Meng, K.
Wang,C.Kim,J.Kim,Nat. Nanotechnol. 2019,14, .
[] H. R. Lim, H. S. Kim, R. Qazi, Y. T. Kwon, J. W. Jeong, W. H. Yeo, Adv.
Mater. 2020,32, .
[] W. Shi, Y. Guo, Y. Liu, Adv. Mater. 2020,32, .
[] T. Q. Trung, S. Ramasundaram, B. U. Hwang, N. E. Lee, Adv. Mater.
2016,28, .
[] N. Gao, R. Zhou, B. Tu, T. Tao, Y. Song, Z. Cai, H. He, G. Chang, Y.
Wu, Y. He, Anal. Chim. Acta 2023,1239, .
[] C. Huang, Z. Hao, Z. Wang, H. Wang, X. Zhao, Y. Pan, Adv. Mater.
Technol. 2022,7, .
[] J. B. Park, M. S. Song, R. Ghosh, R. K. Saroj, Y. Hwang, Y. Tchoe, H.
Oh, H. Baek, Y. Lim, B. Kim, S. W. Kim, G. C. Yi, NPG Asia Mater.
2021,13, .
[] A. Paul, N. Yogeswaran,R. Dahiya, Adv. Intell. Syst. 2022,4, .
[] S. Subbulakshmi Radhakrishnan, A. Sebastian, A. Oberoi, S. Das, S.
Das, Nat. Commun. 2021,12, .
[] C. H. Dai, Y. C. Li, Z. W. Liao, J. Kong, T. C. Wang, Ceram. Int. 2019,
45, .
[] O. S. Kwon, H. S. Song, S. J. Park, S. H. Lee, J. H. An, J. W. Park,
H.Yang,H.Yoon,J.Bae,T.H.Park,J.Jang,Nano Lett. 2015,15,
.
[] C. W. Lee, J. M. Suh, S. Choi, S. E. Jun, T. H. Lee, J. W. Yang, S. A. Lee,
B. R. Lee, D. Yoo, S. Y. Kim, D. S. Kim, H. W. Jang, NPJ 2D Mater.
Appl. 2021,5, .
[] T. Hayasaka, A. Lin, V. C. Copa, L. P. Lopez Jr., R. A. Loberternos, L.
I. M. Ballesteros, Y. Kubota, Y. Liu, A. A. Salvador, L. Lin, Microsyst.
Nanoeng. 2020,6, .
[] K. Persaud, G. Dodd, Nature 1982,299, .
[] J. Yeom, A. Choe, S. Lim, Y. Lee, S. Na, H. Ko, Sci. Adv. 2020,6,
eaba.
[] S. R. Ahn, J. H. An, H. S. Song, J. W. Park, S. H. Lee, J. H. Kim, J.
Jang, T. H. Park, ACS Nano 2016,10, .
[] S. R. Ahn, J. H. An, I. H. Jang, W. Na, H. Yang, K. H. Cho, S. H. Lee,
H. S. Song, J. Jang, T. H. Park, Biosens. Bioelectron. 2018,117, .
[] Y. H. Jung, B. Park, J. U. Kim, T. I. Kim, Adv. Mater. 2019,31, .
[] C. Choi, M. K. Choi, S. Liu, M. S. Kim, O. K. Park, C. Im, J. Kim, X.
Qin, G. J. Lee, K. W. Cho, M. Kim, E. Joh, J. Lee, D. Son, S. H. Kwon,
N. L. Jeon, Y. M. Song, N. Lu, D. H. Kim, Nat. Commun. 2017,8,
.
[] J. Yang, G. Li, W. Wang, J. Shi, M. Li, N. Xi, M. Zhang, L. Liu, Biosens.
Bioelectron. 2021,178, .
[] E. Masvidal-Codina, X. Illa, M. Dasilva, A. B. Calia, T. Dragojevic,
E. E. Vidal-Rosas, E. Prats-Alfonso, J. Martinez-Aguilar, J. M. De la
Cruz, R. Garcia-Cortadella, P. Godignon, G. Rius, A. Camassa, E. Del
Corro,J.Bousquet,C.Hebert,T.Durduran,R.Villa,M.V.Sanchez-
Vives, J. A. Garrido, A. Guimera-Brunet, Nat. Mater. 2019,18, .
[] R. Garcia-Cortadella, G. Schwesig, C. Jeschke, X. Illa, A. L. Gray,
S. Savage, E. Stamatidou, I. Schiessl, E. Masvidal-Codina, K.
Kostarelos, A. Guimera-Brunet, A. Sirota, J. A. Garrido, Nat. Com-
mun. 2021,12, .
[] A. Bonaccini Calia, E. Masvidal-Codina, T. M. Smith, N. Schafer, D.
Rathore, E. Rodriguez-Lucas, X. Illa, J. M. De la Cruz, E. Del Corro, E.
Prats-Alfonso, D. Viana, J. Bousquet, C. Hebert, J. Martinez-Aguilar,
J. R. Sperling, M. Drummond, A. Halder, A. Dodd, K. Barr, S. Savage,
J. Fornell, J. Sort, C. Guger, R. Villa, K. Kostarelos, R. C. Wykes, A.
Guimera-Brunet, J. A. Garrido, Nat. Nanotechnol. 2022,17,.
[] J. A. Hartings, C. Li, J. M. Hinzman, C. W. Shuttleworth, G. L. Ernst,
J. P. Dreier, J. A. Wilson, N. Andaluz, B. Foreman, A. P. Carlson, J.
Cereb. Blood Flow Metab. 2017,37, .
[] R. Garcia-Cortadella, N. Schafer, J. Cisneros-Fernandez, L. Re, X. Illa,
G. Schwesig, A. Moya, S. Santiago, G. Guirado, R. Villa, A. Sirota, F.
Serra-Graells, J. A. Garrido, A. Guimera-Brunet, Nano Lett. 2020,20,
.
[] R. S. Hazra, M. R. Hasan Khan, N. Kale, T. Tanha, J. Khandare, S.
Ganai, M. Quadir, ACS Biomater. Sci. Eng. 2022, https://doi.org/.
/acsbiomaterials.c.
[] E. Piccinini, G. E. Fenoy, A. L. Cantillo, J. A. Allegretto, J. Scotto, J.
M. Piccinini, W. A. Marmisolle, O. Azzaroni, Adv. Mater. Interfaces
2022,9, .
[] A. Maity, X. Sui, B. Jin, H. Pu, K. J. Bottum, X. Huang, J. Chang, G.
Zhou, G. Lu, J. Chen, Anal. Chem. 2018,90, .
[] S. K. Krishnan, N. Nataraj, M. Meyyappan, U. Pal, Anal. Chem. 2023,
95, .
[] M. Pareek, C. Greenaway, Lancet Public Health 2022,7, e.
[] R. Hajian, J. DeCastro, J. Parkinson, A. Kane, A. F. R. Camelo, P.
P. Chou, J. Yang, N. Wong, E. D. O. Hernandez, B. Goldsmith, I.
Conboy, K. Aran, Adv. Biol. 2021,5, e.
[] Z. Hao, Y. Pan, W. Shao, Q. Lin, X. Zhao, Biosens. Bioelectron. 2019,
134, .
[] Y. Chen, D. Kong, L. Qiu, Y. Wu, C. Dai, S. Luo, Z. Huang, Q. Lin, H.
Chen,S.Xie,L.Geng,J.Zhao,W.Tan,Y.Liu,D.Wei,Anal. Chem.
2023,95, .
[] K. H. Kim, S. J. Park, C. S. Park, S. E. Seo, J. Lee, J. Kim, S. H. Lee,
S. Lee, J. S. Kim, C. M. Ryu, D. Yong, H. Yoon, H. S. Song, S. H. Lee,
O. S. Kwon, Biosens. Bioelectron. 2020,167, .
[] S. Samota, R. Rani, S. Chakraverty, A. Kaushik, Mater. Sci. Semicond.
Process. 2022,141, .
[] J. Riu, B. Giussani, Trends Analyt. Chem. 2020,126, .
[] N. Kumar, W. Wang, J. C. Ortiz-Marquez, M. Catalano, M. Gray, N.
Biglari, K. Hikari, X. Ling, J. Gao, T. van Opijnen, K. S. Burch, Biosens.
Bioelectron. 2020,156, .
[] N. Kumar, M. Rana, M. Geiwitz, N. I. Khan, M. Catalano, J. C. Ortiz-
Marquez, H. Kitadai, A. Weber, B. Dweik, X. Ling, T. van Opijnen, A.
A. Argun, K. S. Burch, ACS Nano 2022,16, .
[] X. Tan, M. Yang, L. Zhu, G. Gunathilaka, Z. Zhou, P.-Y. Chen, Y.
Zhang, M. M.-C. Cheng, IEEE Sens. J. 2022,22, .
Adv. Funct. Mater. 2023,  ©  Wiley-VCH GmbH
2301948 (24 of 26)
16163028, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/adfm.202301948 by Fudan University, Wiley Online Library on [11/05/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.afm-journal.de
[] Z. Lin, G. Wu, L. Zhao, K. W. C. Lai, Nanomaterials 2021,11, .
[] Y. Kutovyi, H. Hlukhova, N. Boichuk, M. Menger, A. Oenhausser,
S. Vitusevich, Biosens. Bioelectron. 2020,154, .
[] S. R. Millar, J. Q. Huang, K. J. Schreiber, Y. C. Tsai, J. Won, J. Zhang,
A. M. Moses, J. Y. Youn, Chem. Rev. 2023, https://doi.org/./
acs.chemrev.c.
[] G. Bieri, A. B. Schroer, S. A. Villeda, Nat. Neurosci. 2023,26, .
[] K. Yang, J. C. Chaput, J. Am. Chem. Soc. 2021,143, .
[] B. Udugama, P. Kadhiresan, H. N. Kozlowski, A. Malekjahani, M.
Osborne, V. Y. C. Li, H. Chen, S. Mubareka, J. B. Gubbay, W. C. W.
Chan, ACS Nano 2020,14, .
[] M. Garcia-Finana, I. E. Buchan, Science 2021,372, .
[] D. B. Larremore, B. Wilder, E. Lester, S. Shehata, J. M. Burke, J. A.
Hay, M. Tambe, M. J. Mina, R. Parker, Sci. Adv. 2021,7, eabd.
[] M. A. Tortorici, M. Beltramello, F. A. Lempp, D. Pinto, H. V. Dang, L.
E. Rosen, M. McCallum, J. Bowen, A. Minola, S. Jaconi, F. Zatta, A.
De Marco, B. Guarino, S. Bianchi, E. J. Lauron, H. Tucker, J. Zhou,
A. Peter, C. Havenar-Daughton, J. A. Wojcechowskyj, J. B. Case, R.
E. Chen, H. Kaiser, M. Montiel-Ruiz, M. Meury, N. Czudnochowski,
R. Spreafico, J. Dillen, C. Ng, N. Sprugasci, et al., Science 2020,370,
.
[] A. Panahi, D. Sadighbayan, S. Forouhi, E. Ghafar-Zadeh, Biosensors
2021,11, .
[] K. R. McCarthy, L. J. Rennick, S. Nambulli, L. R. Robinson-
McCarthy, W. G. Bain, G. Haidar, W. P. Duprex, Science 2021,371,
.
[] I. Park, J. Lim, S. You, M. T. Hwang, J. Kwon, K. Koprowski, S. Kim,
J. Heredia, S. A. Stewart de Ramirez, E. Valera, R. Bashir, ACS Sens.
2021,6, .
[] A. Abusukhon, IEEE Internet Things J. 2021,8, .
[] X. Liao, W. Song, X. Zhang, C. Yan, T. Li, H. Ren, C. Liu, Y. Wang, Y.
Zheng, Nat. Commun. 2020,11, .
[] I. Akyildiz, M. Pierobon, S. Balasubramaniam, Y. Koucheryavy, IEEE
Commun. Mag. 2015,53, .
[] P. Li, G. H. Lee, S. Y. Kim, S. Y. Kwon, H. R. Kim, S. Park, ACS Nano
2021,15, .
Changhao Dai received his bachelor’s degree from Nanjing University of Science and Technology in
. He is currently pursuing a Ph. D. at the Department of Macromolecular Science, Fudan Univer-
sity at Shanghai, China, under the guidance of Prof. Dacheng Wei. His research interests center on the
preparation of D materials and their sensing applications.
Derong Kong received his Ph. D. degree from Fudan University in . She is currently a postdoctoral
scholar in Dr.Wei’s research group at Fudan University. Her major research interests are to combine
electrochemical bio-platforms and CRISPR-based technology for point-of-care detection of unpro-
cessed biological samples.
Chang Chen received her master’s degree from Fuzhou University in . She is currently a Ph. D.
candidate at the Department of Macromolecular Science, Fudan University at Shanghai, China, un-
der the guidance of Prof. Dacheng Wei. Her research focuses on the application of graphene-based
electronics in bioelectronic sensing.
Adv. Funct. Mater. 2023,  ©  Wiley-VCH GmbH
2301948 (25 of 26)
16163028, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/adfm.202301948 by Fudan University, Wiley Online Library on [11/05/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.afm-journal.de
YunqiLiu is a professor of Fudan University. He was selected as an academician of the Chinese
Academy of Sciences in . He received his Ph.D. degree from Tokyo Institute of Technology(Japan)
in . He is an adjunct professor of Tsinghua University (China) and a guest professor of Kyoto Uni-
versity (Japan). His research interests include molecular materials and devices.
Dacheng Wei is a professor of Fudan University. He received his bachelor’s degree from Zhejiang
University in  and his Ph. D. from the Institute of Chemistry,Chinese Academy of Sciences, in
. His research interests include controllable growth of D materials and their applications in FET
devices and sensors.
Adv. Funct. Mater. 2023,  ©  Wiley-VCH GmbH
2301948 (26 of 26)
16163028, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/adfm.202301948 by Fudan University, Wiley Online Library on [11/05/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
... The P-O peaks at 132.6 and 133.7 eV correspond to the phosphate groups of DNA aptamers, and the N-C peak at 401.6 eV is attributed to the thymine homo-oligonucleotides of antibodies. [3,30,37,47] Electrochemical impedance spectroscopy (Figure 2c) further validated the successful functionalization of the biological probes in the sensing area. To gain insight into the recognition activity of these probes, we investigated the molecular structures of protein-protein complexes in biofluids ( Figure 2d). ...
... s −1 for different analytes ( Figure S14, Supporting Information), which is two orders of magnitude greater than that of reported probes. [30,37,46] These performance metrics support the conclusion that the DMDP can successfully obtain reliable signals in complex POCT settings. ...
... attracted extensive attention because of their advantages in terms of sensitive detection, rapid response, and easy operation. [30,35,48] Thus, the DMDP working principle is divided into two parts: (i) the exergonic electron transfer reaction of the ECL sensing unit and (ii) the chemoelectric gating effect of the FET sensing unit. [1,14,45,60] Benefiting from the unique combination of ECL and FET sensors, the dual-mode platform generates crossvalidated readouts derived from independent signal transduction processes, physically reducing potential inaccuracy from random disturbances. ...
Article
Full-text available
On‐site diagnostic tests that accurately identify disease biomarkers lay the foundation for self‐healthcare applications. However, these tests routinely rely on single‐mode signals and suffer from insufficient accuracy, especially for multiplexed point‐of‐care tests (POCTs) within a few minutes. Here, this work develops a dual‐mode multiclassification diagnostic platform that integrates an electrochemiluminescence sensor and a field‐effect transistor sensor in a microfluidic chip. The microfluidic channel guides the testing samples to flow across electro‐optical sensor units, which produce dual‐mode readouts by detecting infectious biomarkers of tuberculosis (TB), human rhinovirus (HRV), and group B streptococcus (GBS). Then, machine‐learning classifiers generate three‐dimensional (3D) hyperplanes to diagnose different diseases. Dual‐mode readouts derived from distinct mechanisms enhance the anti‐interference ability physically, and machine‐learning‐aided diagnosis in high‐dimensional space reduces the occasional inaccuracy mathematically. Clinical validation studies with 501 unprocessed samples indicate that the platform has an accuracy approaching 99%, higher than the 77%–93% accuracy of rapid point‐of‐care testing technologies at 100% statistical power (>150 clinical tests). Moreover, the diagnosis time is 5 min without a trade‐off of accuracy. This work solves the occasional inaccuracy issue of rapid on‐site diagnosis, endowing POCT systems with the same accuracy as laboratory tests and holding unique prospects for complicated scenes of personalized healthcare.
... The presence of biological molecules with specific dielectric constants inside this cavity changes the dielectric gate capacitance and causes a shift in the threshold voltage. A change in current due to threshold-voltage shift indicates the presence of the moiety [38,39]. Graphene, a kind of 2D carbon atomic flake material with a six-circular honeycomb lattice structure, has emerged as a widely studied 2D biosensor material. ...
... The presence of biological molecules with specific dielectric constants inside this cavity changes the dielectric gate capacitance and causes a shift in the threshold voltage. A change in current due to threshold-voltage shift indicates the presence of the moiety [38,39]. GFET biosensors have become a powerful diagnostic method for the real-time and on-site detection of COVID-19 because of their advantages of high sensitivity, high selectivity, fast analysis speed, label free, low cost, miniaturization, and integration. ...
Article
Full-text available
Coronavirus disease 2019 (COVID-19) is a disease caused by the infectious agent of severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2). The primary method of diagnosing SARS-CoV-2 is nucleic acid detection, but this method requires specialized equipment and is time consuming. Therefore, a sensitive, simple, rapid, and low-cost diagnostic test is needed. Graphene field-effect transistor (GFET) biosensors have become the most promising diagnostic technology for detecting SARS-CoV-2 due to their advantages of high sensitivity, fast-detection speed, label-free operation, and low detection limit. This review mainly focus on three types of GFET biosensors to detect SARS-CoV-2. GFET biosensors can quickly identify SARS-CoV-2 within ultra-low detection limits. Finally, we will outline the pros and cons of the diagnostic approaches as well as future directions.
Article
Full-text available
Recently, field‐effect transistors (FETs) have emerged as a novel type of multiparameter, high‐performance, highly integrated platform for biochemical detection, leveraging their classical three‐terminal structure, working principles, and fabrication methods. Notably, graphene materials, known for their exceptional electrical and optical properties as well as biocompatibility, serve as a fundamental component of these devices, further enhancing their advantages in biological detection. This review places special emphasis on recent advancements in graphene field‐effect transistor (GFET)‐based biosensors and focuses on four main areas: i) the basic concepts of FETs and the specific electrical properties of GFETs; ii) various state‐of‐the‐art approaches to enhance the performance of GFET‐based biosensors in terms of operating principles and the “3S”—stability, sensitivity, and specificity; iii) multiplexed detection strategies for GFET‐based biosensors; and iv) the current challenges and future perspectives in the field of GFET‐based biosensors. It is hoped that this article can profoundly elucidate the development of GFET biosensors and inspire a broader audience.
Article
Full-text available
Bioartificial livers have showcased significant value in the treatment of acute liver failure (ALF). Current efforts are directed toward overcoming challenges in the development of microcarriers, with a specific emphasis on integrating higher‐density liver cells to enhance detoxification capabilities. Here, inspired by the radial filtration model in hepatic lobules, ion‐specific silk fibroin microcarriers are proposed with biomimetic niches for cultivating functional liver cells at high density. These biomimetic microcarriers are generated by capillary microfluidic device with controllable adjustments of ion type or concentration within the aqueous phase. When cultivating human induced pluripotent stem cell ‐differentiated mature liver cells on these recrystallized microcarriers, notably enhanced cell proliferation activity, as well as increased metabolic and secretory functionality is observed. Based on these features, the microcarrier‐integrated bioreactor can effectively reduce hepatic transaminase levels and significantly improve urea, albumin production, and survival rate in rabbit ALF models is demonstrated. Thus, it is believed that the biomimetic microcarriers and their derived bioreactor may hold potential for clinical applications in managing ALF and other liver diseases.
Article
Accurate detection of biomarkers is essential for disease diagnosis. Although the highly sensitive fluorescence probes are feasible for the above goal, it is typically interfered by the auto-fluorescence and light scattering of the biological samples. Photochemical afterglow system (consisting of photosensitizer, afterglow substrate and emitter) based on cascade photochemical reactions exhibits long-lived luminescence (seconds to hours), thus avoiding background interference. With the assistance of polymers, such as polystyrene microspheres, the photochemical afterglow systems have been transformed into homogeneous and water-soluble nanoparticles, and used for in-vitro biomarker analysis. Here, we summarized the principle, preparation and applications of these afterglow nanoparticles, and evaluated their performance in clinical sample testing by comparing with other nanoparticle-based methods. Finally, several problems and possible solutions of afterglow nanoparticle-based methods in biomarker determination were also mentioned.
Article
Full-text available
Implantable and wearable bioelectronic systems are arising growing interest in the medical field. Linking the microelectronic (electronic conductivity) and biological (ionic conductivity) worlds, the biocompatible conductive materials at the electrode/tissue interface are key components in these systems. We herein focus more particularly on resorbable bioelectronic systems, which can safely degrade in the biological environment once they have completed their purpose, namely, stimulating or sensing biological activity in the tissues. Resorbable conductive materials are also explored in the fields of tissue engineering and 3D cell culture. After a short description of polymer-based substrates and scaffolds, and resorbable electrical conductors, we review how they can be combined to design resorbable conductive materials. Although these materials are still emerging, various medical and biomedical applications are already taking shape that can profoundly modify post-operative and wound healing follow-up. Future challenges and perspectives in the field are proposed.
Article
Optical microcavities are compact structures that confine resonant photons in microscale dimensions for long periods of time, greatly enhancing light–matter interactions. Plentiful and profound physical mechanisms within these microcavities or functional microcavities have been extensively explored, including mode shift/splitting/broadening, lasing and gain enhancements, surface plasmon resonance, fluorescence resonance energy transferring, optical frequency comb spectroscopy, optomechanical interaction, and exceptional point. The versatility in design and the diverse range of materials, particularly composites involving metals and 2-dimensional materials, have paved a way for innovative approaches and improved performance in biochemical sensing applications. Leveraging the advantages ranging from miniaturization, high sensitivity, rapid response, and inherent stability, optical microcavity-based biochemical sensors have emerged to address the growing and increasingly complex demands of biochemical detection. This review commences with an exploration of fundamental mechanisms and structures and then delves into typical applications in recent advancements, covering the detection of biomacromolecules, cells, solid particles, liquid ions, and gas molecules. This review also culminates with a forward-looking perspective, highlighting future development trends and crucial research directions.
Article
Supercapacitors and transistors are two key devices for future electronics that must combine portability, high performance, easy scalability, etc. Graphene‐related materials (GRMs) are frequently chosen as active materials for these applications given their unique physical properties that are tunable via chemical functionalization. Up to date, among GRMs, only reduced graphene oxide (rGO) showed sufficient versatility and processability in mild media, rendering it suitable for integration in these two types of devices. Here, a sound alternative to rGO is provided, namely graphene acetic acid (GAA), whose physico‐chemical features offer specific advantages. In particular, the use of a GAA‐based cathode in a zinc hybrid supercapacitor (Zn‐HSC) delivers state‐of‐the‐art gravimetric capacitance of ≈400 F g ⁻¹ at a current density of 0.05 A g ⁻¹ . Conversely, GAA‐based LGT, supported onto Si/SiO 2 , shows an ambipolar behavior in 0.1 m NaCl, featuring a clear p‐doping quantified by Dirac voltage higher than 100 mV. Such a device is successfully implemented in paper fluidics, thereby demonstrating the feasibility of real‐time monitoring.
Article
Full-text available
Stress granules (SGs) are cytosolic biomolecular condensates that form in response to cellular stress. Weak, multivalent interactions between their protein and RNA constituents drive their rapid, dynamic assembly through phase separation coupled to percolation. Though a consensus model of SG function has yet to be determined, their perceived implication in cytoprotective processes (e.g., antiviral responses and inhibition of apoptosis) and possible role in the pathogenesis of various neurodegenerative diseases (e.g., amyotrophic lateral sclerosis and frontotemporal dementia) have drawn great interest. Consequently, new studies using numerous cell biological, genetic, and proteomic methods have been performed to unravel the mechanisms underlying SG formation, organization, and function and, with them, a more clearly defined SG proteome. Here, we provide a consensus SG proteome through literature curation and an update of the user-friendly database RNAgranuleDB to version 2.0 (http://rnagranuledb.lunenfeld.ca/). With this updated SG proteome, we use next-generation phase separation prediction tools to assess the predisposition of SG proteins for phase separation and aggregation. Next, we analyze the primary sequence features of intrinsically disordered regions (IDRs) within SG-resident proteins. Finally, we review the protein- and RNA-level determinants, including post-translational modifications (PTMs), that regulate SG composition and assembly/disassembly dynamics.
Article
Full-text available
Current healthcare practices are reactive and use limited physiological and clinical information, often collected months or years apart. Moreover, the discovery and profiling of blood biomarkers in clinical and research settings are constrained by geographical barriers, the cost and inconvenience of in-clinic venepuncture, low sampling frequency and the low depth of molecular measurements. Here we describe a strategy for the frequent capture and analysis of thousands of metabolites, lipids, cytokines and proteins in 10 μl of blood alongside physiological information from wearable sensors. We show the advantages of such frequent and dense multi-omics microsampling in two applications: the assessment of the reactions to a complex mixture of dietary interventions, to discover individualized inflammatory and metabolic responses; and deep individualized profiling, to reveal large-scale molecular fluctuations as well as thousands of molecular relationships associated with intra-day physiological variations (in heart rate, for example) and with the levels of clinical biomarkers (specifically, glucose and cortisol) and of physical activity. Combining wearables and multi-omics microsampling for frequent and scalable omics may facilitate dynamic health profiling and biomarker discovery.
Article
Full-text available
It is challenging to balance high biocompability with good mechanical‐electrical sensing performance, especially when triggered inflammatory stress response after in‐vivo implantation. Herein, we reported a bioinspired wrinkle‐reinforced adaptive nanoclay‐interlocked soft strain‐sensor based on a highly stretchable and elastic ionic‐conductive hydrogel. This novel nanoclay‐composite hydrogel exhibited excellent tensile properties and high sensing capacity with steady and reliable sensing performance due to the structural‐mechanical‐electrical integrity of the nanoclay crosslinked and nano‐reinforced interpenetrating network. The incorporation of amphiphilic ions provides the hydrogel with significant protein resistance, reducing its non‐specific adsorption to proteins upon implantation and improving its biosafety as an implanted device and maintaining the authenticity of the sensing results. Based on the revealed sensing enhanced mechanism based on hierarchical ordered structures, as a proof‐of‐concept application, it is for the first time to demonstrate this hydrogel sensor can accurately localize the region where myocardial infarction occurs and may become a novel strategy for real‐time monitoring of pathological changes in heart disease. This article is protected by copyright. All rights reserved
Article
Full-text available
Biosensors based on graphene field-effect transistors have become a promising tool for detecting a broad range of analytes. However, their performance is substantially affected by the functionalization protocol. In this work, we use a controlled in-vacuum physical method for the covalent functionalization of graphene to construct ultrasensitive aptamer-based biosensors (aptasensors) able to detect hepatitis C virus core protein. These devices are highly specific and robust, achieving attomolar detection of the viral protein in human blood plasma. Such an improved sensitivity is rationalized by theoretical calculations showing that induced polarization at the graphene interface, caused by the proximity of covalently bound molecular probe, modulates the charge balance at the graphene/aptamer interface. This charge balance causes a net shift of the Dirac cone providing enhanced sensitivity for the attomolar detection of the target proteins. Such an unexpected effect paves the way for using this kind of graphene-based functionalized platforms for ultrasensitive and real-time diagnostics of different diseases.
Article
Full-text available
The incidence of prostate cancer (PCa) in men globally increases as the standard of living improves. Blood serum biomarker prostate‐specific antigen (PSA) detection is the gold standard assay that do not meet the requirements of early detection. Herein, a solution‐gated graphene transistor (SGGT) biosensor for the ultrasensitive and rapid quantification detection of the early prostate cancer‐relevant biomarker, miRNA‐21 is reported. The designed single‐stranded DNA (ssDNA) probes immobilized on the Au gate can hybridize effectively with the miRNA‐21 molecules targets and induce the Dirac voltage shifts of SGGT transfer curves. The limit of detection (LOD) of the sensor can reach 10⁻²⁰ M without amplification and any chemical or biological labeling. The detection linear range is from 10⁻²⁰ to 10⁻¹² M. The sensor can realize real‐time detection of the miRNA‐21 molecules in less than 5 min and can well distinguish one‐mismatched miRNA‐21 molecule. The blood serum samples from the patients without RNA extraction and amplification are measured. The results demonstrated that the biosensor can well distinguish the cancer patients from the control group and has higher sensitivity (100%) than PSA detection (58.3%). Contrastingly, it can be found that the PSA level is not directly related to PCa.
Article
COVID-19, caused by the severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2), originated a global health crisis, causing over 2 million casualties and altering human daily life all over the world. This pandemic emergency revealed the limitations of current diagnostic tests, highlighting the urgency to develop faster, more precise and sensitive sensors. Graphene field effect transistors (GFET) are analytical platforms that enclose all these requirements. However, the design of a sensitive and robust GFET is not a straightforward objective. In this work, we report a GFET array biosensor for the detection of SARS-CoV-2 spike protein using the human membrane protein involved in the virus internalisation: angiotensin-converting enzyme 2 (ACE2). By finely controlling the graphene functionalisation, by tuning the Debye length, and by deeply characterising the ACE2-spike protein interactions, we have been able to detect the target protein with an extremely low limit of detection (2.94 aM). This work set the basis for a new class of analytical platforms, based on human membrane proteins, with the potential to detect a broad variety of pathogens, even before their isolation, being a powerful tool in the fight against future pandemics.
Article
With the rapid development of wearable electronic devices, health monitoring is undergoing a fundamental shift from hospital-centered treatment to patient-centered diagnosis. Solution-gated graphene transistors provide an effective platform for developing high-sensitivity wearable devices due to their unique signal amplification, low energy consumption, and compatibility for miniaturization. However, it is still a major challenge to perform real-time sweat composition monitoring directly on the dry skin surface. In this work, a skin-based flexible gel electrolyte graphene transistor (GEGT) was successfully designed and fabricated for glucose detection, consisting of a gate electrode decorated with Au nanoparticles modified reduced graphene oxide (AuNPs/RGO) nanocomposites and a monolayer graphene channel. Glycerin gel was used to replace the traditional liquid electrolyte, not only could better fit the human skin, but also play the role of fluid collection, providing stable testing conditions for the sensor. Based on the high electron mobility of graphene channel and the excellent electrocatalytic performance of AuNPs/RGO nanocomposites, the constructed GEGT sensor exhibits excellent sensing performance for glucose with good selectivity, low operating voltage (0.5 V), wide detection range (10 nM - 25 mM), and low detection limit (10 nM). The device maintains stable performance after up to 1000 bending cycles with a bending radius of 4 mm. In addition, the GEGT sensor displays good accuracy in sweat detection and sensitive dynamic response during actual wearing, which provides a guarantee for the construction of wearable transistor devices and real-time health tracking.
Article
The disease caused by severe acute respiratory syndrome coronavirus, SARS-CoV-2, is termed COVID-19. Even though COVID-19 has been out for more than two years, it is still causing a global pandemic. Due to the limitations of sample collection, transportation, and kit performance, the traditional reverse transcription-quantitative polymerase chain reaction (RT-qPCR) method has a long detection period and high testing costs. An increased risk of infection is inevitable, since many patients may not be diagnosed in time. The CRISPR-Cas13a system can be designed for RNA identification and knockdown, as a promising platform for nucleic acid detection. Here, we designed a solution-gated graphene transistor (SGGT) biosensor based on the CRISPR-Cas13a system. Using the gene-targeting capacity of CRISPR-Cas13a and gate functionalization via multilayer modification, SARS-CoV-2 nucleic acid sequences can be quickly and precisely identified without the need for amplification or fluorescence tagging. The limit of detection (LOD) in both buffer and serum reached the aM level, and the reaction time was about 10 min. The results of the detection of COVID-19 clinical samples from throat swabs agree with RT-PCR. In addition, the interchangeable gates significantly minimize the cost and time of device fabrication. In a nutshell, our biosensor technology is broadly applicable and will be suitable for point-of-care (POC) testing.
Article
False detection of SARS-CoV-2 is detrimental to epidemic prevention and control. The scalar nature of the detected signal and the imperfect target recognition property of developed methods are the root causes of generating false signals. Here, we reported a collaborative system of CRISPR-Cas13a coupling with the stabilized graphene field-effect transistor, providing high-intensity vector signals for detecting SARS-CoV-2. In this collaborative system, SARS-CoV-2 RNA generates a “big subtraction” signal with a right-shifted feature, whereas any untargets cause the left-shifted characteristic signal. Thus, the false detection of SARS-CoV-2 is eliminated. High sensitivity with 0.15 copies/μL was obtained. In addition, the wide concerned instability of the graphene field-effect transistor for biosensing in solution environment was solved by the hydrophobic treatment to its substrate, which should be a milestone in advancing it's engineering application. This collaborative system characterized by the high-intensity vector signal and amazing stability significantly advances the accurate SARS-CoV-2 detection from the aspect of signal nature.