SlideShare a Scribd company logo
1 of 16
Download to read offline
Confinement-guided ultrasensitive optical assay with artificial
intelligence for disease diagnostics
Wenjing Zhang,1,6
Yongfeng Lu,1,2,3,6
Chenyi Su,1
Yibo Wang,1
Yong-Fei Wang,1
Bo Zhang,1
Cheng Jiang,1,* Keying Guo,4,5,* and Chuan Xu2,3,*
*Correspondence: jiangcheng@cuhk.edu.cn (C.J.); keying.guo@gtiit.edu.cn (K.G.); xuchuan100@163.com (C.X.)
Received: May 31, 2023; Accepted: August 31, 2023; Published Online: September 1, 2023; https://doi.org/10.59717/j.xinn-med.2023.100023
© 2023 The Author(s). This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
GRAPHICAL ABSTRACT
PUBLIC SUMMARY
■ Advanced optical assays utilize external and internal confinement strategies for disease biomarker detection.
■ Convergence of artificial intelligence (AI) with optical assays boosts diagnostic accuracy and prediction of disease paths.
■ Embracing biotechnology and information technology permits the revolution of biomarker discovery.
Medicine
REVIEW
The Innovation Medicine 1(2): 100023, September 21, 2023 1
Confinement-guided ultrasensitive optical assay with artificial
intelligence for disease diagnostics
Wenjing Zhang,1,6
Yongfeng Lu,1,2,3,6
Chenyi Su,1
Yibo Wang,1
Yong-Fei Wang,1
Bo Zhang,1
Cheng Jiang,1,* Keying Guo,4,5,* and Chuan Xu2,3,*
1
School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China
2
Department of Oncology & Cancer Institute, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital Chengdu, Sichuan 610072, China
3
Department of Laboratory Medicine and Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic
Science and Technology of China, Chengdu 610054, China
4
Biotechnology and Food Engineering, Guangdong Technion-Israel Institute of Technology (GTIIT), Shantou 515063, China
5
Faculty of Biotechnology and Food Engineering, Technion-Israel Institute of Technology (IIT), Haifa 3200003, Israel
6
These authors contributed equally
*Correspondence: jiangcheng@cuhk.edu.cn (C.J.); keying.guo@gtiit.edu.cn (K.G.); xuchuan100@163.com (C.X.)
Received: May 31, 2023; Accepted: August 31, 2023; Published Online: September 1, 2023; https://doi.org/10.59717/j.xinn-med.2023.100023
© 2023 The Author(s). This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
Citation: Zhang W., Lu Y., Su C., et al., (2023). Confinement-guided ultrasensitive optical assay with artificial intelligence for disease diagnostics. The Innovation Medicine 1(2), 100023.
The necessity for ultrasensitive detection is becoming increasingly appar-
ent as it plays a pivotal role in disease early diagnostics and health
management, particularly when it comes to detecting and monitoring low-
abundance biomarkers or precious samples with tiny volumes. In many
disease cases, such as cancer, infectious disease, autoimmune disorder,
and neurodegenerative disease, low-abundant target biomarkers like circu-
lating tumor cells (CTCs), extracellular vesicle (EV) subpopulations, and
post-translational modified proteins (PTMs) are commonly existing and
can be served as early indicators of disease onset or progression. However,
these biomarkers often exist in ultra-low quantities in body fluids, surpass-
ing the detection limits of conventional diagnostic tools like enzyme-linked
immunosorbent assay (ELISA). This leads to the inability to probe disease
evolution at a very early stage from molecular pathology perspective. In
such regard, ultrasensitive optical assays have emerged as a solution to
overcome these limitations and have witnessed significant progress in
recent decades. This review provides a comprehensive overview of the
recent advancements in ultrasensitive optical detection for disease diag-
nostics, particularly focusing on the conjunction of confinement within
micro-/nano-structures and signal amplification to generate distinguish-
able optical readouts. The discussion begins with a meticulous evaluation
of the advantages and disadvantages of these ultra-sensitive optical
assays. Then, the spotlight is turned towards the implementation of artifi-
cial intelligence (AI) algorithms. The ability of AI to process large volumes
of visible reporter signal and clinical data has proven invaluable in identify-
ing unique patterns across multi-center cohort samples. Looking forward,
the review underscores future advancements in developing convergent
biotechnology (BT) and information technology (IT) toolbox, especially opti-
cal biosensors for high-throughput biomarker screening, point-of-care
(PoC) testing with appropriate algorithms for their clinical translation are
highlighted.
INTRODUCTION
Biomarkers discovered and validated from biological fluids hold great
potential for reflecting the molecular pathology and progression of diseases.1-3
Compared to imaging techniques, e.g., computed tomography (CT),4,5
magnetic resonance imaging (MRI)6,7
which highly rely on well-trained clini-
cian to identify nidus of milli-meter to centimeter dimension,8,9
biomarkers
based in vitro diagnostics (IVD) can offer a range of advantages in disease
diagnosis and monitoring,10-13
including high sensitivity,14
real-time monitor-
ing,15-19
facilitation of personalized treatment,20,21
and cost-effectiveness.22,23
However, biomarkers like post-translation modification (PTM) proteins,24-26
microRNA,27-31
extracellular vesicle (EV) subpopulations,19,32-36
circulating
tumor cells (CTCs),37,38
and circulating nucleic acids39,40
are usually present in
low abundance, and their secretion is rare during the early stage of disease
progression, posing significant challenges for the conventional detection
methodologies like enzyme-linked immunosorbent assays (ELISA)41,42
due to
their limited sensitivity and unachievable stable monitoring. Usually, compro-
mised strategies have to be used, such as consumption and conversion of
large input volume into a squeezed detection volume to achieve a decent
enrichment factor. This not only increases the economic and psychological
burden on patients associated with multiple sampling but also constrains the
widespread adoption and implementation of biomarkers in clinical applica-
tions. In such regard, the development of ultrasensitive toolboxes is essential
with high demand.
The pressing need for ultrasensitive detection methods in the realm of
biomarker analysis has led researchers to explore various innovative strate-
gies. Fundamentally, ultrasensitive detection either relies on high-end detec-
tion devices or signal amplification strategies. The former category includes
advanced instruments such as total internal reflection fluorescence (TIRF)
microscopy43,44
and cutting-edge flow cytometry analyzers.45,46
While these
state-of-the-art tools provide exceptional sensitivity and accuracy, their
widespread accessibility may be hindered by factors such as high costs and
the need of specialized expertise in operation and maintenance. The latter, on
the other hand, serves as the primary focus of this review, offering a more
promising and accessible avenue for exploration and translation.
Typically, signal amplification with confinement strategies, e.g., multiple
enzymatical catalysis, nucleic acid amplification, and surface plasmon-
enhanced fluorescence, involve generating sufficiently robust output signals,
generally achieved by confining signal molecules within minuscule spaces.
Such confinement can be realized either using semi-enclosed microwells,
fully enclosed water-in-oil droplets, or by immobilizing ultra-bright or visible
probes on the surfaces of particulate substrates to provide localized tremen-
dous reporter concentration and signal readout (Figure 1). These methodolo-
gies can enhance signal detection and improve the overall sensitivity of the
method, ultimately pushing the detection limits down to even single-entity
level.
In this review, we primarily focus on the development of ultrasensitive opti-
cal sensors, emphasizing the “adhesion” of visible reporters to the outer or
internal microenvironment, presenting external- and internal- confinement
strategies, respectively. Generally, plasmonic47-52
or non-plasmonic materials
can be employed to convert target binding events into digitized signal format
(Figure 1). How artificial intelligence facilitates image processing, particle
counting, and clinical data handling will be emphasized. We introduce repre-
sentative cases, discussing their pivotal features and shortcomings. Finally,
we delve into the prospective developments both in AI technologies and in
the field of ultrasensitive optical detection. Our vision is that the synergy of
these advancements will significantly contribute to reshaping diagnostic
approaches in healthcare, offering a more precise, efficient, and reliable anal-
ysis.
EXTERNAL CONFINEMENT STRATEGY
To date, diverse types of optical reporters have been investigated with their
confinement, either on the outer space or internal space of a nano-/micro-
architecture, to achieve ultra-sensitivity. Therefore, these strategies can be
mainly categorized into two domains: internal confinement and external
confinement.
External confinement, namely restraint of optical reporters (e.g., chemical
dyes, plasmonic particles) to the outer surface of solid support (e.g., spheri-
cal beads). The binding of optical reporter molecules can usually be executed
by various approaches like DNA hybridization, covalent binding, etc. Generally,
REVIEW
2 The Innovation Medicine 1(2): 100023, September 21, 2023 www.the-innovation.org/medicine
in such system, reporter molecules with high-density would generate local-
ized high signal intensity on the individual beads that can “illuminate” the
single-bead microenvironment under excitation, resulting in superior sensitiv-
ity. Such “ultrabright” beads can be easily detected using conventional micro-
scope with low-magnification even mobile phone-based microscope. Differ-
ent feasible algorithms can then be employed for beads digital counting,
which can be converted to the target analyte concentration using appropriate
algorithm. The screened biomarker can be used for toward separation of
different groups in a cohort study to permit disease early diagnostics, stratifi-
cation, therapy monitoring, etc.53-56
So far, plasmonic- and nonplasmonic-
materials have been widely used as optical labels in cooperation with
confinement strategy for ultrasensitive assay development, which will be
discussed in the following sections.
Nanoplasmonics based reporter
When considering the direct signal output, plasmonic objects with size
larger than ~30 nm can be visualized under dark-field microscopy (DFM)
due to their strong scattering capabilities. This arises from the localized
surface plasmon resonance (LSPR) effect. When the size of the plasmonic
particles exceeds a certain threshold—yet remains smaller than the wave-
length of light—there is a significant enhancement in light scattering.48,57-60
On the other hand, plasmon can also enhance fluorescence through surface
plasmon interactions by mediating the distance (e.g. adjusting the spacer
thickness) between the plasmonic materials and the fluorophore. These
interactions amplify the local electromagnetic field. This, in turn, increases the
excitation rate and the quantum yield of the fluorophores, resulting in ulti-
mately enhanced fluorescence signals.61,62
These intrinsic advantages permit
plasmonics-related detection with achievable ultrasensitivity. The most
widely explored formats are plasmonic label by its alone and surface plas-
mon-enhanced fluorescence.
Nanoplasmonics alone. The surface plasmon effect refers to the collec-
tive oscillation of electrons at the interface
between a metal and a dielectric material, lead-
ing to unique optical phenomena.49
Dark-field
spectroscopy (DFS) is a typical plasmonic parti-
cle spectroscopy technique that employs
microscopy in combination with a spectrometer
to observe plasmonic particles.63,64
Lee group65
developed a plasmonic exosome assay using
transmission surface plasmon resonance with
antibodies functionalized nanohole arrays to
identify ovarian cancer exosomal surface
biomarkers (Figure 2A). They demonstrated
improved signal amplification through
secondary labeling with nanoprobes such as
gold nanostars of variable sizes and dimen-
sions for detection of 12 surface markers. The
sensitivity enhancement factors were 104
-fold
and 102
-fold compared to Western blot and
chemiluminescence ELISA, respectively. The
plasmonic structure sensing mechanism was
confirmed through Three-Dimensional Finite-
Difference Time-Domain simulations (3D-FDTD)
simulations, showing enhancement with the
immune-sandwich format on a glass substrate.
By using this technique, exosomal EpCAM and
CD24 were stably probed, and were demon-
strated the ability to differentiate ovarian cancer
patients (n=20) from non-cancer patients
(n=10).
In another work, Hu and co-workers66
built a
nanoplasmon-enhanced scattering (nPES)
assay to directly probe the membrane antigens from tumour-derived EVs, in
which the detection antibody-conjugated gold nanospheres (50 nm) and
nanorods (25×60 nm) were bind to EVs, which were captured by antibodies
pre-decorated glass chip to form the “sandwich” format, producing a local
plasmon effect that enhances tumour-derived EV detection sensitivity and
specificity (Figure 2B). The unique plasmonic color of gold nanoparticles
(AuNPs) and gold nanorods (AuNRs) allows the duplexed profiling of EV
membrane marker, and their coupling caused red shift and color change can
indicate the co-existence of the membrane antigens. They leveraged the
Image J to analyze batches of DFM images. The method can automatically
selected image areas with a brightness value of 255. After identifying these
areas, the software calculated the ratios between the selected areas and the
total areas of the images. These ratios were used as a metric for the specific
nPES EV signal. In their study, the authors demonstrated that nanoplasmon-
enhanced scattering assay could effectively distinguish early-stage pancre-
atic cancer from pancreatitis and controls in a cohort of 155 subjects.
However, the assay capacity for high-throughput surface marker profiling is
limited considering the proximity of surface antigens and the steric hindrance
of AuNPs and AuNRs during recognition and binding to the exosomal
surface.
Gooding group67
developed a core-satellite formation of Au nanoparticles
(i.e., 67 nm-AuNP-analyte-enzyme coated 10 nm-AuNP) for the detection of
interleukin 6 (IL-6). They utilized a CMOS-equipped digital camera and a dark-
field microscope to analyze thousands of dropcasted-gold nanospheres
within seconds using Image J (Figure 2C). The research team developed a
MATLAB script to transform images from the RGB color space into the HSV
(i.e. Hue, Saturation, Value) color space, facilitating the separation of intensity
and color variables. Following this conversion, a bandpass filter was applied
to remove noisy pixels and enhance the image quality. The next step involved
the identification of single nanoparticles based on their distinct morphology
and intensity of the point-spread-functions. Each identified nanoparticle was
Medicine
Figure 1. Schematic illustration of confinement
based ultrasensitive optical detection
REVIEW
The Innovation Medicine 1(2): 100023, September 21, 2023 3
then labeled, and its color data (hue) is extracted using an intensity-weighted
averaging method. This process entails creating a mask around the pixel with
the highest intensity within a 3-pixel radius. By employing this robust
process, the research team can independently count nanoparticles of various
colors, enabling a comprehensive assessment of the nanoparticle population
in the image. Compared to the LoD of ELISA and Western blot (~0.5 ng/mL),
this dark-field imaging approach shows a 50-fold decrease in LoD due to the
plasmon coupling. This method can circumvent the influence of non-specific
adsorption due to the localized surface plasmon resonance (LSPR) sandwich
assay, which is tolerant to non-specific binding effects, a major problem in
the field of biosensing.
Wang group68
designed another plasmonic nanobiosensor based on
Au@Ag core-shell structure for detecting microRNA 21 (miR-21) (Figure 2D).
After capturing miR-21, an average localized surface plasmon resonance
(LSPR) scattering wavelength shift of about 0.4 nm can be obtained. In this
system, the “pyramidal” DNA tetrahedral structure was utilized as the recog-
nition probe for miR-21 capped on noble-metal nanoparticles (e.g., gold or
silver nanoparticles) by employing the shifts of the LSPR scattering spec-
trum peak (λmax) as the output signal, permitting real-time detection of miR-
21 with sensitivity at the aM level within a large dynamic range of 1 aM ‒ 1
nM. However, the DNA-microarray platform could only give rise to tiny LSPR
scattering spectral wavelength shift (~0.4 nm) on this nanobiosensor, which
may not be enough to be captured intuitively by a common instrument.
Plasmon coupled with fluorescence. Noble metal-based plasmonic parti-
cles can not only provide signal output by themselves, but also have the
unique ability to couple with other fluorescent emission luminogens or quan-
tum dots to enhance emission properties, permitting dark-filed and fluores-
cence dual-channel signal output. The physicochemical properties of plas-
monic nanoparticles are specifically designed to facilitate plasmon-enhanced
fluorescence (PEF) pathway,49
which can significantly enhance the fluores-
cence of nearby fluorophore zone using spacers like SiO2 or polymers with
appropriate manipulation for the avoidance of quenching. While conventional
fluorescent molecules and optical nanoparticles suffer from low brightness,
poor photostability, and photobleaching, plasmon-enhanced fluorescence
offers improved performance in terms of higher sensitivity and long-term
photo-stability. Quantum dots (QDs) are a promising class of fluorescent
probes for biological imaging owing to their unique stokes shift, powerful
stability, and relatively long fluorescence lifetime. However, the fluorescence
intensity of a single quantum dot is not sufficient for observation under a
conventional optical microscope. Wang group51
presented bioorthogonal
nanoparticle detection (BOND) technology in a PEF-based suspension
microarray using QDs as a signal reporter. The spherical gold-coated
magnetic beads and flat gold film-coated glass were compared and served
as solid support. They were decorated with anti-PSA (prostate-specific anti-
gen) capture antibodies, followed by recognition and binding of the target
analyte and the QDs-modified detection antibody, generating the final sand-
wich immunocomplex. After collection of fluorescence images of the plas-
monic slide, the imagines were analyzed by software Genepix 6.1. This soft-
ware can automatically identify the features and report the fluorescence
intensities of each spot with background correction. After collecting the
intensity data for each point, the mean fluorescence intensity was calculated
for each set of spots, which can be converted to original concentration using
Poisson equation. The designed platform utilizes BOND- and PEF- amplifica-
tion strategies for ultrasensitive detection of proteins with detection sensitiv-
ity (1 fM) and dynamic range (5 orders of magnitude) was achieved for PSA
detection (Figure 3A). They demonstrated that both spherical and flat plas-
monic surfaces can enhance the fluorescence of QDs, and the immunocom-
plex served as an excellent spacer.
To ensure multiplexed profiling, Shao group62
developed a templated plas-
monics for exosomes (TPEX) platform for multiparametric exosome analysis.
This advanced method has been employed to analyze samples from 20
subjects (12 with colorectal cancer and 8 with gastric cancer), and has
demonstrated its potential in predicting patient prognosis using exosomal
proteins (MUC1, EpCAM, CD24, CD63) (Figure 3B). Exosomes were incubated
Figure 2. Ultrasensitive plasmonic detection strategies (A) Detection of tumor-derived extracellular vesicles near a metallic array, with FDTD simulations highlighting confined
electromagnetic fields near nanopores.65
(Nature Biotechnology, 2014). (B) Nanoplasmon-enhanced scattering (nPES) assay for EVs detection.66
(copyright Springer Nature,
2017). (C) Core-satellite immunoassay for the detection of IL-6.67
(copyright Elsevier Ltd., 2018). (D) Smart nanobiosensor for single molecule analysis using tsDNA17-modified
Au@Ag nanocubes on an ITO glass to capture miR-21.68
(copyright American Chemical Society, 2018).
REVIEW
4 The Innovation Medicine 1(2): 100023, September 21, 2023 www.the-innovation.org/medicine
with fluorescent molecular probes and AuNPs. The exosome-bound AuNP
can develop into a gold nanoshell-coated exosomes by seeding method, and
the authors demonstrated that a large red shift in its plasmonic resonance
can effectively quench the fluorescence signal of probes attached to the
same vesicle. Besides, all the experimental steps were integrated into the
miniaturized microfluidic chip that facilitated TPEX measurements of target in
the complex clinical biofluids. A custom-designed smartphone-based optical
detector can then be used for the image-based data acquisition and analysis,
which can be completed within 15 min.
Recently, Singamaneni and co-authors52
showed that a plasmonic
nanoscale construct based on bovine serum albumin (BSA)-fluorophore
decorated AuNRs which served as an “add-on” label for a broad range of
bioassays with improved signal-to-noise ratio and dynamic range without
altering their workflow and readout devices (Figure 3C). In this work, BSA was
used as a scaffold in the design of nanoconstructs, acting as a stabilizing
agent to prevent nanoconstruct aggregation and as a blocking agent to mini-
mize non-specific binding of the plasmonic-fluor. Compared to ELISA, the
presented plasmonic assay exhibited 189-fold lower LOD and more than two
orders of magnitude larger dynamic range. Such device was successfully
utilized to scan an array of biomarkers associated with kidney disease.
However, it should be noted that the exposed fluorophore on the AuNRs
surface may be suffered from photostability, a protection shell would be
favorable. Zheng and co-workers fabricated Ag@SiO2 core-shell structure
dopped with FITC, which are single ultrabright green particles that can be
visualized by a conventional fluorescence microscope. This system permits
long-term stability (6 months) with negligible attenuation of fluorescence
intensity due to the silica shell protection and the resistance to the dye
desorption.63
Magnetic beads and nucleic acid amplification based detection
In addition to the plasmonic reporters, there are other non-plasmonic
reporters available, which can be complementary candidates and have been
actively explored for ultrasensitive assay. In the context of using fluores-
cence probes for ultrasensitive detection, traditional method using repeated
labeling of fluorophores is often laborious and can lead to photobleaching,
false-positive signal as well as a low signal-to-noise ratio69
. Consequently,
there is a pressing need to amplify the fluorescence signal toward single-
entity detection in a robust way. In such regard, nucleic acid-labelled fluo-
rophore-based detection probe in cooperation with solid supports like
magnetic beads is a good option. Nucleic acid amplification (NAA) is a valu-
able technique for detecting pathogens or other biomolecules by amplifying
specific nucleic acid sequences.70
Numerous isothermal NAA techniques,
including the catalyzed hairpin assembly (CHA),71
hybridization chain reac-
tion (HCR),72
and rolling circle amplification (RCA),73,74
have emerged as
promising alternatives for achieving rapid and efficient signal amplification
without the need of thermocycling. The Walt group75
developed an ultrasen-
sitive single-molecule detection platform that employed rolling circle isother-
mal amplification and a sandwich structure for Brachyury detection, achiev-
ing an impressive sensitivity of 244.6 aM (Figure 4A). In this design, magnetic
microbeads (MBs) conjugated with antibodies were used to probe human
cytokines interleukin-1β (IL-1β) and IL-10. Then, single-stranded DNA
(ssDNA) with a specific detection antibody and an RCA trigger were used to
capture target exosomes. The RCA reaction enabled numerous fluorescent
probes to hybridize with the RCA products, amplifying fluorescence signals.
Beads were then placed on a slide and dried, forming monodispersed beads
for digital counting using colocalized red (MBs)-green (probe) fluorescence.
Following the similar principle, the Gao group76
introduced modifications by
using an aptamer as a recognition element instead of antibodies. They
employed an addressable DNA nanoflower (DNF) attached to the surface of
magnetic beads, which introduced a large number of fluorescence probes
onto the nanoflower through base complementary pairing. This configuration
enabled fluorescence imaging for the detection of 17β-estradiol (E2). a typi-
cal environmental estrogen, and a small molecule model target (Figure 4B).
The fluorescence image was automatically analyzed by Fiji image analysis
Medicine
Figure 3. Strategies utilizing plasmonic coupled fluorescence (A) BOND overview and its role in plasmonic suspension microarray.51
(copyright Elsevier Ltd., 2023).
(B) Schematic of plasmonic-fluor as a biolabel boosting fluorescence intensity and improving signal-to-noise in assays.62
(copyright Springer Nature, 2020). (C) Templated plas-
monics for exosomes (TPEX) platform showing labeled exosomes for multiparametric molecular profiling through SPR redshift signals.52
(copyright Science Advances, 2020).
REVIEW
The Innovation Medicine 1(2): 100023, September 21, 2023 5
software, simplifying the testing workflow. The surface confinement strategy,
which involves using NAA and attaching optical probes to solid support, is
promising but comes with a set of challenges. The intricate and prolonged
wash procedure during binding and polymerization stages often results in
significant product loss. Moreover, the inherent negative charge of DNA tends
to non-specifically adsorb positively charged molecules, leading to undesir-
able false positives and high background interference. There is also a notable
heterogeneity in the brightness of individual beads due to different amplifica-
tion cycles, stemming from the sensitivity of enzyme activity to environmen-
tal influences.
To tackle these issues, appropriate antifouling strategies should be
employed, such as the use of nonionic and non-protein blockers to suppress
non-specific adsorption.77-79
They can help reduce DNA-induced false posi-
tive signals, thereby pushing the detection limit further and improving the
current fluorescence threshold. Automation for capture-detection would be
another solution to overcome the multi-step manual handling caused errors.
Additionally, rational fundamental design on sequences, the recently
proposed one-pot amplification strategy might be a good alternative for its
further evolution and improvement.80,81
It is anticipated that RCA can be
further improved by using signal enhancement and noise reduction
approaches. Finally, the ability to control the efficiency of aggregation on indi-
vidual magnetic beads results in discrepancies in fluorescence intensity
among beads, which can have a substantial influence on the outcomes. It is
worth noting that reactions such as RCA, LAMP, and PCR all require
enzymes, whose activity can be easily affected by the environment, leading to
low stability and assay accuracy. Hu et al.82
ingeniously utilized size encod-
ing approach to accomplish multiplexed detection by employing microbeads
of four different sizes, followed by decoration with aptamers to target extra-
cellular vesicle (EV) surface markers (Figure 4C). They designed a microflu-
idic chip featuring an array of differently sized channels, which allowed
magnetic beads of corresponding sizes to enter separate channels to reach
their blockade with spatial resolution. This combination of size coding and
spatial coding permitted simultaneous analysis of PD-L1, EpCAM, and CD63
from intact EVs for cancer diagnosis (healthy control n=10, cancer patients
n=25). RCA amplification with hybridization of complementary DNA probe
endowed the signal amplification capability. This study demonstrated that
Figure 4. Various single-molecule detection methods based on nucleic acid amplification (A) Schematic of dropcasted single molecule assays.75
(copyright American Chemi-
cal Society, 2020). (B) Principle of single-molecule detection of E2 used by addressable DNA nanoflowers and automatic digital quantitative analysis.76
(copyright American
Chemical Society, 2022). (C) Schematic of size-coded affinity matrix microfluidic chip strategy for multiplex EV phenotyping.82
(copyright American Chemical Society, 2022).
REVIEW
6 The Innovation Medicine 1(2): 100023, September 21, 2023 www.the-innovation.org/medicine
this tEV phenotyping method can rapidly and simultaneously detect six
different tEV phenotypes with high sensitivity (Table 1).
Visible microsphere
In the aforementioned discussion, signal amplification techniques are high-
lighted, which enables the conversion of undetectable signals into observ-
able fluorescent outputs through confinement-based signal enhancement.
This method involves directly doping reporters like fluorescent tags to micro-
spheres. The number of doped fluorophores and their intrinsic quantum yield
determines the brightness of resultant fluorescent microbeads. Alternatively,
the microbeads can be used as reporters as they can easily be visualized by
conventional microscope. Noji et al.83
introduced a sensitive wash-free detec-
tion method for PSA (Figure 5A). In their work, PSA reacted with antibody-
coated magnetic nanoparticles (~800 nm) and were then pooled into a
femtoliter-sized reactor by magnetic force. The images were analyzed using
Image J and Trackmate softwares. These tools can localize the center of
individual particles by calculating the intensity of light spots. As a result, the
number of tethered particles could directly correlate with the concentration of
the target antigen. The number of particles tethered was proportional to the
concentration of the target antigen. This method directly treated magnetic
microbeads as reporters without the need of additional dyes. However, this
method requires extensive mathematical calculations of the Brownian motion
of molecules and imaging validation through other instruments. Yiping Chen84
and his team encoded multiple targets using polystyrene microspheres with
five different particle sizes, and the unconjugated microspheres in the super-
natant after immunoreaction were recorded for subsequent visible signals
under an optical microscope (Figure 5B). The obtained images were further
analyzed using artificial intelligence computer vision technology to decode
the inherent characteristics of the polystyrene microspheres and revealed the
type and concentration of the target. This method sidesteps the complexities
of chip production and signal amplification but requires precise size control of
the microspheres. Non-specific adsorption and precipitation must also be
considered. The bright-field microscopy is straightforward but impurities or
dusts can interfere, offering opportunity for the fluorescently encoded micro-
spheres. To this end, He et al.85
directly labeled magnetic beads (~1 µm) with
fluorescent microspheres (~120 nm) to capture the target using a compo-
nents of capture DNA-bead, analyte, and detection DNA-fluorescent micro-
sphere in an order (Figure 5C). Using SARS-CoV-2 ssDNA as the model, they
achieved a 1.5 fM detection limit by counting and decoding bead-micro-
sphere pairs and analyzed the data with NIH ImageJ software.86
This study
highlights the potential of single-particle luminescence for single-molecule
detection. Noteworthy materials include aggregation-induced emission (AIE)
Medicine
Table 1. The typical examples using external-/internal- confinement strategy for ultrasensitive optical assay.
Confinement Type Analyte LOD Working Range Volume Ref.
External confinement Plasmonic alone CD63 NA NA 1 μL 62
External confinement Plasmonic alone EphA2 0.2 ng/μL 0.1–10000 ng/uL 1 μL 133
External confinement Plasmonic alone miR-21 0.1 aM 1 aM to 1 nM NA 68
External confinement Plasmonic with fluorescence ALP 5.8 μU/mL 0.06 to 2.48 mU/mL 100 μL 134
External confinement Plasmonic with fluorescence PSA 1 fM 1 fM-1 nM NA 135
External confinement Nucleic acid amplification 17β-estradiol (E2) 63.09 × 10−6
nM 0.001836 to 183.6 nM 10 μL 76
External confinement Nucleic acid amplification IFNγ and IL-2 (3×) 30 aM and 20 aM NA 2 μL 136
External confinement Nucleic acid amplification IL-6 13 pM NA 250 μL 137
External confinement Nucleic acid amplification PSA 97.2 aM 1.62 nm 2 μL 138
External confinement Nucleic acid amplification CEACAM-7 2.82 pM NA 25 μL 139
External confinement Nucleic acid amplification LwaCas13a 15.8 fM 10 cp/μL up to 106
cp/μL 2 μL 140
External confinement without amplification SARS-CoV-2 (ssDNA) 1.5 fM 0 to 1 pM 5 μL 85
External confinement without amplification CEA 1.2 pg/mL NA NA 141
External confinement without amplification Aflatoxin B1 0.17 ng/mL 3.13 to 125.00 ng/mL 15 μL 142
External confinement without amplification PSA 1.2 pg /mL 10 pg/mL and 1 ng/mL 100 μL 143
Internal confinement Droptlet(oil in water) β-gal NA 0.1 cpd-3 cpd(40-1202 fM) NA 144
Internal confinement Droptlet(oil in water) PSA 0.48 ng/mL 0.5–30 ng /mL 30 μL 145
Internal confinement Droptlet(oil in water) HPV’s target DNA 2000 copies/mL 107
copies/mL to 103
copies/mL 100 μL 92
Internal confinement Droptlet(oil in water) BβG 930 zM 1 aM to 1 fM 400 μL 146
Internal confinement Droptlet(oil in water) Exosomes 10−17
M 10-105
Exosomes/uL NA 93
Internal confinement Droptlet(hydrogel) IL-6 1.36 fM 0-50 fM 10 μL 147
Internal confinement microwell femtomolar proteins 2.84 fM 493 aM (32 fg/mL)-
120 fM (7.8 pg/mL)
2 μL 148
Internal confinement microwell Thyroid stimulating
hormone
0.0013 μIU/mL 0.02-1 μIU/mL 100 μL 100
Internal confinement microwell CD9+/CD63+ EVs NA 30-22800 ng/uL 3-8 μL 149
Internal confinement microwell serum cytokines 0.198pg/ml 0.16 pg/mL- 2.5 ng/mL <15 μL 103
ALP, alkaline phosphatase; PSA, prostate-specific antigen; β-Gal, β-galatosidase; BβG, biotinylated β-galactosidase; EphA2, Ephrin type-A receptor 2;
CEACAM-7, CEA Cell Adhesion Molecule 7
REVIEW
The Innovation Medicine 1(2): 100023, September 21, 2023 7
molecules embedded within metal-organic frameworks (MOFs) that intensify
fluorescence emission with the restraint movement by the MOF cage, making
it favorable optical reporter. Similarly, upconversion nanoparticles (UCNPs)87
are anti-Stokes luminescent materials that transform low-energy near-
infrared (NIR) radiation into higher-energy radiation. They are favorable for
detection with reduced interference and resistance to photobleaching.
Carbon dots (CDs)88,89
are photoluminescent nanomaterials that are water-
soluble, biocompatible, and photostable, fitting the need for biological and
environmental sample detection. Ongoing research in luminescent materials
will bolster single-particle detection, broadening its applications in diagnos-
tics, environmental monitoring, and chemical sensing.
To conclude, the external confinement strategies widely employ micro-/
nano- architectures with the adhesion of optical reporters (e.g., organic dyes,
plasmonic nanoparticles, quantum dots, etc.) on to their outer surface. In
cooperation with signal amplification strategies like nucleic acid amplifica-
tion or surface plasmon-enhanced fluorescence, the signal output can be
dramatically enhanced to permit the stable probing of target at ultra-low
levels, even single-entity level. It is expected that more novel formats on the
nanoarchitecture design or signal enhancement and the cooperation with
internal confinement will be introduced to achieve a synergetic effect, which
would be the next hot spot.
INTERNAL CONFINEMENT STRATEGY
Besides the external confinement strategy, the internal strategy has also
been actively explored as a vital complementary way. In such system, the
signal output reporters, e.g., fluorophores or catalyzed photonic/colorful
product, are usually confined within a small minuscule internal compartment
of an architecture like microwell, droplet, metal-organic-frame (MOF), hydro-
gel, etc. The signal output molecules are restricted, producing localized ultra-
high concentrations with a resultant amplified signal. In the case of droplet-
based systems, soft materials like emulsions or hydrogels are used to
confine the signal molecules, restricting the signal output within the liquid
droplet boundaries. For microwell-based systems, rigid materials such as
plastics or silicon are used to form an array of small wells, limiting the signal
output within the confined space of individual wells.
Confinement within droplet
In droplet system, mineral oil wrapped aqueous phase is commonly used.
By application of positive external pressure to the inlet or negative pressure to
the outlet generated through syringe pump90
or syringe with a binder clip91
,
respectively, the stable water-in-oil droplet format can be formed. Only if the
enzyme and substrate are encapsulated in the same droplet, the enzyme can
catalyze and produce a fluorescent product thus illuminate the droplet under
Figure 5. Various single-molecule detection methods based on microspheres (A) Digital HoNon-ELISA: Detection of protein biomarkers using antibody-coated magnetic
nanoparticles.83
(copyright American Chemical Society, 2019). (B) APT Immunoassay: AI-driven assay using 2D microspheres for multi-target detection.84
(copyright American
Chemical Society, 2023). (C) Enzyme-free Assay: Detects SARS-CoV-2 ssDNA with a bead-based sandwich structure.85
(copyright Elsevier Ltd, 2023).
REVIEW
8 The Innovation Medicine 1(2): 100023, September 21, 2023 www.the-innovation.org/medicine
excitation. Then, digital readout (“1” or “0”) can be generated once the
number of droplets is far more than that of the target analyte.92
Zheng et al.,93
utilized the capture antibody (anti-CD63)-detection antibody (anti-GPC-1) co-
localization strategy for the single-exosome detection with an achievable
LOD down to ~10−17
M and a dynamic range of 5 log of the linear regime
(Figure 6A). Although the droplet system using enzyme-catalyzed fluores-
cence within a confined volume can generate a strong signal for ultrasensi-
tive measurement and save sample consumption, it requires complex
manual operations, e.g., the stepwise capture processes must be done
outside the chip, and it only permits singplexing using current format. In such
regard, the capability for automation, integration, and multiplexed profiling is
favorable. Moreover, to further enhance the effectiveness of the droplet-
based chip, it is crucial to develop a matched mobile platform that facilitates
fast and easy access to assay data by integrating mobile device, thus high-
lighting the importance of improved connectivity and user-friendly interfaces.
The Walt team94
has developed a platform for rapid, sub-picogram-per-
milliliter, multiplexed digital droplet detection by using color-encoded beads
and droplet microfluidics comprising of partition-incubation-detection
compartments (Figure 6B). This innovative approach employs a mobile
phone-based imaging technique and coding-based duplexed profiling, elimi-
nating the need for costly benchtop optics and ensuring a detection speed
100 times faster than conventional assays due to time domain-encoded
mobile phone imaging. The performance of this assay was characterized by
spiking recombinant proteins in fetal bovine serum (FBS) with a limit of
detection (LOD) at 4 fg/mL (i.e., 300 aM). By integrating the coding-based
duplexed profiling, they further optimized the system performance, enhanc-
ing its detection accuracy and reliability. This development will aid its
commercialization, making a user-friendly system for self-testing anytime
and anywhere. Liang’s group95
devised a microfluidic system, known as digi-
tal droplet with auto-catalytic hairpin assembly (ddaCHA), for the digital
quantification of single microRNAs (miRNA) (Figure 6C). This process
involves partitioning miRNA molecules into individual picoliter-sized droplets,
generating a digital readout in which the target molecule is classified as either
present ("positive") or absent ("negative"). The researchers successfully
implemented an enzyme-free auto-catalytic hairpin assembly (aCHA) within
the droplets, demonstrating its effectiveness in minimizing the impact of
external environmental factors and temperature fluctuations on droplet
performance. In their investigation, an automated Python program was
employed to monitor and compute the fluorescence intensities of individual
droplets when they are flowing through a high-throughput microfluidic chip.
By assigning a distinct identifier to each droplet within the flow channel, the
script ensured that each droplet was only quantified once, thereby eliminat-
ing the risk of replicated counting. The significance of automation in such
detection systems is obvious as it considerably reduces manual labor and
minimizes potential errors in the process.
However, the complexity of these systems can be daunting for untrained
end users, prompting the development of new approaches that aim to
simplify or automate the microfluidic steps. Wang group96
presented a novel
lab-on-a-particle assay that employs hydrogel particles for simultaneous
immobilization of protein molecules and emulsification templating, enabling
digital counting of β-galactosidase. Following this process, image analysis
algorithms in MATLAB are used to analyze the fluorescence signal of each
droplet by averaging signals over the particle. A signal is considered to be
positive if it exceeds 2.5 × standard deviations above the mean of the back-
ground signals (Figure 6D). Moving forward, the efforts will be paid for refin-
ing this process and measuring actual targets further using appropriate affin-
Medicine
Figure 6. Droplet-based strategies for single-molecule detection (A) Droplet digital ExoELISA for exosome quantification using magnetic beads in microdroplets for fluorescent
readout.93
(copyright Springer Nature, 2018). (B) The µMD's droplet generation and detection for mini dELISA on mobile platforms with antibody-functionalized, color-coded
beads.94
(copyright National Academy of Science, 2019). (C) Droplet-generating CFF junction design for single-molecule miRNA imaging.95
(copyright American Chemical Society,
2022). (D) Production of Hydrogel particles through step emulsification at low pH.96
(copyright Royal Society of Chemistry, 2021).
REVIEW
The Innovation Medicine 1(2): 100023, September 21, 2023 9
ity capture probes.
Droplet-based single-molecule detection has shown great potential in vari-
ous applications, however, it presents certain drawbacks. For instance,
droplet generation and manipulation can be technically challenging, and
droplet stability may be affected by evaporation or coalescence over time.97,98
Additionally, droplet-based approaches may not be suitable for all sample
types or analytes, such as those with high viscosity or complex molecular
structures due to challenges in generating uniform droplets and potential loss
of analyte integrity. Furthermore, recovering analytes from droplets for further
analysis could be difficult. These limitations call for the need of alternative
approaches like microwell-based single particle detection, which may provide
a more robust and versatile solution for many applications.
Confinement within microwell
Microwell strategies serve as an alternative to droplet-based techniques by
confining chromatic or luminescent products within a tiny-volume microw-
ells rather than the water-in-oil droplets. The microwell system is usually
generated by nano-/micro-fabrication. Microwell-based techniques offer a
controlled and stable environment for target analysis, as they do not rely on
droplet generator and the complex manipulation. By employing microwell
arrays, researchers can effectively isolate and analyze individual particles.
Microwell confinement-based detection was originally developed by the Walt
group99
. This detection method, also referred to as Single Molecular Array
(Simoa), has been successfully commercialized. In the Simoa approach, each
femtoliter-sized reaction chamber in the array contains a single magnetic
bead, and the number of microbeads (MBs) is deliberately maintained to
significantly exceed the number of target molecules from highly diluted
sample. In essence, it utilizes the concept of transformation of on-bead
confinement to the micro-well confinement system, the enzymatically
catalyzed fluorescence can be monitored by ultrasensitive CCD camera. The
beads with the sandwich complexes are then loaded into these chambers,
with each chamber designed to accommodate one bead. Subsequently, a
fluorogenic substrate is introduced, which allows the enzyme on the detec-
tion antibody to convert the substrate into a fluorescent product. This
process results in a binary "0" or "1" signal output, corresponding to the
absence or presence of the target analyte, enabling highly sensitive detection.
However, the Simoa system highly relies on the pricey bulk instrument and
micro-well plates, which is not affordable to many remote areas and source-
limited regions. Also, the current format is mainly designed for singleplexed
profiling while multiplexing ability is still limited.
As microwell detection has evolved with continuous improvements and
refinements, researchers have explored its potential in various applications.
For instance, Karen group100
has developed a novel digital microfluidics (DMF)
chip-based method for detecting thyroid-stimulating hormone (TSH), a criti-
cal marker for assessing thyroid function. Such device requires a tiny sample
volume of 1.1 µL and offers a LOD at 1.3 nanoIU/mL (Figure 7A). Microwells
Figure 7. Microwell confinement-based detection platforms (A) Schematic illustration of the digital microfluidics (DMF) chip with microwell arrays.100
(copyright American
Chemical Society, 2022). (B) Schematic illustrations of bead trapping.101
(copyright Elsevier Ltd., 2019). (C) Schematic of the microwell-based biosensor with edge
enhancement.102
(copyright American Chemical Society, 2021). (D) Schematic illustration of the digital microfluidics (DMF) chip with microwell arrays.103
(copyright Elsevier Ltd.,
2021).
REVIEW
10 The Innovation Medicine 1(2): 100023, September 21, 2023 www.the-innovation.org/medicine
were fabricated on a PET substrate using a UV imprinting process and resist
material through a roll-to-roll fabrication method. In the DMF chip, precise
control of electric fields between the top and bottom electrodes enables effi-
cient droplet manipulation within these microwells, and the hydrophobic-in-
hydrophobic microwells on the chip also boost a higher seeding efficiency
(97.6% ± 0.6%) due to minimized surface energy, ensuring stable droplet
confinement, reduced merging or spreading. In another study, the Sun
group101
has developed a microfluidic chip using polydimethylsiloxane
(PDMS) based microwell patterned arrays to detect tumor necrosis factor α
(TNF-α) (Figure 7B). Beads were injected into a chip and were allowed to
settle for several minutes due to gravity. Subsequently, oil was injected
through the inlet to seal an aqueous solution containing a single bead in the
well. This power-free oil-sealed water phase eliminates the need for external
pumping systems or valves, making it a cost-effective approach. Further-
more, the fabrication cost of these chips is low, and there is no need for
complex surface modifications. The method also maintains good sensitivity
with a limit of detection (LOD) of 12.62 fg/mL, which is comparable to perfor-
mance using the commercial Simoa HD-1 Analyzer (14 fg/mL). In order to
amplify optical signals and increase sensitivity, Li group102
used edge-
enhanced microwell (40 μm depth) immunoassay for detection of interferon-
γ (Figure 7C). The microwells are coated with capture antibodies, which facili-
tate the binding of IFN-γ and the following Texas conjugated detection anti-
bodies to form sandwich immune-complex. Due to larger axial resolution
along with the vertical detection, the fluorescence emission was enhaced,
then the signals of the analytes will be amplified greatly, which formed fluo-
rescent rings. The edge enhancement effect of the microwell vertical side-
wall shows a 6-fold sensitivity enhancement compared to those obtained on
a flat surface.
It is worth mentioning that all the examples presented above are focusing
on detecting single target, which may not reflect disease evolution and
progression accurately. Also, lots of diseases originate by complex multiple
factors, thus there is a need for multiplexed assays to permit multi-analyte
profiling. In conventional assays, the detection of multiple biomarkers from
one set of samples is challenging due to the complexity of encoding and
decoding of numerous signal output from different analytes, and various error
sources can lead to a decrease in the accuracy of the assay.54
Therefore,
rational design that allows distinguishable signal readouts, decent sensitivity,
and specificity is the key pathway. Recognizing the existing limitations, Sung
group103
introduced the pre-equilibrium digital ELISA (PEdELISA) microarray
platform that leverages spatial-spectral encoding and machine learning on a
microfluidic chip (Figure 7D). This allows 12-cytokine assay using only 15 μL
of serum. The 12-plex PEdELISA microarray uses a CNN-based parallel
computing algorithm, ensuring unsupervised image data analysis with high
accuracy. It autonomously classifies and segments image features at a high
throughput (1 min/analyte), achieving 8‒10 times higher accuracy than tradi-
tional GTS-based algorithms without manual corrections. The entire detec-
tion and analysis cycle can be completed within 40 min. In essence,
PEdELISA enhances multiplexing efficiency by integrating advanced
microfluidics with a machine learning image processing method.
AI APPROACH FOR OPTICAL DETECTION IN CLINICAL ANALYSIS
Indeed, with the remarkable advancements in hardware technology, there
has been an extraordinary surge in the development and implementation of
artificial intelligence (AI) tools that are specifically designed for disease diag-
nosis. These bespoke AI systems excel at processing vast volumes of data
and images sourced from a myriad of equipment. By incorporating AI models,
such as Linear Discriminant Analysis (LDA)104-106
, Support Vector Machines
(SVM)107,108
, Logistic Regression, Linear Regression, Linear Mixed Models and
Random Forest Models (RFM)109,110
, these systems significantly enhance both
the efficiency and accuracy of diagnostic procedures. Within the scope of our
specialized system, the primary objective is the automated detection and
quantification of visible particle signals as particle count can act as a surro-
gate for biomarker concentration. By accurately quantifying specific protein
biomarkers in patients, these AI-driven approaches can significantly advance
medical research and patient care, potentially leading to earlier interventions
and improved treatment outcomes. The schematic diagram of this AI appli-
cation in our review can be summarized as follows: Image acquisition →
Particle detection and counting → Biomarker concentration representation →
Data Analysis→Diagnosis (Figure 8). This progression succinctly captures
the role of AI in augmenting diagnostic procedures in healthcare.
Automation and standardization of image processing
AI technology has revolutionized optical detection by automating data
processing, boosting efficiency, and reducing human error. In image process-
Medicine
Figure 8. The general process of AI techniques for data interpretation
REVIEW
The Innovation Medicine 1(2): 100023, September 21, 2023 11
ing, AI excels in automating particle counting through several steps:
(1) Image Preprocessing: AI algorithms enhance image quality by decreas-
ing noise and elevating contrast. Techniques like Gaussian filtering, thresh-
olding, and contrast normalization111,112
are used for clarity, facilitating better
detection and accurate particle counting. (2) Object Detection: Depending on
image complexity, methods range from connectivity analysis for simple
images to advanced techniques like edge detection and machine learning for
intricate ones. (3) Object Counting: After segmentation, objects are enumer-
ated using tools like ImageJ/FIJI, MATLAB, and Python libraries such as
OpenCV and Scikit-Image. The Chen group113
demonstrated this, differentiat-
ing polystyrene (PS) microspheres with a CV technique, achieving 93.5% to
100% accuracy, surpassing traditional methods. For example, the Chen
group113
utilized computer vision (CV) methods to differentiate and quantify
polystyrene (PS) microspheres of varying sizes. (Figure 9A) Through tech-
niques like binarization thresholding and a customized algorithm, they
achieved impressive accuracy ranging from 93.5% to 100%, surpassing tradi-
tional manual methods.
To sum up, ultra-sensitive optical detection can be realized through vari-
ous rational signal amplification strategies, translating minuscule signals into
countable particles, bypassing the need for complex AI technologies typically
required in other systems. However, when a scenario necessitates more intri-
cate analysis, such as distinguishing signals based on their intensity or corre-
lating signal strength with specific biomarker concentrations, advanced algo-
rithms and strategies become invaluable. Nevertheless, the incorporation of
AI, even within these simplified environments and small-scale clinical sample
analysis, significantly enhances the accuracy and reliability of our method-
ologies.
AI approaches to data interpretation
Analysis of collected biomarker data plays a crucial role in gaining a
comprehensive understanding and diagnosis of diseases. The abundance
and diversity of biomarkers offer valuable insights for disease diagnosis,
monitoring treatment response, tracking disease progression, and identifying
disease subtypes. With the advent of AI techniques, the analysis of biomarker
data has become more efficient and accurate, enabling the delivery of
personalized healthcare services to patients. In the following section, we will
elaborate how various AI techniques work on these critical medical areas.
In the domain of early disease diagnosis, SVM-based methods are notably
adept at dealing with high-dimensional data, like gene expression profiles.
Their proficiency in extracting complex patterns from this data allows them to
discern distinct gene expression signatures that could separate cancer cells
apart from healthy cells. This distinguishing capability is pivotal in facilitating
the early detection of cancer.
Given the prominence of high-dimensional gene expression data in cancer
research, the application of SVMs might be more prevalent in this field than in
others.114,115
However, their performance depends on the correct selection of a
kernel function and adjustment of parameters, which may require specialized
domain knowledge. Further research in this area could greatly enhance the
applicability of SVMs.
Moving on to the monitoring of treatment responses, Random Forest
models play a central role in this domain.116-119
These models are ensemble
learning methods that combine multiple decision trees. This approach could
provide an understanding of the intricate interactions between various
biomarkers and treatment outcomes. For instance, when managing chronic
diseases like diabetes, Random Forest models can effectively handle diverse
Figure 9. AI-assisted strategies for disease diagnosis (A) Schematic illustration of workflow and principles of CV-enabled image processing for simultaneous detection.113
(copyright American Chemical Society, 2023). (B) Schematic illustration of machine learning-based SERS detection by PCA-LDA.125
(copyright American Chemical Society, 2023).
REVIEW
12 The Innovation Medicine 1(2): 100023, September 21, 2023 www.the-innovation.org/medicine
data, ranging from glucose levels and insulin doses to lifestyle factors like diet
and physical activity.120
This capability could help develop a more personal-
ized and effective treatment strategies. However, the complexity of the model
can increase as the number of features or variables grows, making it chal-
lenging to interpret the results and understand the underlying patterns effec-
tively. Therefore, future research may explore more effective ways to opti-
mize the number of decision trees in the ensemble or propose new data
dimensionality reduction techniques.
Linear Discriminant Analysis (LDA) plays an important role in tracking
disease progression.121-124
Its capability to simplify high-dimensional
biomarker data makes it instrumental in identifying longitudinal changes in
biomarker levels, paving the way for a deeper understanding of disease
trajectories over time. For instance, when monitoring heart disease progres-
sion, LDA can be applied to unravel dynamic alterations in multiple biomark-
ers, such as cholesterol levels, blood pressure readings, and inflammation
markers. This analytical power aids in understanding the fluid nature of
disease progression and enabling timely interventions. For example, Jin
group 125
employed a combined approach of principal component analysis
(PCA) and Linear Discriminant Analysis (LDA) to extract features from high-
dimensional Surface-Enhanced Raman Scattering (SERS) spectra (Figure
9B). First, they performed dimensionality reduction on the SERS spectra using
PCA, reducing them to 46 principal components that accounted for 95% of
the total variance. Next, they applied LDA to further reduce the dimensionality
of these 46 principal components to just two LDA factors. Remarkably, this
combined approach resulted in a clear separation and accurate identification
of exosomes derived from normal cells and cancerous cells.
In the field of disease subtyping, linear and logistic regression models serve
as potent tools for grouping.126,127
These models can shed light on the associ-
ations between biomarkers and specific disease subtypes. In addition, these
models can integrate an array of biomarkers, such as gene expression
profiles and metabolic markers, to estimate disease subtypes, enabling more
effective and personalized management plans.
In conclusion, AI techniques have demonstrated their effectiveness in
analyzing biomarker data, facilitating a deeper understanding and diagnosis
of diseases. These techniques contribute to early disease detection, treat-
ment response monitoring, disease subtyping, ultimately providing more
accurate and tailored healthcare services for patients. However, it is also
essential to be aware of potential challenges. AI models heavily rely on the
quality of the training data. Therefore, ensuring the accuracy, completeness,
and standardization of biomarker data is critical to yield reliable results. In
addition, these models often involve complex mathematical computations
and require domain knowledge for optimal parameter tuning, highlighting the
need for interdisciplinary collaborations between clinicians, data scientists,
and bioinformaticians. Another potential challenge in embracing AI advance-
ments is overfitting due to sample size. Overfitting occurs when a model is
excessively complex and learns noise or specific patterns from the training
data that do not generalize well to real data.
Another area that warrants attention is the interpretability of these models.
While AI models can make highly accurate predictions, the underlying
reasoning can be opaque, a phenomenon often termed as the "black box"
problem. This lack of transparency can hinder physicians' trust in the model
predictions, thereby limiting their adoption in clinical practice. Lastly, ethical
and legal considerations surrounding data privacy and security, and algorith-
mic bias should also be considered when deploying AI in healthcare. As we
move towards this future, it is imperative to address these challenges head-
on to fully harness the potential of AI in transforming patient care and
outcomes. The amalgamation of AI with medical science is truly an exciting
frontier. It promises not only advancements in disease understanding and
management but also a new era of personalized medicine. With careful navi-
gation, we stand on the cusp of a healthcare revolution.
CONCLUSION AND PERSPECTIVE
This review provides a comprehensive view of confinement-based ultra-
sensitive optical assays (Table 1), enabling the (digital) measurement of
analytes without relying on advanced imaging systems such as total internal
reflection fluorescence (TIRF)43,44
microscopy or photo-activated localization
microscopy (PALM).128,129
As the investigation of novel and uncommon
disease markers expands, along with the need to decipher multifactorial
diseases, there is a rising demand for the development of unsophisticated,
ultrasensitive, and multiplexed detection platforms to address these complex
challenges. Luminescent systems, either through self-illuminating
compounds or through the products of enzymatically induced luminescence
or color systems, can be confined to internal or external spaces. These
systems can cooperate with numerous signal amplification approaches, such
as incorporating surface plasmon enhancement and nucleic acid amplifica-
tion, to realize ultra-bright and ultra-strong signal readouts. The core aim is to
harmonize the cooperation of signal amplification with noise reduction,
reaching a balance that is crucial to achieving a distinguishable signal from
the background at a single-entity level. This balance plays a key role in
enabling early diagnosis and treatment. However, this cutting-edge detection
approach still faces significant obstacles and challenges, highlighting the
need for further investigation and resolution to fully exploit its potential in
diagnostic and therapeutic applications.
Ultrasensitive optical detection requires specific probes to avoid false
results. Enhancing probe specificity can be achieved through unique molecu-
lar design, directed evolution, high-throughput screening, multi-domain
probes, and computational modeling. These steps can substantially improve
the accuracy and reliability in applications like disease diagnosis. Various
signal amplification strategies aim for ultrasensitivity, with the reduction of
background noise being paramount. Incorporating anti-fouling techniques,
such as PEGylation and zwitterionic coatings, can minimize contaminants,
enhancing detection accuracy.130
With the rise of automation and AI, their synergy with ultrasensitive detec-
tion is gaining traction. As the detection process requires sample purification
to eliminate interference, automation offers improved efficiency and accu-
racy over traditional methods. AI is particularly adept at rapidly interpreting
images obtained through microscopy. Moreover, training AI on vast patient
datasets allows for predicting disease trajectories and crafting tailored treat-
ment plans. By fusing AI with particle-guided optical assays, we pave the way
for innovative disease diagnostics and treatments. While many of these tech-
nologies remain in their nascent stages, leveraging larger sample sizes in the
future could lead to significant breakthroughs. This evolution symbolizes the
shift from mere big data to its profound mining, optimizing the union of AI
and detection hardware.131
The intersection of biotechnology (BT) and information technology (IT) is
undeniably shaping our present and future. To achieve impactful outcomes,
strategies in confinement, signal amplification, and encoding must seam-
lessly integrate. The overarching goal is to craft miniaturized, automated, and
smart systems for monitoring evolving disease threats with validated mark-
ers, a venture that underscores the importance of interdisciplinary teamwork.
The next decade promises transformative advancements, especially in single-
entity detection technology.132
With potential applications spanning molecu-
lar biology, biomedicine, to nanotechnology, it is essential to address any
challenges and champion continued innovation for its broader acceptance.
Such progression is vital for proactive disease management, improving
human health, and realizing novel technological leaps.
REFERENCES
Cheung, A.H.K., Chow, C., and To, K.F. (2018). Latest development of liquid biopsy.
J.Thorac. Dis. 10, S1645-S1651.
1.
Miller, D.B., and O’Callaghan, J.P. (2015). Biomarkers of Parkinson’s disease: present
and future. Metabolism 64, S40-S46.
2.
Counts, S.E., Ikonomovic, M.D., Mercado, N., et al. (2017). Biomarkers for the early
detection and progression of Alzheimer’s disease. Neurotherapeutics 14, 35-53.
3.
Gangi, S., Fletcher, J., Nathan, M.A., et al. (2004). Time interval between
abnormalities seen on CT and the clinical diagnosis of pancreatic cancer:
retrospective review of CT scans obtained before diagnosis. Am. J. Roentgenol. 182,
897-903.
4.
Metintas, M., Ak, G., Dundar, E., et al. (2010). Medical thoracoscopy vs CT scan-
guided Abrams pleural needle biopsy for diagnosis of patients with pleural effusions:
a randomized, controlled trial. Chest 137, 1362-1368.
5.
Helpern, J.A., Jensen, J., Lee, S.P., and Falangola, M.F. (2004). Quantitative MRI
assessment of Alzheimer's disease. J. Mol. Neurosci. 24, 45-48.
6.
Morrow, M., Waters, J., and Morris, E. (2011). MRI for breast cancer screening,
diagnosis, and treatment. Lancet 378, 1804-1811.
7.
Gao, Q., Asthana, A., Tong, T., and Rueckert, D. (2014). Multi-scale feature learning
8.
Medicine
REVIEW
The Innovation Medicine 1(2): 100023, September 21, 2023 13
on pixels and super-pixels for seminal vesicles MRI segmentation. Medical Imaging
2014: Image Processing. Spie, 2014, 9034, 36-41.
Braum, L.S., McGonagle, D., Bruns, A., et al. (2013). Characterisation of hand small
joints arthropathy using high-resolution MRI —limited discrimination between
osteoarthritis and psoriatic arthritis. Eur radiol 23, 1686-1693.
9.
Zarovni, N., Corrado, A., Guazzi, P., et al. (2015). Integrated isolation and quantitative
analysis of exosome shuttled proteins and nucleic acids using immunocapture
approaches. Methods 87, 46-58.
10.
Chi, X., Huang, D., Zhao, Z., et al. (2012). Nanoprobes for in vitro diagnostics of
cancer and infectious diseases. Biomaterials 33, 189-206.
11.
Zhang, Z., and Chan, D. W. (2010).. The Road from discovery to clinical diagnostics:
lessons learned from the first FDA-cleared in vitro diagnostic multivariate index
assay of proteomic biomarkers. Cancer Epidem. Biomar. 19, 2995-2999.
12.
Pritchard, C.C., Cheng, H.H., and Tewari, M. (2012). MicroRNA profiling: approaches
and considerations. Nat. Rev. Genet. 13, 358-369.
13.
St John, A., and Price, C.P. (2014). Existing and emerging technologies for point-of-
care testing. Clin Biochem. Rev. 35, 155-167.
14.
Whitesides, G.M. (2006). The origins and the future of microfluidics. Nature 442, 368-
373.
15.
Han, F., Wang, T., Liu, G., et al. (2022). Materials with tunable optical properties for
wearable epidermal sensing in health monitoring. Adv. Mater. 34, e2109055.
16.
Kaur, J., Jiang, C., and Liu, G. (2019). Different strategies for detection of HbA1c
emphasizing on biosensors and point-of-care analyzers. Biosens. Bioelectron. 123,
85-100.
17.
Liu, G., Jiang, C., Lin, X., and Yang, Y.J.V. (2021). Point‐ of‐ care detection of
cytokines in cytokine storm management and beyond: significance and challenges.
View 2, 20210003.
18.
Fu, Y., Jiang, C., Tofaris, G.K., and Davis, J.J. (2020). Facile impedimetric analysis of
neuronal exosome markers in Parkinson’s disease diagnostics. Anal. Chem. 92,
13647-13651.
19.
Chen, R., Mias, G.I., Li-Pook-Than, J., et al. (2012). Personal omics profiling reveals
dynamic molecular and medical phenotypes. Cell 148, 1293-1307.
20.
Sharafeldin, M., Yan, S., Jiang, C., et al. (2023). Alternating magnetic field-promoted
nanoparticle mixing: the on-chip immunocapture of serum neuronal exosomes for
Parkinson’s disease diagnostics. Anal. Chem. 95, 7906–7913.
21.
Jiang, C., Alam, M.T., Silva, S.M., et al. (2016). Unique sensing interface that allows
the development of an electrochemical immunosensor for the detection of tumor
necrosis factor α in whole blood. ACS Sens. 1, 1432-1438.
22.
Cao, C., Zhang, Y., Jiang, C., et al. (2017). Advances on aryldiazonium salt chemistry
based interfacial fabrication for sensing applications. ACS Appl. Mater. Interfaces 9,
5031-5049.
23.
Vasilieva, A., Yurina, L., Azarova, D.Y., et al. (2022). Development of a diagnostic
approach based on the detection of post-translation modifications of fibrinogen
associated with oxidative stress by the method of high efficiency liquid
chromatography. J Phys Chem B.16, 118-122.
24.
Chiki, A. (2020). Development of novel methods and tools to decipher the huntingtin
post-translation modifications code. EPFL.
25.
Jiang, C., Hopfner, F., Katsikoudi, A., et al. (2020). Serum neuronal exosomes predict
and differentiate Parkinson’s disease from atypical parkinsonism. J Neurol
Neurosurg Psychiatry 91, 720-729.
26.
Mitchell, P.S., Parkin, R.K., Kroh, E.M., et al. (2008). Circulating microRNAs as stable
blood-based markers for cancer detection. PNAS 105, 10513-10518.
27.
Weber, J.A., Baxter, D.H., Zhang, S., et al. (2010). The microRNA spectrum in 12 body
fluids. Clin. Chem. 56, 1733-1741.
28.
Chen, X., Ba, Y., Ma, L., et al. (2008). Characterization of microRNAs in serum: a novel
class of biomarkers for diagnosis of cancer and other diseases. Cell Res. 18, 997-
1006.
29.
Lu, X., Hu, C., Jia, D., et al. (2021). Amplification-free and mix-and-read analysis of
multiplexed microRNAs on a single plasmonic microbead. Nano Lett. 21, 6718-
6724.
30.
Wang, G., Tian, W., Liu, X., et al. (2020). New CRISPR-derived microRNA sensing
mechanism based on Cas12a self-powered and rolling circle transcription-
unleashed real-time crRNA recruiting. Anal. Chem. 92, 6702-6708.
31.
Goetzl, L., Merabova, N., Darbinian, N., et al. (2018). Diagnostic potential of neural
exosome cargo as biomarkers for acute brain injury. Ann. Clin. Transl. Neurol. 5, 4-
10.
32.
Singh, K., Nalabotala, R., Koo, K.M., et al. (2021). Separation of distinct exosome
subpopulations: isolation and characterization approaches and their associated
challenges. Analyst 146, 3731-3749.
33.
Picciolini, S., Gualerzi, A., Vanna, R., et al. (2018). Detection and characterization of
different brain-derived subpopulations of plasma exosomes by surface plasmon
resonance imaging. Anal. Chem. 90, 8873-8880.
34.
Yousif, G., Qadri, S., Haik, M., et al. (2021). Circulating exosomes of neuronal origin
as potential early biomarkers for development of stroke. Mol Diagn Ther 25, 163-
180.
35.
Yan, S., Jiang, C., Davis, J.J., and Tofaris, G.K. (2023). Methodological considerations
in neuronal extracellular vesicle isolation for α-synuclein biomarkers.Brain,
awad169.
36.
Kang, Y.T., Hadlock, T., Lo, T.W., et al. (2020). Dual‐ isolation and profiling of
circulating tumor cells and cancer exosomes from blood samples with melanoma
using immunoaffinity‐based microfluidic interfaces. Adv. Sci. 7, 2001581.
37.
Słomka, A., Wang, B., Mocan, T., et al. (2022). Extracellular vesicles and circulating
tumour cells-complementary liquid biopsies or standalone concepts? Theranostics
12, 5836.
38.
Schwarzenbach, H., Hoon, D.S., and Pantel, K.J. (2011). Cell-free nucleic acids as
biomarkers in cancer patients. Nat. Rev. Cancer 11, 426-437.
39.
Jahr, S., Hentze, H., Englisch, S., et al. (2001). DNA fragments in the blood plasma of
cancer patients: quantitations and evidence for their origin from apoptotic and
necrotic cells. Cancer Res. 61, 1659-1665.
40.
Zhang, H.Y., Wang, S., and Fang, G.Z. (2011). Applications and recent developments
of multi-analyte simultaneous analysis by enzyme-linked immunosorbent assays. J.
Immunol. Methods 368, 1-23.
41.
Lequin, R.M. (2005). Enzyme immunoassay (EIA)/enzyme-linked immunosorbent
assay (ELISA). Clin. Chem. 51, 2415-2418.
42.
Steyer, J.A., and Almers, W.J.N.r.M.c.b. (2001). A real-time view of life within 100 nm
of the plasma membrane. Nat. Rev. Mol. Cell Biol. 2, 268-275.
43.
Axelrod, D. (2001). Total internal reflection fluorescence microscopy in cell biology.
Traffic 2, 764-774.
44.
Hartmann, F.J., and Bendall, S.C. (2020). Immune monitoring using mass cytometry
and related high-dimensional imaging approaches. Nat. Rev. Rheumatol. 16, 87-99.
45.
Robinson, J.P., Rajwa, B., Patsekin, V., and Davisson, V.J. (2012). Computational
analysis of high-throughput flow cytometry data. Expert Opin Drug Discov 7, 679-
693.
46.
Lu, Y., Cheng, H., Li, G.C., et al. (2022). Dynamic cryptography through plasmon‐
enhanced fluorescence blinking. Adv. Funct. Mater. 32, 2201372.
47.
Zheng, Y., Jiang, C., Ng, S.H., et al. (2016). Unclonable plasmonic security labels
achieved by shadow‐mask‐lithography‐assisted self‐assembly. Adv. Mater.
28, 2330-2336.
48.
Su, Q., Jiang, C., Gou, D., and Long, Y. (2021). Surface plasmon-assisted
fluorescence enhancing and quenching: from theory to application. ACS Appl. Bio
Mater. 4, 4684-4705.
49.
Zong, H., Wang, X., Mu, X., et al. (2019). Plasmon-enhanced fluorescence resonance
energy transfer. Chem. Rec. 19, 818-842.
50.
Min, X., Cao, B., Huang, S., et al. (2023). Bioorthogonal chemistry-based high-
efficient quantum dots binding boosts the detection sensitivity of plasmon-
enhanced fluorescence platform for immunoassay. Sens. Actuators, B 382, 133516.
51.
Luan, J., Seth, A., Gupta, R., et al. (2020). Ultrabright fluorescent nanoscale labels for
the femtomolar detection of analytes with standard bioassays. Nat. Biomed. Eng. 4,
518-530.
52.
Fan, Z., Jiang, C., Wang, Y., et al. (2022). Engineered extracellular vesicles as
intelligent nanosystem for next-generation of nanomedicine. Nanoscale Horiz. 7,
682-714.
53.
Jiang, C., Fu, Y., Liu, G., et al. (2022). Multiplexed profiling of extracellular vesicles for
biomarker development. Nano-Micro Lett. 14, 3.
54.
Jiang, C., Hopfner, F., Berg, D., et al. (2021). Validation of α‐synuclein in L1CAM‐
immunocaptured exosomes as a biomarker for the stratification of Parkinsonian
syndromes. Movement. Disord. 36, 2663-2669.
55.
Fan, W., Ren, W., and Liu, C. (2023). Advances in optical counting and imaging of
micro/nano single-entity reactors for biomolecular analysis. Anal. Bioanal. Chem.
415, 97-117.
56.
Zheng, Y., Soeriyadi, A.H., Rosa, L., et al. (2015). Reversible gating of smart
plasmonic molecular traps using thermoresponsive polymers for single-molecule
detection. Nat. Commun. 6, 8797.
57.
Zheng, Y., Rosa, L., Thai, T., et al. (2015). Asymmetric gold nanodimer arrays:
electrostatic self-assembly and SERS activity. J. Mater. Chem. A 3, 240-249.
58.
Lu, Y., Chen, H., Cheng, H., et al. (2022). Plasmonic physical unclonable function
labels based on tricolored silver nanoparticles: implications for anticounterfeiting
applications. ACS Appl. Nano Mater. 5, 9298-9305.
59.
Lu, Y., Cheng, H., Francis, P.S., and Zheng, Y. (2023). Nanomaterials and artificial
intelligence in anti-counterfeiting. Intelligent Nanotechnology, (Elsevier), 361-398.
60.
Zhao, J., Liu, C., Li, Y., et al. (2020). Thermophoretic detection of exosomal
microRNAs by nanoflares. J. Am. Chem. Soc. 142, 4996-5001.
61.
Wu, X., Zhao, H., Natalia, A., et al. (2020). Exosome-templated nanoplasmonics for
multiparametric molecular profiling. Sci. Adv. 6, eaba2556.
62.
Lu, Y., Cheng, H., Li, G.C., et al. (2022). Dynamic cryptography through plasmon‐
enhanced fluorescence blinking. Adv. Funct. Mater. 32, 2201372,
63.
Lu, Y., Chen, H., Cheng, H., et al. (2022). Plasmonic physical unclonable function
labels based on tricolored silver nanoparticles: implications for anticounterfeiting
applications. Adv. Funct. Mater. 5, 9298-9305.
64.
Im, H., Shao, H., Park, Y.I., et al. (2014). Label-free detection and molecular profiling
of exosomes with a nano-plasmonic sensor. Nat. Biotechnol. 32, 490-495.
65.
Liang, K., Liu, F., Fan, J., et al. (2017). Nanoplasmonic quantification of tumour-
derived extracellular vesicles in plasma microsamples for diagnosis and treatment
monitoring. Nat. Biomed. Eng. 1, 0021.
66.
Sriram, M., Markhali, B. P., Nicovich, P. R., et al. (2018) A rapid readout for many
single plasmonic nanoparticles using dark-field microscopy and digital color
67.
REVIEW
14 The Innovation Medicine 1(2): 100023, September 21, 2023 www.the-innovation.org/medicine
analysis. Biosens. Bioelectron. 117, 530-536.
Zhang, Y., Shuai, Z., Zhou, H., et al. (2018). Single-molecule analysis of microRNA
and logic operations using a smart plasmonic Nanobiosensor. J. Am. Chem. Soc.
140, 3988-3993.
68.
Lee, K., Fraser, K., Ghaddar, B., et al. (2018). Multiplexed profiling of single
extracellular vesicles. ACS Nano 12, 494-503.
69.
Craw, P., and Balachandran, W. (2012). Isothermal nucleic acid amplification
technologies for point-of-care diagnostics: a critical review. Lab Chip 12, 2469-
2486.
70.
Zhao, Y., Xiang, J., Cheng, H., et al. (2021). Flexible photoelectrochemical biosensor
for ultrasensitive microRNA detection based on concatenated multiplex signal
amplification. Biosens. Bioelectron. 194, 113581.
71.
Fu, R., and Xianyu, Y. (2023). Gold nanomaterials‐ implemented CRISPR‐ Cas
systems for biosensing. Small 2300057.
72.
Long, Y., Zhou, X., and Xing, D. (2011). Sensitive and isothermal
electrochemiluminescence gene-sensing of Listeria monocytogenes with
hyperbranching rolling circle amplification technology. Biosens. Bioelectron. 26,
2897-2904.
73.
Long, Y., Zhou, X., and Xing, D. (2013). An isothermal and sensitive nucleic acids
assay by target sequence recycled rolling circle amplification. Biosens. Bioelectron.
46, 102-107.
74.
Wu, C., Garden, P.M., and Walt, D.R. (2020). Ultrasensitive detection of attomolar
protein concentrations by dropcast single molecule assays. J. Am. Chem. Soc. 142,
12314-12323.
75.
Wang, W., Wu, J., Zhao, Z., et al. (2022). Ultrasensitive automatic detection of small
molecules by membrane imaging of single molecule assays. ACS Appl. Mater.
Interfaces 14, 54914-54923.
76.
Jiang, C., Alam, M.T., Silva, S.M., et al. (2016). Unique sensing interface that allows
the development of an electrochemical immunosensor for the detection of tumor
necrosis factor α in whole blood. ACS Sens. 1, 1432-1438.
77.
Jiang, C., Wang, G., Hein, R., et al. (2020). Antifouling strategies for selective in vitro
and in vivo sensing. Chem. Rev. 120, 3852-3889.
78.
Sun, J., Wang, G., Cheng, H., et al. (2022). An antifouling electrochemical aptasensor
based on hyaluronic acid functionalized polydopamine for thrombin detection in
human serum. Bioelectrochemistry 145, 108073.
79.
Yan, H., Wen, Y., Tian, Z., et al. (2023). A one-pot isothermal Cas12-based assay for
the sensitive detection of microRNAs. Nat. Biomed. Eng., 1-19.
80.
Zhu, Z., Guo, Y., Wang, C., et al. (2023). An ultra-sensitive one-pot RNA-templated
DNA ligation rolling circle amplification-assisted CRISPR/Cas12a detector assay for
rapid detection of SARS-CoV-2. Biosens. Bioelectron. 228, 115179.
81.
Wu, J., Lin, Z., Zou, Z., et al. (2022). Identifying the phenotypes of tumor-derived
extracellular vesicles using size-coded affinity microbeads. J. Am. Chem. Soc. 144,
23483-23491.
82.
Akama, K., Iwanaga, N., Yamawaki, K., et al. (2019). Wash- and amplification-free
digital immunoassay based on single-particle motion analysis. ACS Nano 13, 13116-
13126.
83.
Zhou, Y., Zhao, W., Feng, Y., et al. (2023). Artificial intelligence-assisted digital
immunoassay based on a programmable-particle-decoding technique for
multitarget ultrasensitive detection. Anal. Chem. 95, 1589-1598.
84.
Gong, F., Yang, Y., Shan, X., et al. (2023). A microchamber-free and enzyme-free
digital assay based on ultrabright fluorescent microspheres. Sens. Actuators, B 380,
133358.
85.
Zhang, J., Li, Y., Chai, F., et al. (2022). Ultrasensitive point-of-care biochemical
sensor based on metal-AIEgen frameworks. Sci. Adv. 8, eabo1874.
86.
Wang, X., and Yan, X.P. (2018). Analyte-driven self-assembly of graphene oxide
sheets onto hydroxycamptothecin-functionalized upconversion nanoparticles for
the determination of type I topoisomerases in cell extracts. Anal. Bioanal. Chem.
410, 6761-6769.
87.
Li, Y.F., Lin, Z.Z., Hong, C.Y., and Huang, Z.Y. (2021). Histamine detection in fish
samples based on indirect competitive ELISA method using iron-cobalt co-doped
carbon dots labeled histamine antibody. Food Chem. 345, 128812.
88.
Yang, H., Liu, Y., Guo, Z., et al. (2019). Hydrophobic carbon dots with blue dispersed
emission and red aggregation-induced emission. Nat. Commun. 10, 1789.
89.
Sun, C., Liu, L., Pérez, L., et al. (2022). Droplet-microfluidics-assisted sequencing of
HIV proviruses and their integration sites in cells from people on antiretroviral
therapy. Nat. Biomed. Eng. 6, 1004-1012.
90.
Tian, T., Shu, B., Jiang, Y., et al. (2021). An ultralocalized Cas13a assay enables
universal and nucleic acid amplification-free single-molecule RNA diagnostics. ACS
Nano 15, 1167-1178.
91.
Mou, L., Hong, H., Xu, X., et al. (2021). Digital hybridization human papillomavirus
assay with attomolar sensitivity without amplification. ACS Nano 15, 13077-13084.
92.
Liu, C., Xu, X., Li, B., et al. (2018). Single-exosome-counting immunoassays for
cancer diagnostics. Nano Lett. 18, 4226-4232.
93.
Yelleswarapu, V., Buser, J.R., Haber, M., et al. (2019). Mobile platform for rapid
sub–picogram-per-milliliter, multiplexed, digital droplet detection of proteins. PNAS
116, 4489-4495.
94.
Chen, Z.P., Yang, P., Yang, Z.Z., et al. (2022). One-step digital droplet auto-catalytic
nucleic acid amplification with high-throughput fluorescence imaging and droplet
95.
tracking computation. Anal. Chem. 94, 9166-9175.
Wang, Y., Shah, V., Lu, A., et al. (2021). Counting of enzymatically amplified affinity
reactions in hydrogel particle-templated drops. Lab Chip 21.
96.
Song, H., Chen, D.L., and Ismagilov, R.F. (2006). Reactions in droplets in microfluidic
channels. Angew Chem Int Ed Engl 45, 7336-7356.
97.
Garstecki, P., Fuerstman, M.J., Stone, H.A., et al. (2006). Formation of droplets and
bubbles in a microfluidic T-junction —scaling and mechanism of break-up. Lab
Chip 6, 437-446.
98.
Rissin, D.M., Kan, C.W., Campbell, T.G., et al. (2010). Single-molecule enzyme-linked
immunosorbent assay detects serum proteins at subfemtomolar concentrations.
Nat Biotechnol 28, 595-599.
99.
Leirs, K., Dal Dosso, F., Perez-Ruiz, E., et al. (2022). Bridging the gap between digital
assays and point-of-care testing: automated, low cost, and ultrasensitive detection
of thyroid stimulating hormone. Anal Chem 94, 8919-8927.
100.
Sun, J.J., Hu, J.M., Gou, T., et al. (2019). Power-free polydimethylsiloxane femtoliter-
sized arrays for bead-based digital immunoassays. Biosens. Bioelectron. 139,
111339.
101.
Li, Q., Bencherif, S.A., and Su, M. (2021). Edge-enhanced microwell immunoassay for
highly sensitive protein detection. Anal. Chem. 93, 10292-10300.
102.
Song, Y., Zhao, J., Cai, T., et al. (2021). Machine learning-based cytokine microarray
digital immunoassay analysis. Biosens. Bioelectron. 180, 113088.
103.
Jeng, M.-J., Sharma, M., Sharma, L., et al. (2019). Raman spectroscopy analysis for
optical diagnosis of oral cancer detection. J. Clin. Med. 8, 1313,
104.
Alkhuder, K. (2023). Raman scattering-based optical sensing of chronic liver
diseases. Photodiagn. Photodyn. Ther. 42, 103505.
105.
Lü, G., Zheng, X., Lü, X., et al. (2021). Label-free detection of echinococcosis and liver
cirrhosis based on serum Raman spectroscopy combined with multivariate
analysis. Photodiagn. Photodyn. Ther. 33, 102164.
106.
Wang, C., Zhang, T., Wang, P., et al. (2021). Bone metabolic biomarker-based
diagnosis of type 2 diabetes osteoporosis by support vector machine. Ann. Transl.
Med. 9.
107.
Eke, C.S., Jammeh, E., Li, X., et al. (2020). Early detection of Alzheimer's disease with
blood plasma proteins using support vector machines. IEEE J. Biomed. Health. Inf.
25, 218-226.
108.
Javeed, A., Zhou, S., Yongjian, L., et al. (2019). An intelligent learning system based
on random search algorithm and optimized random forest model for improved heart
disease detection. IEEE Access 7, 180235-180243.
109.
Wang, S., Wang, Y., Wang, D., et al. (2020). An improved random forest-based rule
extraction method for breast cancer diagnosis. Appl. Soft Comput. 86, 105941.
110.
Kumar, A., and Sodhi, S.S. (2020). Comparative analysis of gaussian filter, median
filter and denoise autoenocoder. 2020 7th International Conference on Computing
for Sustainable Global Development (INDIACom). IEEE, 45-51.
111.
Noor, A., Zhao, Y., Khan, R., et al. (2020). Median filters combined with denoising
convolutional neural network for Gaussian and impulse noises. Multimed. Tools.
Appl. 79, 18553-18568.
112.
Feng, N., Wang, S., Wei, L., et al. (2023). Artificial intelligence-based imaging
transcoding system for multiplex screening of viable foodborne pathogens. Anal.
Chem. 95, 8649-8659.
113.
Zhang, F., Kaufman, H.L., Deng, Y., and Drabier, R. (2013). Recursive SVM biomarker
selection for early detection of breast cancer in peripheral blood. BMC Med.
Genomics 6, 1-10,
114.
Zheng, R., Su, R., Xing, F., et al. (2022). Metabolic-dysregulation-based iEESI-MS
reveals potential biomarkers associated with early-stage and progressive colorectal
cancer. Anal. Chem. 94, 11821-11830.
115.
Li, Y., Pan, J., Zhou, N., et al. (2021). A random forest model predicts responses to
infliximab in Crohn’s disease based on clinical and serological parameters. Scand. J.
Gastroenterol 56, 1030-1039.
116.
Wager, S., and Athey, S. (2018). Estimation and inference of heterogeneous
treatment effects using random forests. J. Am. Stat. Assoc. 113, 1228-1242.
117.
Tabib, S., and Larocque, D. (2020). Non-parametric individual treatment effect
estimation for survival data with random forests. Bioinform. 36, 629-636.
118.
Waljee, A.K., Joyce, J.C., Wang, S., et al. (2010). Algorithms outperform metabolite
tests in predicting response of patients with inflammatory bowel disease to
thiopurines. CGH 8, 143-150.
119.
Ellahham, S. (2020). Artificial intelligence: the future for diabetes care. AM. J. Med.
133, 895-900.
120.
Blanco, F.C., Bigi, F., and Soria, M.A. (2014). Identification of potential biomarkers of
disease progression in bovine tuberculosis. Vet. Immunol. Immunopathol. 160, 177-
183.
121.
Rizk-Jackson, A., Stoffers, D., Sheldon, S., et al. (2011). Evaluating imaging
biomarkers for neurodegeneration in pre-symptomatic Huntington's disease using
machine learning techniques. NeuroImage 56, 788-796.
122.
Lin, C.-H., Chiu, S.-I., Chen, T.-F., et al. (2020). Classifications of neurodegenerative
disorders using a multiplex blood biomarkers-based machine learning model. Int. J.
Mol. Sci. 21, 6914.
123.
Ghiti Moghadam, M., Lamers-Karnebeek, F.B., Vonkeman, H.E., et al. (2018). Multi-
biomarker disease activity score as a predictor of disease relapse in patients with
rheumatoid arthritis stopping TNF inhibitor treatment. PLoS One 13, e0192425.
124.
Medicine
REVIEW
The Innovation Medicine 1(2): 100023, September 21, 2023 15
Diao, X., Li, X., Hou, S., et al. (2023). Machine learning-based label-free SERS profiling
of exosomes for accurate fuzzy diagnosis of cancer and dynamic monitoring of
drug therapeutic processes. Anal. Chem. 19, 7552-7559.
125.
Guinney, J., Dienstmann, R., Wang, X., et al. (2015). The consensus molecular
subtypes of colorectal cancer. Nat. Med. 21, 1350-1356.
126.
Menyhárt, O., and Győrffy, B. (2021). Multi-omics approaches in cancer research
with applications in tumor subtyping, prognosis, and diagnosis. Comput. Struct.
Biotechnol. J. 19, 949-960.
127.
Khater, I.M., Nabi, I.R., and Hamarneh, G. (2020). A review of super-resolution single-
molecule localization microscopy cluster analysis and quantification methods.
Patterns 1, 100038.
128.
Scalisi, S., Pisignano, D., Cella Zanacchi, F. (2023). Single‐ molecule localization
microscopy goes quantitative. Microsc Res Tech., 86(4), 494-504.
129.
Mahmoudpour M, Jouyban A, Soleymani J, et al. (2022) Rational design of smart
nano-platforms based on antifouling-nanomaterials toward multifunctional
bioanalysis. Adv. Colloid Interface Sci. 302, 102637.
130.
Dou, B., Zhu, Z., Merkurjev, E., et al. (2023). Machine learning methods for small data
challenges in molecular science. Chem. Rev. 123, 8736-8780.
131.
Fan, W., Dong, Y., Ren, W., and Liu, C. (2023). Single microentity analysis-based
ultrasensitive bioassays: Recent advances, applications, and perspectives. TrAC,
Trends Anal. Chem. 162, 117035.
132.
Liang, K., Liu, F., Fan, J., et al. (2017). Nanoplasmonic quantification of tumour-
derived extracellular vesicles in plasma microsamples for diagnosis and treatment
monitoring. Nat. Biomed. Eng. 1, 0021.
133.
Wang, F., Li, Y., Han, Y., et al. (2019). Single-particle enzyme activity assay with
spectral-resolved dark-field optical microscopy. Anal. Chem. 91, 6329-6339.
134.
Min, X., Cao, B., Huang, S., et al. (2023). Bioorthogonal chemistry-based high-
efficient quantum dots binding boosts the detection sensitivity of plasmon-
enhanced fluorescence platform for immunoassay. Sens. Actuators B: Chem. 382,
133516.
135.
Cohen, L., Cui, N., Cai, Y., et al. (2020). Single molecule protein detection with
attomolar sensitivity using droplet digital enzyme-linked immunosorbent assay.
ACS Nano 14, 9491-9501.
136.
Schmidt, K., Hageneder, S., Lechner, B., et al. (2022). Rolling circle amplification
tailored for plasmonic biosensors: from ensemble to single-molecule detection. ACS
Appl. Mater. Interfaces 14, 55017-55027.
137.
Chen, D., Zhang, X., Zhu, L., et al. (2022). All on size-coded single bead set: a modular
enrich-amplify-amplify strategy for attomolar level multi-immunoassay. Chem. Sci.
13, 3501-3506.
138.
You, M., Peng, P., Xue, Z., et al. (2021). A fast and ultrasensitive ELISA based on
rolling circle amplification. Analyst 146, 2871-2877.
139.
Cui, J.Q., Liu, F.X., Park, H., et al. (2022). Droplet digital recombinase polymerase
amplification (ddRPA) reaction unlocking via picoinjection. Biosens. Bioelectron. 202,
114019.
140.
Xu, S., Wu, J., Chen, C., et al. (2021). A micro-chamber free digital biodetection
method via the “sphere-labeled-sphere” strategy. Sens. Actuators B: Chem. 337,
129794.
141.
Wang, F., Han, Y., Wang, S., et al. (2019). Single-particle LRET aptasensor for the
sensitive detection of aflatoxin B(1) with upconversion nanoparticles. Anal. Chem.
91, 11856-11863.
142.
Farka, Z., Mickert, M.J., Hlavacek, A., et al. (2017). Single molecule upconversion-
linked immunosorbent assay with extended dynamic range for the sensitive
detection of diagnostic biomarkers. Anal. Chem. 89, 11825-11830.
143.
Guan, Z., Zou, Y., Zhang, M., et al. (2014). A highly parallel microfluidic droplet
method enabling single-molecule counting for digital enzyme detection.
Biomicrofluidics 8, 014110.
144.
Zhou, W. (2018). Development of immunomagnetic droplet-based digital immuno-
PCR for the quantification of prostate specific antigen. Anal. Methods 10, 3690-
3695.
145.
Sun, J., Hu, J., Gou, T., et al. (2019). Power-free polydimethylsiloxane femtoliter-
sized arrays for bead-based digital immunoassays. Biosens. Bioelectron. 139,
111339.
146.
Maley, A.M., Garden, P.M., and Walt, D.R. (2020). Simplified digital enzyme-linked
immunosorbent assay using tyramide signal amplification and fibrin hydrogels. ACS
Sens. 5, 3037-3042.
147.
Hu, J., Gou, T., Wu, W., et al. (2019). Proximity ligation assays for precise
quantification of femtomolar proteins in single cells using self-priming microfluidic
dPCR chip. Anal. Chim. Acta 1076, 118-124.
148.
Morasso, C., Ricciardi, A., Sproviero, D., et al. (2022). Fast quantification of
extracellular vesicles levels in early breast cancer patients by Single Molecule
Detection Array (SiMoA). Breast Cancer Res Treat. 192, 65-74.
149.
ACKNOWLEDGMENTS
The work is supported by the Major Research Plan of the National Natural Science
Foundation of China (Grant No.92259102), Dr Cheng Jiang appreciates the financial
support from the Chinese University of Hong Kong (Shenzhen) startup funding
(K10120220253), Shenzhen-Hong Kong Cooperation Zone for Technology and Innova-
tion (HZQBKCZYB-2020056), Shenzhen Science and Technology Program
(2022A1515110206), Shenzhen International Science and Technology Cooperation
Project (GJHZ20220913144002005).
AUTHOR CONTRIBUTIONS
C.J., K.G. and C.X. supervised and revised the manuscript. W.Z. and Y.L. wrote and
edited the manuscript. All authors contributed to the article and approved the submitted
version.
DECLARATION OF INTERESTS
The authors declare no competing interests.
LEAD CONTACT WEBSITE
https://lhs.cuhk.edu.cn/en/teacher/326
REVIEW
16 The Innovation Medicine 1(2): 100023, September 21, 2023 www.the-innovation.org/medicine

More Related Content

Similar to Confinement-guided+ultrasensitive+optical+assay+with+artificial+intelligence+for+disease+diagnostics.pdf

A decision support system for tuberculosis
A decision support system for tuberculosisA decision support system for tuberculosis
A decision support system for tuberculosisijsc
 
A Decision Support System for Tuberculosis Diagnosability
A Decision Support System for Tuberculosis Diagnosability  A Decision Support System for Tuberculosis Diagnosability
A Decision Support System for Tuberculosis Diagnosability ijsc
 
Novel Molecular techniques.pptx
Novel Molecular techniques.pptxNovel Molecular techniques.pptx
Novel Molecular techniques.pptxIbrahimAboAlasaad
 
Lung Nodule Feature Extraction and Classification using Improved Neural Netwo...
Lung Nodule Feature Extraction and Classification using Improved Neural Netwo...Lung Nodule Feature Extraction and Classification using Improved Neural Netwo...
Lung Nodule Feature Extraction and Classification using Improved Neural Netwo...IRJET Journal
 
deep learning applications in medical image analysis brain tumor
deep learning applications in medical image analysis brain tumordeep learning applications in medical image analysis brain tumor
deep learning applications in medical image analysis brain tumorVenkat Projects
 
How is machine learning significant to computational pathology in the pharmac...
How is machine learning significant to computational pathology in the pharmac...How is machine learning significant to computational pathology in the pharmac...
How is machine learning significant to computational pathology in the pharmac...Pubrica
 
5 Best Companies in Digital Pathology Market, March 2023
5 Best Companies in Digital Pathology Market, March 20235 Best Companies in Digital Pathology Market, March 2023
5 Best Companies in Digital Pathology Market, March 2023insightscare
 
IRJET- Intelligent Prediction of Lung Cancer Via MRI Images using Morphologic...
IRJET- Intelligent Prediction of Lung Cancer Via MRI Images using Morphologic...IRJET- Intelligent Prediction of Lung Cancer Via MRI Images using Morphologic...
IRJET- Intelligent Prediction of Lung Cancer Via MRI Images using Morphologic...IRJET Journal
 
Modern diagnosis
Modern diagnosisModern diagnosis
Modern diagnosisAnmol Gupta
 
Important Aspects of Digital Pathology- A Focus on Whole Slide Imaging/Tissue...
Important Aspects of Digital Pathology- A Focus on Whole Slide Imaging/Tissue...Important Aspects of Digital Pathology- A Focus on Whole Slide Imaging/Tissue...
Important Aspects of Digital Pathology- A Focus on Whole Slide Imaging/Tissue...The Lifesciences Magazine
 
E-book Thesis Sara Carvalho
E-book Thesis  Sara CarvalhoE-book Thesis  Sara Carvalho
E-book Thesis Sara CarvalhoSara Carvalho
 
IRJET- Lung Cancer Detection using Digital Image Processing and Artificia...
IRJET-  	  Lung Cancer Detection using Digital Image Processing and Artificia...IRJET-  	  Lung Cancer Detection using Digital Image Processing and Artificia...
IRJET- Lung Cancer Detection using Digital Image Processing and Artificia...IRJET Journal
 
Application of Biomedical Informatics in Clinical Problem Solving
Application of Biomedical Informatics in Clinical Problem SolvingApplication of Biomedical Informatics in Clinical Problem Solving
Application of Biomedical Informatics in Clinical Problem Solvingimprovemed
 
ICBO 2014, October 8, 2014
ICBO 2014, October 8, 2014ICBO 2014, October 8, 2014
ICBO 2014, October 8, 2014Warren Kibbe
 
A Review of Super Resolution and Tumor Detection Techniques in Medical Imaging
A Review of Super Resolution and Tumor Detection Techniques in Medical ImagingA Review of Super Resolution and Tumor Detection Techniques in Medical Imaging
A Review of Super Resolution and Tumor Detection Techniques in Medical Imagingijtsrd
 

Similar to Confinement-guided+ultrasensitive+optical+assay+with+artificial+intelligence+for+disease+diagnostics.pdf (20)

A decision support system for tuberculosis
A decision support system for tuberculosisA decision support system for tuberculosis
A decision support system for tuberculosis
 
A Decision Support System for Tuberculosis Diagnosability
A Decision Support System for Tuberculosis Diagnosability  A Decision Support System for Tuberculosis Diagnosability
A Decision Support System for Tuberculosis Diagnosability
 
biomedicines-03-00203
biomedicines-03-00203biomedicines-03-00203
biomedicines-03-00203
 
Novel Molecular techniques.pptx
Novel Molecular techniques.pptxNovel Molecular techniques.pptx
Novel Molecular techniques.pptx
 
Lung Nodule Feature Extraction and Classification using Improved Neural Netwo...
Lung Nodule Feature Extraction and Classification using Improved Neural Netwo...Lung Nodule Feature Extraction and Classification using Improved Neural Netwo...
Lung Nodule Feature Extraction and Classification using Improved Neural Netwo...
 
deep learning applications in medical image analysis brain tumor
deep learning applications in medical image analysis brain tumordeep learning applications in medical image analysis brain tumor
deep learning applications in medical image analysis brain tumor
 
How is machine learning significant to computational pathology in the pharmac...
How is machine learning significant to computational pathology in the pharmac...How is machine learning significant to computational pathology in the pharmac...
How is machine learning significant to computational pathology in the pharmac...
 
5 Best Companies in Digital Pathology Market, March 2023
5 Best Companies in Digital Pathology Market, March 20235 Best Companies in Digital Pathology Market, March 2023
5 Best Companies in Digital Pathology Market, March 2023
 
Radiology 2040
Radiology 2040Radiology 2040
Radiology 2040
 
IRJET- Intelligent Prediction of Lung Cancer Via MRI Images using Morphologic...
IRJET- Intelligent Prediction of Lung Cancer Via MRI Images using Morphologic...IRJET- Intelligent Prediction of Lung Cancer Via MRI Images using Morphologic...
IRJET- Intelligent Prediction of Lung Cancer Via MRI Images using Morphologic...
 
Modern diagnosis
Modern diagnosisModern diagnosis
Modern diagnosis
 
s12951-020-00613-6.pdf
s12951-020-00613-6.pdfs12951-020-00613-6.pdf
s12951-020-00613-6.pdf
 
Important Aspects of Digital Pathology- A Focus on Whole Slide Imaging/Tissue...
Important Aspects of Digital Pathology- A Focus on Whole Slide Imaging/Tissue...Important Aspects of Digital Pathology- A Focus on Whole Slide Imaging/Tissue...
Important Aspects of Digital Pathology- A Focus on Whole Slide Imaging/Tissue...
 
Use cases
Use casesUse cases
Use cases
 
E-book Thesis Sara Carvalho
E-book Thesis  Sara CarvalhoE-book Thesis  Sara Carvalho
E-book Thesis Sara Carvalho
 
IRJET- Lung Cancer Detection using Digital Image Processing and Artificia...
IRJET-  	  Lung Cancer Detection using Digital Image Processing and Artificia...IRJET-  	  Lung Cancer Detection using Digital Image Processing and Artificia...
IRJET- Lung Cancer Detection using Digital Image Processing and Artificia...
 
Application of Biomedical Informatics in Clinical Problem Solving
Application of Biomedical Informatics in Clinical Problem SolvingApplication of Biomedical Informatics in Clinical Problem Solving
Application of Biomedical Informatics in Clinical Problem Solving
 
ICBO 2014, October 8, 2014
ICBO 2014, October 8, 2014ICBO 2014, October 8, 2014
ICBO 2014, October 8, 2014
 
Tteh.000542
Tteh.000542Tteh.000542
Tteh.000542
 
A Review of Super Resolution and Tumor Detection Techniques in Medical Imaging
A Review of Super Resolution and Tumor Detection Techniques in Medical ImagingA Review of Super Resolution and Tumor Detection Techniques in Medical Imaging
A Review of Super Resolution and Tumor Detection Techniques in Medical Imaging
 

Recently uploaded

AmericanHighSchoolsprezentacijaoskolama.
AmericanHighSchoolsprezentacijaoskolama.AmericanHighSchoolsprezentacijaoskolama.
AmericanHighSchoolsprezentacijaoskolama.arsicmarija21
 
Framing an Appropriate Research Question 6b9b26d93da94caf993c038d9efcdedb.pdf
Framing an Appropriate Research Question 6b9b26d93da94caf993c038d9efcdedb.pdfFraming an Appropriate Research Question 6b9b26d93da94caf993c038d9efcdedb.pdf
Framing an Appropriate Research Question 6b9b26d93da94caf993c038d9efcdedb.pdfUjwalaBharambe
 
DATA STRUCTURE AND ALGORITHM for beginners
DATA STRUCTURE AND ALGORITHM for beginnersDATA STRUCTURE AND ALGORITHM for beginners
DATA STRUCTURE AND ALGORITHM for beginnersSabitha Banu
 
Pharmacognosy Flower 3. Compositae 2023.pdf
Pharmacognosy Flower 3. Compositae 2023.pdfPharmacognosy Flower 3. Compositae 2023.pdf
Pharmacognosy Flower 3. Compositae 2023.pdfMahmoud M. Sallam
 
Alper Gobel In Media Res Media Component
Alper Gobel In Media Res Media ComponentAlper Gobel In Media Res Media Component
Alper Gobel In Media Res Media ComponentInMediaRes1
 
Procuring digital preservation CAN be quick and painless with our new dynamic...
Procuring digital preservation CAN be quick and painless with our new dynamic...Procuring digital preservation CAN be quick and painless with our new dynamic...
Procuring digital preservation CAN be quick and painless with our new dynamic...Jisc
 
Blooming Together_ Growing a Community Garden Worksheet.docx
Blooming Together_ Growing a Community Garden Worksheet.docxBlooming Together_ Growing a Community Garden Worksheet.docx
Blooming Together_ Growing a Community Garden Worksheet.docxUnboundStockton
 
EPANDING THE CONTENT OF AN OUTLINE using notes.pptx
EPANDING THE CONTENT OF AN OUTLINE using notes.pptxEPANDING THE CONTENT OF AN OUTLINE using notes.pptx
EPANDING THE CONTENT OF AN OUTLINE using notes.pptxRaymartEstabillo3
 
Proudly South Africa powerpoint Thorisha.pptx
Proudly South Africa powerpoint Thorisha.pptxProudly South Africa powerpoint Thorisha.pptx
Proudly South Africa powerpoint Thorisha.pptxthorishapillay1
 
Final demo Grade 9 for demo Plan dessert.pptx
Final demo Grade 9 for demo Plan dessert.pptxFinal demo Grade 9 for demo Plan dessert.pptx
Final demo Grade 9 for demo Plan dessert.pptxAvyJaneVismanos
 
Hierarchy of management that covers different levels of management
Hierarchy of management that covers different levels of managementHierarchy of management that covers different levels of management
Hierarchy of management that covers different levels of managementmkooblal
 
ECONOMIC CONTEXT - LONG FORM TV DRAMA - PPT
ECONOMIC CONTEXT - LONG FORM TV DRAMA - PPTECONOMIC CONTEXT - LONG FORM TV DRAMA - PPT
ECONOMIC CONTEXT - LONG FORM TV DRAMA - PPTiammrhaywood
 
MICROBIOLOGY biochemical test detailed.pptx
MICROBIOLOGY biochemical test detailed.pptxMICROBIOLOGY biochemical test detailed.pptx
MICROBIOLOGY biochemical test detailed.pptxabhijeetpadhi001
 
Introduction to AI in Higher Education_draft.pptx
Introduction to AI in Higher Education_draft.pptxIntroduction to AI in Higher Education_draft.pptx
Introduction to AI in Higher Education_draft.pptxpboyjonauth
 
call girls in Kamla Market (DELHI) 🔝 >༒9953330565🔝 genuine Escort Service 🔝✔️✔️
call girls in Kamla Market (DELHI) 🔝 >༒9953330565🔝 genuine Escort Service 🔝✔️✔️call girls in Kamla Market (DELHI) 🔝 >༒9953330565🔝 genuine Escort Service 🔝✔️✔️
call girls in Kamla Market (DELHI) 🔝 >༒9953330565🔝 genuine Escort Service 🔝✔️✔️9953056974 Low Rate Call Girls In Saket, Delhi NCR
 
Employee wellbeing at the workplace.pptx
Employee wellbeing at the workplace.pptxEmployee wellbeing at the workplace.pptx
Employee wellbeing at the workplace.pptxNirmalaLoungPoorunde1
 
Difference Between Search & Browse Methods in Odoo 17
Difference Between Search & Browse Methods in Odoo 17Difference Between Search & Browse Methods in Odoo 17
Difference Between Search & Browse Methods in Odoo 17Celine George
 

Recently uploaded (20)

AmericanHighSchoolsprezentacijaoskolama.
AmericanHighSchoolsprezentacijaoskolama.AmericanHighSchoolsprezentacijaoskolama.
AmericanHighSchoolsprezentacijaoskolama.
 
Framing an Appropriate Research Question 6b9b26d93da94caf993c038d9efcdedb.pdf
Framing an Appropriate Research Question 6b9b26d93da94caf993c038d9efcdedb.pdfFraming an Appropriate Research Question 6b9b26d93da94caf993c038d9efcdedb.pdf
Framing an Appropriate Research Question 6b9b26d93da94caf993c038d9efcdedb.pdf
 
DATA STRUCTURE AND ALGORITHM for beginners
DATA STRUCTURE AND ALGORITHM for beginnersDATA STRUCTURE AND ALGORITHM for beginners
DATA STRUCTURE AND ALGORITHM for beginners
 
Pharmacognosy Flower 3. Compositae 2023.pdf
Pharmacognosy Flower 3. Compositae 2023.pdfPharmacognosy Flower 3. Compositae 2023.pdf
Pharmacognosy Flower 3. Compositae 2023.pdf
 
Model Call Girl in Tilak Nagar Delhi reach out to us at 🔝9953056974🔝
Model Call Girl in Tilak Nagar Delhi reach out to us at 🔝9953056974🔝Model Call Girl in Tilak Nagar Delhi reach out to us at 🔝9953056974🔝
Model Call Girl in Tilak Nagar Delhi reach out to us at 🔝9953056974🔝
 
Alper Gobel In Media Res Media Component
Alper Gobel In Media Res Media ComponentAlper Gobel In Media Res Media Component
Alper Gobel In Media Res Media Component
 
Procuring digital preservation CAN be quick and painless with our new dynamic...
Procuring digital preservation CAN be quick and painless with our new dynamic...Procuring digital preservation CAN be quick and painless with our new dynamic...
Procuring digital preservation CAN be quick and painless with our new dynamic...
 
Blooming Together_ Growing a Community Garden Worksheet.docx
Blooming Together_ Growing a Community Garden Worksheet.docxBlooming Together_ Growing a Community Garden Worksheet.docx
Blooming Together_ Growing a Community Garden Worksheet.docx
 
EPANDING THE CONTENT OF AN OUTLINE using notes.pptx
EPANDING THE CONTENT OF AN OUTLINE using notes.pptxEPANDING THE CONTENT OF AN OUTLINE using notes.pptx
EPANDING THE CONTENT OF AN OUTLINE using notes.pptx
 
Proudly South Africa powerpoint Thorisha.pptx
Proudly South Africa powerpoint Thorisha.pptxProudly South Africa powerpoint Thorisha.pptx
Proudly South Africa powerpoint Thorisha.pptx
 
Final demo Grade 9 for demo Plan dessert.pptx
Final demo Grade 9 for demo Plan dessert.pptxFinal demo Grade 9 for demo Plan dessert.pptx
Final demo Grade 9 for demo Plan dessert.pptx
 
Hierarchy of management that covers different levels of management
Hierarchy of management that covers different levels of managementHierarchy of management that covers different levels of management
Hierarchy of management that covers different levels of management
 
ECONOMIC CONTEXT - LONG FORM TV DRAMA - PPT
ECONOMIC CONTEXT - LONG FORM TV DRAMA - PPTECONOMIC CONTEXT - LONG FORM TV DRAMA - PPT
ECONOMIC CONTEXT - LONG FORM TV DRAMA - PPT
 
OS-operating systems- ch04 (Threads) ...
OS-operating systems- ch04 (Threads) ...OS-operating systems- ch04 (Threads) ...
OS-operating systems- ch04 (Threads) ...
 
MICROBIOLOGY biochemical test detailed.pptx
MICROBIOLOGY biochemical test detailed.pptxMICROBIOLOGY biochemical test detailed.pptx
MICROBIOLOGY biochemical test detailed.pptx
 
TataKelola dan KamSiber Kecerdasan Buatan v022.pdf
TataKelola dan KamSiber Kecerdasan Buatan v022.pdfTataKelola dan KamSiber Kecerdasan Buatan v022.pdf
TataKelola dan KamSiber Kecerdasan Buatan v022.pdf
 
Introduction to AI in Higher Education_draft.pptx
Introduction to AI in Higher Education_draft.pptxIntroduction to AI in Higher Education_draft.pptx
Introduction to AI in Higher Education_draft.pptx
 
call girls in Kamla Market (DELHI) 🔝 >༒9953330565🔝 genuine Escort Service 🔝✔️✔️
call girls in Kamla Market (DELHI) 🔝 >༒9953330565🔝 genuine Escort Service 🔝✔️✔️call girls in Kamla Market (DELHI) 🔝 >༒9953330565🔝 genuine Escort Service 🔝✔️✔️
call girls in Kamla Market (DELHI) 🔝 >༒9953330565🔝 genuine Escort Service 🔝✔️✔️
 
Employee wellbeing at the workplace.pptx
Employee wellbeing at the workplace.pptxEmployee wellbeing at the workplace.pptx
Employee wellbeing at the workplace.pptx
 
Difference Between Search & Browse Methods in Odoo 17
Difference Between Search & Browse Methods in Odoo 17Difference Between Search & Browse Methods in Odoo 17
Difference Between Search & Browse Methods in Odoo 17
 

Confinement-guided+ultrasensitive+optical+assay+with+artificial+intelligence+for+disease+diagnostics.pdf

  • 1. Confinement-guided ultrasensitive optical assay with artificial intelligence for disease diagnostics Wenjing Zhang,1,6 Yongfeng Lu,1,2,3,6 Chenyi Su,1 Yibo Wang,1 Yong-Fei Wang,1 Bo Zhang,1 Cheng Jiang,1,* Keying Guo,4,5,* and Chuan Xu2,3,* *Correspondence: jiangcheng@cuhk.edu.cn (C.J.); keying.guo@gtiit.edu.cn (K.G.); xuchuan100@163.com (C.X.) Received: May 31, 2023; Accepted: August 31, 2023; Published Online: September 1, 2023; https://doi.org/10.59717/j.xinn-med.2023.100023 © 2023 The Author(s). This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/). GRAPHICAL ABSTRACT PUBLIC SUMMARY ■ Advanced optical assays utilize external and internal confinement strategies for disease biomarker detection. ■ Convergence of artificial intelligence (AI) with optical assays boosts diagnostic accuracy and prediction of disease paths. ■ Embracing biotechnology and information technology permits the revolution of biomarker discovery. Medicine REVIEW The Innovation Medicine 1(2): 100023, September 21, 2023 1
  • 2. Confinement-guided ultrasensitive optical assay with artificial intelligence for disease diagnostics Wenjing Zhang,1,6 Yongfeng Lu,1,2,3,6 Chenyi Su,1 Yibo Wang,1 Yong-Fei Wang,1 Bo Zhang,1 Cheng Jiang,1,* Keying Guo,4,5,* and Chuan Xu2,3,* 1 School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China 2 Department of Oncology & Cancer Institute, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital Chengdu, Sichuan 610072, China 3 Department of Laboratory Medicine and Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610054, China 4 Biotechnology and Food Engineering, Guangdong Technion-Israel Institute of Technology (GTIIT), Shantou 515063, China 5 Faculty of Biotechnology and Food Engineering, Technion-Israel Institute of Technology (IIT), Haifa 3200003, Israel 6 These authors contributed equally *Correspondence: jiangcheng@cuhk.edu.cn (C.J.); keying.guo@gtiit.edu.cn (K.G.); xuchuan100@163.com (C.X.) Received: May 31, 2023; Accepted: August 31, 2023; Published Online: September 1, 2023; https://doi.org/10.59717/j.xinn-med.2023.100023 © 2023 The Author(s). This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/). Citation: Zhang W., Lu Y., Su C., et al., (2023). Confinement-guided ultrasensitive optical assay with artificial intelligence for disease diagnostics. The Innovation Medicine 1(2), 100023. The necessity for ultrasensitive detection is becoming increasingly appar- ent as it plays a pivotal role in disease early diagnostics and health management, particularly when it comes to detecting and monitoring low- abundance biomarkers or precious samples with tiny volumes. In many disease cases, such as cancer, infectious disease, autoimmune disorder, and neurodegenerative disease, low-abundant target biomarkers like circu- lating tumor cells (CTCs), extracellular vesicle (EV) subpopulations, and post-translational modified proteins (PTMs) are commonly existing and can be served as early indicators of disease onset or progression. However, these biomarkers often exist in ultra-low quantities in body fluids, surpass- ing the detection limits of conventional diagnostic tools like enzyme-linked immunosorbent assay (ELISA). This leads to the inability to probe disease evolution at a very early stage from molecular pathology perspective. In such regard, ultrasensitive optical assays have emerged as a solution to overcome these limitations and have witnessed significant progress in recent decades. This review provides a comprehensive overview of the recent advancements in ultrasensitive optical detection for disease diag- nostics, particularly focusing on the conjunction of confinement within micro-/nano-structures and signal amplification to generate distinguish- able optical readouts. The discussion begins with a meticulous evaluation of the advantages and disadvantages of these ultra-sensitive optical assays. Then, the spotlight is turned towards the implementation of artifi- cial intelligence (AI) algorithms. The ability of AI to process large volumes of visible reporter signal and clinical data has proven invaluable in identify- ing unique patterns across multi-center cohort samples. Looking forward, the review underscores future advancements in developing convergent biotechnology (BT) and information technology (IT) toolbox, especially opti- cal biosensors for high-throughput biomarker screening, point-of-care (PoC) testing with appropriate algorithms for their clinical translation are highlighted. INTRODUCTION Biomarkers discovered and validated from biological fluids hold great potential for reflecting the molecular pathology and progression of diseases.1-3 Compared to imaging techniques, e.g., computed tomography (CT),4,5 magnetic resonance imaging (MRI)6,7 which highly rely on well-trained clini- cian to identify nidus of milli-meter to centimeter dimension,8,9 biomarkers based in vitro diagnostics (IVD) can offer a range of advantages in disease diagnosis and monitoring,10-13 including high sensitivity,14 real-time monitor- ing,15-19 facilitation of personalized treatment,20,21 and cost-effectiveness.22,23 However, biomarkers like post-translation modification (PTM) proteins,24-26 microRNA,27-31 extracellular vesicle (EV) subpopulations,19,32-36 circulating tumor cells (CTCs),37,38 and circulating nucleic acids39,40 are usually present in low abundance, and their secretion is rare during the early stage of disease progression, posing significant challenges for the conventional detection methodologies like enzyme-linked immunosorbent assays (ELISA)41,42 due to their limited sensitivity and unachievable stable monitoring. Usually, compro- mised strategies have to be used, such as consumption and conversion of large input volume into a squeezed detection volume to achieve a decent enrichment factor. This not only increases the economic and psychological burden on patients associated with multiple sampling but also constrains the widespread adoption and implementation of biomarkers in clinical applica- tions. In such regard, the development of ultrasensitive toolboxes is essential with high demand. The pressing need for ultrasensitive detection methods in the realm of biomarker analysis has led researchers to explore various innovative strate- gies. Fundamentally, ultrasensitive detection either relies on high-end detec- tion devices or signal amplification strategies. The former category includes advanced instruments such as total internal reflection fluorescence (TIRF) microscopy43,44 and cutting-edge flow cytometry analyzers.45,46 While these state-of-the-art tools provide exceptional sensitivity and accuracy, their widespread accessibility may be hindered by factors such as high costs and the need of specialized expertise in operation and maintenance. The latter, on the other hand, serves as the primary focus of this review, offering a more promising and accessible avenue for exploration and translation. Typically, signal amplification with confinement strategies, e.g., multiple enzymatical catalysis, nucleic acid amplification, and surface plasmon- enhanced fluorescence, involve generating sufficiently robust output signals, generally achieved by confining signal molecules within minuscule spaces. Such confinement can be realized either using semi-enclosed microwells, fully enclosed water-in-oil droplets, or by immobilizing ultra-bright or visible probes on the surfaces of particulate substrates to provide localized tremen- dous reporter concentration and signal readout (Figure 1). These methodolo- gies can enhance signal detection and improve the overall sensitivity of the method, ultimately pushing the detection limits down to even single-entity level. In this review, we primarily focus on the development of ultrasensitive opti- cal sensors, emphasizing the “adhesion” of visible reporters to the outer or internal microenvironment, presenting external- and internal- confinement strategies, respectively. Generally, plasmonic47-52 or non-plasmonic materials can be employed to convert target binding events into digitized signal format (Figure 1). How artificial intelligence facilitates image processing, particle counting, and clinical data handling will be emphasized. We introduce repre- sentative cases, discussing their pivotal features and shortcomings. Finally, we delve into the prospective developments both in AI technologies and in the field of ultrasensitive optical detection. Our vision is that the synergy of these advancements will significantly contribute to reshaping diagnostic approaches in healthcare, offering a more precise, efficient, and reliable anal- ysis. EXTERNAL CONFINEMENT STRATEGY To date, diverse types of optical reporters have been investigated with their confinement, either on the outer space or internal space of a nano-/micro- architecture, to achieve ultra-sensitivity. Therefore, these strategies can be mainly categorized into two domains: internal confinement and external confinement. External confinement, namely restraint of optical reporters (e.g., chemical dyes, plasmonic particles) to the outer surface of solid support (e.g., spheri- cal beads). The binding of optical reporter molecules can usually be executed by various approaches like DNA hybridization, covalent binding, etc. Generally, REVIEW 2 The Innovation Medicine 1(2): 100023, September 21, 2023 www.the-innovation.org/medicine
  • 3. in such system, reporter molecules with high-density would generate local- ized high signal intensity on the individual beads that can “illuminate” the single-bead microenvironment under excitation, resulting in superior sensitiv- ity. Such “ultrabright” beads can be easily detected using conventional micro- scope with low-magnification even mobile phone-based microscope. Differ- ent feasible algorithms can then be employed for beads digital counting, which can be converted to the target analyte concentration using appropriate algorithm. The screened biomarker can be used for toward separation of different groups in a cohort study to permit disease early diagnostics, stratifi- cation, therapy monitoring, etc.53-56 So far, plasmonic- and nonplasmonic- materials have been widely used as optical labels in cooperation with confinement strategy for ultrasensitive assay development, which will be discussed in the following sections. Nanoplasmonics based reporter When considering the direct signal output, plasmonic objects with size larger than ~30 nm can be visualized under dark-field microscopy (DFM) due to their strong scattering capabilities. This arises from the localized surface plasmon resonance (LSPR) effect. When the size of the plasmonic particles exceeds a certain threshold—yet remains smaller than the wave- length of light—there is a significant enhancement in light scattering.48,57-60 On the other hand, plasmon can also enhance fluorescence through surface plasmon interactions by mediating the distance (e.g. adjusting the spacer thickness) between the plasmonic materials and the fluorophore. These interactions amplify the local electromagnetic field. This, in turn, increases the excitation rate and the quantum yield of the fluorophores, resulting in ulti- mately enhanced fluorescence signals.61,62 These intrinsic advantages permit plasmonics-related detection with achievable ultrasensitivity. The most widely explored formats are plasmonic label by its alone and surface plas- mon-enhanced fluorescence. Nanoplasmonics alone. The surface plasmon effect refers to the collec- tive oscillation of electrons at the interface between a metal and a dielectric material, lead- ing to unique optical phenomena.49 Dark-field spectroscopy (DFS) is a typical plasmonic parti- cle spectroscopy technique that employs microscopy in combination with a spectrometer to observe plasmonic particles.63,64 Lee group65 developed a plasmonic exosome assay using transmission surface plasmon resonance with antibodies functionalized nanohole arrays to identify ovarian cancer exosomal surface biomarkers (Figure 2A). They demonstrated improved signal amplification through secondary labeling with nanoprobes such as gold nanostars of variable sizes and dimen- sions for detection of 12 surface markers. The sensitivity enhancement factors were 104 -fold and 102 -fold compared to Western blot and chemiluminescence ELISA, respectively. The plasmonic structure sensing mechanism was confirmed through Three-Dimensional Finite- Difference Time-Domain simulations (3D-FDTD) simulations, showing enhancement with the immune-sandwich format on a glass substrate. By using this technique, exosomal EpCAM and CD24 were stably probed, and were demon- strated the ability to differentiate ovarian cancer patients (n=20) from non-cancer patients (n=10). In another work, Hu and co-workers66 built a nanoplasmon-enhanced scattering (nPES) assay to directly probe the membrane antigens from tumour-derived EVs, in which the detection antibody-conjugated gold nanospheres (50 nm) and nanorods (25×60 nm) were bind to EVs, which were captured by antibodies pre-decorated glass chip to form the “sandwich” format, producing a local plasmon effect that enhances tumour-derived EV detection sensitivity and specificity (Figure 2B). The unique plasmonic color of gold nanoparticles (AuNPs) and gold nanorods (AuNRs) allows the duplexed profiling of EV membrane marker, and their coupling caused red shift and color change can indicate the co-existence of the membrane antigens. They leveraged the Image J to analyze batches of DFM images. The method can automatically selected image areas with a brightness value of 255. After identifying these areas, the software calculated the ratios between the selected areas and the total areas of the images. These ratios were used as a metric for the specific nPES EV signal. In their study, the authors demonstrated that nanoplasmon- enhanced scattering assay could effectively distinguish early-stage pancre- atic cancer from pancreatitis and controls in a cohort of 155 subjects. However, the assay capacity for high-throughput surface marker profiling is limited considering the proximity of surface antigens and the steric hindrance of AuNPs and AuNRs during recognition and binding to the exosomal surface. Gooding group67 developed a core-satellite formation of Au nanoparticles (i.e., 67 nm-AuNP-analyte-enzyme coated 10 nm-AuNP) for the detection of interleukin 6 (IL-6). They utilized a CMOS-equipped digital camera and a dark- field microscope to analyze thousands of dropcasted-gold nanospheres within seconds using Image J (Figure 2C). The research team developed a MATLAB script to transform images from the RGB color space into the HSV (i.e. Hue, Saturation, Value) color space, facilitating the separation of intensity and color variables. Following this conversion, a bandpass filter was applied to remove noisy pixels and enhance the image quality. The next step involved the identification of single nanoparticles based on their distinct morphology and intensity of the point-spread-functions. Each identified nanoparticle was Medicine Figure 1. Schematic illustration of confinement based ultrasensitive optical detection REVIEW The Innovation Medicine 1(2): 100023, September 21, 2023 3
  • 4. then labeled, and its color data (hue) is extracted using an intensity-weighted averaging method. This process entails creating a mask around the pixel with the highest intensity within a 3-pixel radius. By employing this robust process, the research team can independently count nanoparticles of various colors, enabling a comprehensive assessment of the nanoparticle population in the image. Compared to the LoD of ELISA and Western blot (~0.5 ng/mL), this dark-field imaging approach shows a 50-fold decrease in LoD due to the plasmon coupling. This method can circumvent the influence of non-specific adsorption due to the localized surface plasmon resonance (LSPR) sandwich assay, which is tolerant to non-specific binding effects, a major problem in the field of biosensing. Wang group68 designed another plasmonic nanobiosensor based on Au@Ag core-shell structure for detecting microRNA 21 (miR-21) (Figure 2D). After capturing miR-21, an average localized surface plasmon resonance (LSPR) scattering wavelength shift of about 0.4 nm can be obtained. In this system, the “pyramidal” DNA tetrahedral structure was utilized as the recog- nition probe for miR-21 capped on noble-metal nanoparticles (e.g., gold or silver nanoparticles) by employing the shifts of the LSPR scattering spec- trum peak (λmax) as the output signal, permitting real-time detection of miR- 21 with sensitivity at the aM level within a large dynamic range of 1 aM ‒ 1 nM. However, the DNA-microarray platform could only give rise to tiny LSPR scattering spectral wavelength shift (~0.4 nm) on this nanobiosensor, which may not be enough to be captured intuitively by a common instrument. Plasmon coupled with fluorescence. Noble metal-based plasmonic parti- cles can not only provide signal output by themselves, but also have the unique ability to couple with other fluorescent emission luminogens or quan- tum dots to enhance emission properties, permitting dark-filed and fluores- cence dual-channel signal output. The physicochemical properties of plas- monic nanoparticles are specifically designed to facilitate plasmon-enhanced fluorescence (PEF) pathway,49 which can significantly enhance the fluores- cence of nearby fluorophore zone using spacers like SiO2 or polymers with appropriate manipulation for the avoidance of quenching. While conventional fluorescent molecules and optical nanoparticles suffer from low brightness, poor photostability, and photobleaching, plasmon-enhanced fluorescence offers improved performance in terms of higher sensitivity and long-term photo-stability. Quantum dots (QDs) are a promising class of fluorescent probes for biological imaging owing to their unique stokes shift, powerful stability, and relatively long fluorescence lifetime. However, the fluorescence intensity of a single quantum dot is not sufficient for observation under a conventional optical microscope. Wang group51 presented bioorthogonal nanoparticle detection (BOND) technology in a PEF-based suspension microarray using QDs as a signal reporter. The spherical gold-coated magnetic beads and flat gold film-coated glass were compared and served as solid support. They were decorated with anti-PSA (prostate-specific anti- gen) capture antibodies, followed by recognition and binding of the target analyte and the QDs-modified detection antibody, generating the final sand- wich immunocomplex. After collection of fluorescence images of the plas- monic slide, the imagines were analyzed by software Genepix 6.1. This soft- ware can automatically identify the features and report the fluorescence intensities of each spot with background correction. After collecting the intensity data for each point, the mean fluorescence intensity was calculated for each set of spots, which can be converted to original concentration using Poisson equation. The designed platform utilizes BOND- and PEF- amplifica- tion strategies for ultrasensitive detection of proteins with detection sensitiv- ity (1 fM) and dynamic range (5 orders of magnitude) was achieved for PSA detection (Figure 3A). They demonstrated that both spherical and flat plas- monic surfaces can enhance the fluorescence of QDs, and the immunocom- plex served as an excellent spacer. To ensure multiplexed profiling, Shao group62 developed a templated plas- monics for exosomes (TPEX) platform for multiparametric exosome analysis. This advanced method has been employed to analyze samples from 20 subjects (12 with colorectal cancer and 8 with gastric cancer), and has demonstrated its potential in predicting patient prognosis using exosomal proteins (MUC1, EpCAM, CD24, CD63) (Figure 3B). Exosomes were incubated Figure 2. Ultrasensitive plasmonic detection strategies (A) Detection of tumor-derived extracellular vesicles near a metallic array, with FDTD simulations highlighting confined electromagnetic fields near nanopores.65 (Nature Biotechnology, 2014). (B) Nanoplasmon-enhanced scattering (nPES) assay for EVs detection.66 (copyright Springer Nature, 2017). (C) Core-satellite immunoassay for the detection of IL-6.67 (copyright Elsevier Ltd., 2018). (D) Smart nanobiosensor for single molecule analysis using tsDNA17-modified Au@Ag nanocubes on an ITO glass to capture miR-21.68 (copyright American Chemical Society, 2018). REVIEW 4 The Innovation Medicine 1(2): 100023, September 21, 2023 www.the-innovation.org/medicine
  • 5. with fluorescent molecular probes and AuNPs. The exosome-bound AuNP can develop into a gold nanoshell-coated exosomes by seeding method, and the authors demonstrated that a large red shift in its plasmonic resonance can effectively quench the fluorescence signal of probes attached to the same vesicle. Besides, all the experimental steps were integrated into the miniaturized microfluidic chip that facilitated TPEX measurements of target in the complex clinical biofluids. A custom-designed smartphone-based optical detector can then be used for the image-based data acquisition and analysis, which can be completed within 15 min. Recently, Singamaneni and co-authors52 showed that a plasmonic nanoscale construct based on bovine serum albumin (BSA)-fluorophore decorated AuNRs which served as an “add-on” label for a broad range of bioassays with improved signal-to-noise ratio and dynamic range without altering their workflow and readout devices (Figure 3C). In this work, BSA was used as a scaffold in the design of nanoconstructs, acting as a stabilizing agent to prevent nanoconstruct aggregation and as a blocking agent to mini- mize non-specific binding of the plasmonic-fluor. Compared to ELISA, the presented plasmonic assay exhibited 189-fold lower LOD and more than two orders of magnitude larger dynamic range. Such device was successfully utilized to scan an array of biomarkers associated with kidney disease. However, it should be noted that the exposed fluorophore on the AuNRs surface may be suffered from photostability, a protection shell would be favorable. Zheng and co-workers fabricated Ag@SiO2 core-shell structure dopped with FITC, which are single ultrabright green particles that can be visualized by a conventional fluorescence microscope. This system permits long-term stability (6 months) with negligible attenuation of fluorescence intensity due to the silica shell protection and the resistance to the dye desorption.63 Magnetic beads and nucleic acid amplification based detection In addition to the plasmonic reporters, there are other non-plasmonic reporters available, which can be complementary candidates and have been actively explored for ultrasensitive assay. In the context of using fluores- cence probes for ultrasensitive detection, traditional method using repeated labeling of fluorophores is often laborious and can lead to photobleaching, false-positive signal as well as a low signal-to-noise ratio69 . Consequently, there is a pressing need to amplify the fluorescence signal toward single- entity detection in a robust way. In such regard, nucleic acid-labelled fluo- rophore-based detection probe in cooperation with solid supports like magnetic beads is a good option. Nucleic acid amplification (NAA) is a valu- able technique for detecting pathogens or other biomolecules by amplifying specific nucleic acid sequences.70 Numerous isothermal NAA techniques, including the catalyzed hairpin assembly (CHA),71 hybridization chain reac- tion (HCR),72 and rolling circle amplification (RCA),73,74 have emerged as promising alternatives for achieving rapid and efficient signal amplification without the need of thermocycling. The Walt group75 developed an ultrasen- sitive single-molecule detection platform that employed rolling circle isother- mal amplification and a sandwich structure for Brachyury detection, achiev- ing an impressive sensitivity of 244.6 aM (Figure 4A). In this design, magnetic microbeads (MBs) conjugated with antibodies were used to probe human cytokines interleukin-1β (IL-1β) and IL-10. Then, single-stranded DNA (ssDNA) with a specific detection antibody and an RCA trigger were used to capture target exosomes. The RCA reaction enabled numerous fluorescent probes to hybridize with the RCA products, amplifying fluorescence signals. Beads were then placed on a slide and dried, forming monodispersed beads for digital counting using colocalized red (MBs)-green (probe) fluorescence. Following the similar principle, the Gao group76 introduced modifications by using an aptamer as a recognition element instead of antibodies. They employed an addressable DNA nanoflower (DNF) attached to the surface of magnetic beads, which introduced a large number of fluorescence probes onto the nanoflower through base complementary pairing. This configuration enabled fluorescence imaging for the detection of 17β-estradiol (E2). a typi- cal environmental estrogen, and a small molecule model target (Figure 4B). The fluorescence image was automatically analyzed by Fiji image analysis Medicine Figure 3. Strategies utilizing plasmonic coupled fluorescence (A) BOND overview and its role in plasmonic suspension microarray.51 (copyright Elsevier Ltd., 2023). (B) Schematic of plasmonic-fluor as a biolabel boosting fluorescence intensity and improving signal-to-noise in assays.62 (copyright Springer Nature, 2020). (C) Templated plas- monics for exosomes (TPEX) platform showing labeled exosomes for multiparametric molecular profiling through SPR redshift signals.52 (copyright Science Advances, 2020). REVIEW The Innovation Medicine 1(2): 100023, September 21, 2023 5
  • 6. software, simplifying the testing workflow. The surface confinement strategy, which involves using NAA and attaching optical probes to solid support, is promising but comes with a set of challenges. The intricate and prolonged wash procedure during binding and polymerization stages often results in significant product loss. Moreover, the inherent negative charge of DNA tends to non-specifically adsorb positively charged molecules, leading to undesir- able false positives and high background interference. There is also a notable heterogeneity in the brightness of individual beads due to different amplifica- tion cycles, stemming from the sensitivity of enzyme activity to environmen- tal influences. To tackle these issues, appropriate antifouling strategies should be employed, such as the use of nonionic and non-protein blockers to suppress non-specific adsorption.77-79 They can help reduce DNA-induced false posi- tive signals, thereby pushing the detection limit further and improving the current fluorescence threshold. Automation for capture-detection would be another solution to overcome the multi-step manual handling caused errors. Additionally, rational fundamental design on sequences, the recently proposed one-pot amplification strategy might be a good alternative for its further evolution and improvement.80,81 It is anticipated that RCA can be further improved by using signal enhancement and noise reduction approaches. Finally, the ability to control the efficiency of aggregation on indi- vidual magnetic beads results in discrepancies in fluorescence intensity among beads, which can have a substantial influence on the outcomes. It is worth noting that reactions such as RCA, LAMP, and PCR all require enzymes, whose activity can be easily affected by the environment, leading to low stability and assay accuracy. Hu et al.82 ingeniously utilized size encod- ing approach to accomplish multiplexed detection by employing microbeads of four different sizes, followed by decoration with aptamers to target extra- cellular vesicle (EV) surface markers (Figure 4C). They designed a microflu- idic chip featuring an array of differently sized channels, which allowed magnetic beads of corresponding sizes to enter separate channels to reach their blockade with spatial resolution. This combination of size coding and spatial coding permitted simultaneous analysis of PD-L1, EpCAM, and CD63 from intact EVs for cancer diagnosis (healthy control n=10, cancer patients n=25). RCA amplification with hybridization of complementary DNA probe endowed the signal amplification capability. This study demonstrated that Figure 4. Various single-molecule detection methods based on nucleic acid amplification (A) Schematic of dropcasted single molecule assays.75 (copyright American Chemi- cal Society, 2020). (B) Principle of single-molecule detection of E2 used by addressable DNA nanoflowers and automatic digital quantitative analysis.76 (copyright American Chemical Society, 2022). (C) Schematic of size-coded affinity matrix microfluidic chip strategy for multiplex EV phenotyping.82 (copyright American Chemical Society, 2022). REVIEW 6 The Innovation Medicine 1(2): 100023, September 21, 2023 www.the-innovation.org/medicine
  • 7. this tEV phenotyping method can rapidly and simultaneously detect six different tEV phenotypes with high sensitivity (Table 1). Visible microsphere In the aforementioned discussion, signal amplification techniques are high- lighted, which enables the conversion of undetectable signals into observ- able fluorescent outputs through confinement-based signal enhancement. This method involves directly doping reporters like fluorescent tags to micro- spheres. The number of doped fluorophores and their intrinsic quantum yield determines the brightness of resultant fluorescent microbeads. Alternatively, the microbeads can be used as reporters as they can easily be visualized by conventional microscope. Noji et al.83 introduced a sensitive wash-free detec- tion method for PSA (Figure 5A). In their work, PSA reacted with antibody- coated magnetic nanoparticles (~800 nm) and were then pooled into a femtoliter-sized reactor by magnetic force. The images were analyzed using Image J and Trackmate softwares. These tools can localize the center of individual particles by calculating the intensity of light spots. As a result, the number of tethered particles could directly correlate with the concentration of the target antigen. The number of particles tethered was proportional to the concentration of the target antigen. This method directly treated magnetic microbeads as reporters without the need of additional dyes. However, this method requires extensive mathematical calculations of the Brownian motion of molecules and imaging validation through other instruments. Yiping Chen84 and his team encoded multiple targets using polystyrene microspheres with five different particle sizes, and the unconjugated microspheres in the super- natant after immunoreaction were recorded for subsequent visible signals under an optical microscope (Figure 5B). The obtained images were further analyzed using artificial intelligence computer vision technology to decode the inherent characteristics of the polystyrene microspheres and revealed the type and concentration of the target. This method sidesteps the complexities of chip production and signal amplification but requires precise size control of the microspheres. Non-specific adsorption and precipitation must also be considered. The bright-field microscopy is straightforward but impurities or dusts can interfere, offering opportunity for the fluorescently encoded micro- spheres. To this end, He et al.85 directly labeled magnetic beads (~1 µm) with fluorescent microspheres (~120 nm) to capture the target using a compo- nents of capture DNA-bead, analyte, and detection DNA-fluorescent micro- sphere in an order (Figure 5C). Using SARS-CoV-2 ssDNA as the model, they achieved a 1.5 fM detection limit by counting and decoding bead-micro- sphere pairs and analyzed the data with NIH ImageJ software.86 This study highlights the potential of single-particle luminescence for single-molecule detection. Noteworthy materials include aggregation-induced emission (AIE) Medicine Table 1. The typical examples using external-/internal- confinement strategy for ultrasensitive optical assay. Confinement Type Analyte LOD Working Range Volume Ref. External confinement Plasmonic alone CD63 NA NA 1 μL 62 External confinement Plasmonic alone EphA2 0.2 ng/μL 0.1–10000 ng/uL 1 μL 133 External confinement Plasmonic alone miR-21 0.1 aM 1 aM to 1 nM NA 68 External confinement Plasmonic with fluorescence ALP 5.8 μU/mL 0.06 to 2.48 mU/mL 100 μL 134 External confinement Plasmonic with fluorescence PSA 1 fM 1 fM-1 nM NA 135 External confinement Nucleic acid amplification 17β-estradiol (E2) 63.09 × 10−6 nM 0.001836 to 183.6 nM 10 μL 76 External confinement Nucleic acid amplification IFNγ and IL-2 (3×) 30 aM and 20 aM NA 2 μL 136 External confinement Nucleic acid amplification IL-6 13 pM NA 250 μL 137 External confinement Nucleic acid amplification PSA 97.2 aM 1.62 nm 2 μL 138 External confinement Nucleic acid amplification CEACAM-7 2.82 pM NA 25 μL 139 External confinement Nucleic acid amplification LwaCas13a 15.8 fM 10 cp/μL up to 106 cp/μL 2 μL 140 External confinement without amplification SARS-CoV-2 (ssDNA) 1.5 fM 0 to 1 pM 5 μL 85 External confinement without amplification CEA 1.2 pg/mL NA NA 141 External confinement without amplification Aflatoxin B1 0.17 ng/mL 3.13 to 125.00 ng/mL 15 μL 142 External confinement without amplification PSA 1.2 pg /mL 10 pg/mL and 1 ng/mL 100 μL 143 Internal confinement Droptlet(oil in water) β-gal NA 0.1 cpd-3 cpd(40-1202 fM) NA 144 Internal confinement Droptlet(oil in water) PSA 0.48 ng/mL 0.5–30 ng /mL 30 μL 145 Internal confinement Droptlet(oil in water) HPV’s target DNA 2000 copies/mL 107 copies/mL to 103 copies/mL 100 μL 92 Internal confinement Droptlet(oil in water) BβG 930 zM 1 aM to 1 fM 400 μL 146 Internal confinement Droptlet(oil in water) Exosomes 10−17 M 10-105 Exosomes/uL NA 93 Internal confinement Droptlet(hydrogel) IL-6 1.36 fM 0-50 fM 10 μL 147 Internal confinement microwell femtomolar proteins 2.84 fM 493 aM (32 fg/mL)- 120 fM (7.8 pg/mL) 2 μL 148 Internal confinement microwell Thyroid stimulating hormone 0.0013 μIU/mL 0.02-1 μIU/mL 100 μL 100 Internal confinement microwell CD9+/CD63+ EVs NA 30-22800 ng/uL 3-8 μL 149 Internal confinement microwell serum cytokines 0.198pg/ml 0.16 pg/mL- 2.5 ng/mL <15 μL 103 ALP, alkaline phosphatase; PSA, prostate-specific antigen; β-Gal, β-galatosidase; BβG, biotinylated β-galactosidase; EphA2, Ephrin type-A receptor 2; CEACAM-7, CEA Cell Adhesion Molecule 7 REVIEW The Innovation Medicine 1(2): 100023, September 21, 2023 7
  • 8. molecules embedded within metal-organic frameworks (MOFs) that intensify fluorescence emission with the restraint movement by the MOF cage, making it favorable optical reporter. Similarly, upconversion nanoparticles (UCNPs)87 are anti-Stokes luminescent materials that transform low-energy near- infrared (NIR) radiation into higher-energy radiation. They are favorable for detection with reduced interference and resistance to photobleaching. Carbon dots (CDs)88,89 are photoluminescent nanomaterials that are water- soluble, biocompatible, and photostable, fitting the need for biological and environmental sample detection. Ongoing research in luminescent materials will bolster single-particle detection, broadening its applications in diagnos- tics, environmental monitoring, and chemical sensing. To conclude, the external confinement strategies widely employ micro-/ nano- architectures with the adhesion of optical reporters (e.g., organic dyes, plasmonic nanoparticles, quantum dots, etc.) on to their outer surface. In cooperation with signal amplification strategies like nucleic acid amplifica- tion or surface plasmon-enhanced fluorescence, the signal output can be dramatically enhanced to permit the stable probing of target at ultra-low levels, even single-entity level. It is expected that more novel formats on the nanoarchitecture design or signal enhancement and the cooperation with internal confinement will be introduced to achieve a synergetic effect, which would be the next hot spot. INTERNAL CONFINEMENT STRATEGY Besides the external confinement strategy, the internal strategy has also been actively explored as a vital complementary way. In such system, the signal output reporters, e.g., fluorophores or catalyzed photonic/colorful product, are usually confined within a small minuscule internal compartment of an architecture like microwell, droplet, metal-organic-frame (MOF), hydro- gel, etc. The signal output molecules are restricted, producing localized ultra- high concentrations with a resultant amplified signal. In the case of droplet- based systems, soft materials like emulsions or hydrogels are used to confine the signal molecules, restricting the signal output within the liquid droplet boundaries. For microwell-based systems, rigid materials such as plastics or silicon are used to form an array of small wells, limiting the signal output within the confined space of individual wells. Confinement within droplet In droplet system, mineral oil wrapped aqueous phase is commonly used. By application of positive external pressure to the inlet or negative pressure to the outlet generated through syringe pump90 or syringe with a binder clip91 , respectively, the stable water-in-oil droplet format can be formed. Only if the enzyme and substrate are encapsulated in the same droplet, the enzyme can catalyze and produce a fluorescent product thus illuminate the droplet under Figure 5. Various single-molecule detection methods based on microspheres (A) Digital HoNon-ELISA: Detection of protein biomarkers using antibody-coated magnetic nanoparticles.83 (copyright American Chemical Society, 2019). (B) APT Immunoassay: AI-driven assay using 2D microspheres for multi-target detection.84 (copyright American Chemical Society, 2023). (C) Enzyme-free Assay: Detects SARS-CoV-2 ssDNA with a bead-based sandwich structure.85 (copyright Elsevier Ltd, 2023). REVIEW 8 The Innovation Medicine 1(2): 100023, September 21, 2023 www.the-innovation.org/medicine
  • 9. excitation. Then, digital readout (“1” or “0”) can be generated once the number of droplets is far more than that of the target analyte.92 Zheng et al.,93 utilized the capture antibody (anti-CD63)-detection antibody (anti-GPC-1) co- localization strategy for the single-exosome detection with an achievable LOD down to ~10−17 M and a dynamic range of 5 log of the linear regime (Figure 6A). Although the droplet system using enzyme-catalyzed fluores- cence within a confined volume can generate a strong signal for ultrasensi- tive measurement and save sample consumption, it requires complex manual operations, e.g., the stepwise capture processes must be done outside the chip, and it only permits singplexing using current format. In such regard, the capability for automation, integration, and multiplexed profiling is favorable. Moreover, to further enhance the effectiveness of the droplet- based chip, it is crucial to develop a matched mobile platform that facilitates fast and easy access to assay data by integrating mobile device, thus high- lighting the importance of improved connectivity and user-friendly interfaces. The Walt team94 has developed a platform for rapid, sub-picogram-per- milliliter, multiplexed digital droplet detection by using color-encoded beads and droplet microfluidics comprising of partition-incubation-detection compartments (Figure 6B). This innovative approach employs a mobile phone-based imaging technique and coding-based duplexed profiling, elimi- nating the need for costly benchtop optics and ensuring a detection speed 100 times faster than conventional assays due to time domain-encoded mobile phone imaging. The performance of this assay was characterized by spiking recombinant proteins in fetal bovine serum (FBS) with a limit of detection (LOD) at 4 fg/mL (i.e., 300 aM). By integrating the coding-based duplexed profiling, they further optimized the system performance, enhanc- ing its detection accuracy and reliability. This development will aid its commercialization, making a user-friendly system for self-testing anytime and anywhere. Liang’s group95 devised a microfluidic system, known as digi- tal droplet with auto-catalytic hairpin assembly (ddaCHA), for the digital quantification of single microRNAs (miRNA) (Figure 6C). This process involves partitioning miRNA molecules into individual picoliter-sized droplets, generating a digital readout in which the target molecule is classified as either present ("positive") or absent ("negative"). The researchers successfully implemented an enzyme-free auto-catalytic hairpin assembly (aCHA) within the droplets, demonstrating its effectiveness in minimizing the impact of external environmental factors and temperature fluctuations on droplet performance. In their investigation, an automated Python program was employed to monitor and compute the fluorescence intensities of individual droplets when they are flowing through a high-throughput microfluidic chip. By assigning a distinct identifier to each droplet within the flow channel, the script ensured that each droplet was only quantified once, thereby eliminat- ing the risk of replicated counting. The significance of automation in such detection systems is obvious as it considerably reduces manual labor and minimizes potential errors in the process. However, the complexity of these systems can be daunting for untrained end users, prompting the development of new approaches that aim to simplify or automate the microfluidic steps. Wang group96 presented a novel lab-on-a-particle assay that employs hydrogel particles for simultaneous immobilization of protein molecules and emulsification templating, enabling digital counting of β-galactosidase. Following this process, image analysis algorithms in MATLAB are used to analyze the fluorescence signal of each droplet by averaging signals over the particle. A signal is considered to be positive if it exceeds 2.5 × standard deviations above the mean of the back- ground signals (Figure 6D). Moving forward, the efforts will be paid for refin- ing this process and measuring actual targets further using appropriate affin- Medicine Figure 6. Droplet-based strategies for single-molecule detection (A) Droplet digital ExoELISA for exosome quantification using magnetic beads in microdroplets for fluorescent readout.93 (copyright Springer Nature, 2018). (B) The µMD's droplet generation and detection for mini dELISA on mobile platforms with antibody-functionalized, color-coded beads.94 (copyright National Academy of Science, 2019). (C) Droplet-generating CFF junction design for single-molecule miRNA imaging.95 (copyright American Chemical Society, 2022). (D) Production of Hydrogel particles through step emulsification at low pH.96 (copyright Royal Society of Chemistry, 2021). REVIEW The Innovation Medicine 1(2): 100023, September 21, 2023 9
  • 10. ity capture probes. Droplet-based single-molecule detection has shown great potential in vari- ous applications, however, it presents certain drawbacks. For instance, droplet generation and manipulation can be technically challenging, and droplet stability may be affected by evaporation or coalescence over time.97,98 Additionally, droplet-based approaches may not be suitable for all sample types or analytes, such as those with high viscosity or complex molecular structures due to challenges in generating uniform droplets and potential loss of analyte integrity. Furthermore, recovering analytes from droplets for further analysis could be difficult. These limitations call for the need of alternative approaches like microwell-based single particle detection, which may provide a more robust and versatile solution for many applications. Confinement within microwell Microwell strategies serve as an alternative to droplet-based techniques by confining chromatic or luminescent products within a tiny-volume microw- ells rather than the water-in-oil droplets. The microwell system is usually generated by nano-/micro-fabrication. Microwell-based techniques offer a controlled and stable environment for target analysis, as they do not rely on droplet generator and the complex manipulation. By employing microwell arrays, researchers can effectively isolate and analyze individual particles. Microwell confinement-based detection was originally developed by the Walt group99 . This detection method, also referred to as Single Molecular Array (Simoa), has been successfully commercialized. In the Simoa approach, each femtoliter-sized reaction chamber in the array contains a single magnetic bead, and the number of microbeads (MBs) is deliberately maintained to significantly exceed the number of target molecules from highly diluted sample. In essence, it utilizes the concept of transformation of on-bead confinement to the micro-well confinement system, the enzymatically catalyzed fluorescence can be monitored by ultrasensitive CCD camera. The beads with the sandwich complexes are then loaded into these chambers, with each chamber designed to accommodate one bead. Subsequently, a fluorogenic substrate is introduced, which allows the enzyme on the detec- tion antibody to convert the substrate into a fluorescent product. This process results in a binary "0" or "1" signal output, corresponding to the absence or presence of the target analyte, enabling highly sensitive detection. However, the Simoa system highly relies on the pricey bulk instrument and micro-well plates, which is not affordable to many remote areas and source- limited regions. Also, the current format is mainly designed for singleplexed profiling while multiplexing ability is still limited. As microwell detection has evolved with continuous improvements and refinements, researchers have explored its potential in various applications. For instance, Karen group100 has developed a novel digital microfluidics (DMF) chip-based method for detecting thyroid-stimulating hormone (TSH), a criti- cal marker for assessing thyroid function. Such device requires a tiny sample volume of 1.1 µL and offers a LOD at 1.3 nanoIU/mL (Figure 7A). Microwells Figure 7. Microwell confinement-based detection platforms (A) Schematic illustration of the digital microfluidics (DMF) chip with microwell arrays.100 (copyright American Chemical Society, 2022). (B) Schematic illustrations of bead trapping.101 (copyright Elsevier Ltd., 2019). (C) Schematic of the microwell-based biosensor with edge enhancement.102 (copyright American Chemical Society, 2021). (D) Schematic illustration of the digital microfluidics (DMF) chip with microwell arrays.103 (copyright Elsevier Ltd., 2021). REVIEW 10 The Innovation Medicine 1(2): 100023, September 21, 2023 www.the-innovation.org/medicine
  • 11. were fabricated on a PET substrate using a UV imprinting process and resist material through a roll-to-roll fabrication method. In the DMF chip, precise control of electric fields between the top and bottom electrodes enables effi- cient droplet manipulation within these microwells, and the hydrophobic-in- hydrophobic microwells on the chip also boost a higher seeding efficiency (97.6% ± 0.6%) due to minimized surface energy, ensuring stable droplet confinement, reduced merging or spreading. In another study, the Sun group101 has developed a microfluidic chip using polydimethylsiloxane (PDMS) based microwell patterned arrays to detect tumor necrosis factor α (TNF-α) (Figure 7B). Beads were injected into a chip and were allowed to settle for several minutes due to gravity. Subsequently, oil was injected through the inlet to seal an aqueous solution containing a single bead in the well. This power-free oil-sealed water phase eliminates the need for external pumping systems or valves, making it a cost-effective approach. Further- more, the fabrication cost of these chips is low, and there is no need for complex surface modifications. The method also maintains good sensitivity with a limit of detection (LOD) of 12.62 fg/mL, which is comparable to perfor- mance using the commercial Simoa HD-1 Analyzer (14 fg/mL). In order to amplify optical signals and increase sensitivity, Li group102 used edge- enhanced microwell (40 μm depth) immunoassay for detection of interferon- γ (Figure 7C). The microwells are coated with capture antibodies, which facili- tate the binding of IFN-γ and the following Texas conjugated detection anti- bodies to form sandwich immune-complex. Due to larger axial resolution along with the vertical detection, the fluorescence emission was enhaced, then the signals of the analytes will be amplified greatly, which formed fluo- rescent rings. The edge enhancement effect of the microwell vertical side- wall shows a 6-fold sensitivity enhancement compared to those obtained on a flat surface. It is worth mentioning that all the examples presented above are focusing on detecting single target, which may not reflect disease evolution and progression accurately. Also, lots of diseases originate by complex multiple factors, thus there is a need for multiplexed assays to permit multi-analyte profiling. In conventional assays, the detection of multiple biomarkers from one set of samples is challenging due to the complexity of encoding and decoding of numerous signal output from different analytes, and various error sources can lead to a decrease in the accuracy of the assay.54 Therefore, rational design that allows distinguishable signal readouts, decent sensitivity, and specificity is the key pathway. Recognizing the existing limitations, Sung group103 introduced the pre-equilibrium digital ELISA (PEdELISA) microarray platform that leverages spatial-spectral encoding and machine learning on a microfluidic chip (Figure 7D). This allows 12-cytokine assay using only 15 μL of serum. The 12-plex PEdELISA microarray uses a CNN-based parallel computing algorithm, ensuring unsupervised image data analysis with high accuracy. It autonomously classifies and segments image features at a high throughput (1 min/analyte), achieving 8‒10 times higher accuracy than tradi- tional GTS-based algorithms without manual corrections. The entire detec- tion and analysis cycle can be completed within 40 min. In essence, PEdELISA enhances multiplexing efficiency by integrating advanced microfluidics with a machine learning image processing method. AI APPROACH FOR OPTICAL DETECTION IN CLINICAL ANALYSIS Indeed, with the remarkable advancements in hardware technology, there has been an extraordinary surge in the development and implementation of artificial intelligence (AI) tools that are specifically designed for disease diag- nosis. These bespoke AI systems excel at processing vast volumes of data and images sourced from a myriad of equipment. By incorporating AI models, such as Linear Discriminant Analysis (LDA)104-106 , Support Vector Machines (SVM)107,108 , Logistic Regression, Linear Regression, Linear Mixed Models and Random Forest Models (RFM)109,110 , these systems significantly enhance both the efficiency and accuracy of diagnostic procedures. Within the scope of our specialized system, the primary objective is the automated detection and quantification of visible particle signals as particle count can act as a surro- gate for biomarker concentration. By accurately quantifying specific protein biomarkers in patients, these AI-driven approaches can significantly advance medical research and patient care, potentially leading to earlier interventions and improved treatment outcomes. The schematic diagram of this AI appli- cation in our review can be summarized as follows: Image acquisition → Particle detection and counting → Biomarker concentration representation → Data Analysis→Diagnosis (Figure 8). This progression succinctly captures the role of AI in augmenting diagnostic procedures in healthcare. Automation and standardization of image processing AI technology has revolutionized optical detection by automating data processing, boosting efficiency, and reducing human error. In image process- Medicine Figure 8. The general process of AI techniques for data interpretation REVIEW The Innovation Medicine 1(2): 100023, September 21, 2023 11
  • 12. ing, AI excels in automating particle counting through several steps: (1) Image Preprocessing: AI algorithms enhance image quality by decreas- ing noise and elevating contrast. Techniques like Gaussian filtering, thresh- olding, and contrast normalization111,112 are used for clarity, facilitating better detection and accurate particle counting. (2) Object Detection: Depending on image complexity, methods range from connectivity analysis for simple images to advanced techniques like edge detection and machine learning for intricate ones. (3) Object Counting: After segmentation, objects are enumer- ated using tools like ImageJ/FIJI, MATLAB, and Python libraries such as OpenCV and Scikit-Image. The Chen group113 demonstrated this, differentiat- ing polystyrene (PS) microspheres with a CV technique, achieving 93.5% to 100% accuracy, surpassing traditional methods. For example, the Chen group113 utilized computer vision (CV) methods to differentiate and quantify polystyrene (PS) microspheres of varying sizes. (Figure 9A) Through tech- niques like binarization thresholding and a customized algorithm, they achieved impressive accuracy ranging from 93.5% to 100%, surpassing tradi- tional manual methods. To sum up, ultra-sensitive optical detection can be realized through vari- ous rational signal amplification strategies, translating minuscule signals into countable particles, bypassing the need for complex AI technologies typically required in other systems. However, when a scenario necessitates more intri- cate analysis, such as distinguishing signals based on their intensity or corre- lating signal strength with specific biomarker concentrations, advanced algo- rithms and strategies become invaluable. Nevertheless, the incorporation of AI, even within these simplified environments and small-scale clinical sample analysis, significantly enhances the accuracy and reliability of our method- ologies. AI approaches to data interpretation Analysis of collected biomarker data plays a crucial role in gaining a comprehensive understanding and diagnosis of diseases. The abundance and diversity of biomarkers offer valuable insights for disease diagnosis, monitoring treatment response, tracking disease progression, and identifying disease subtypes. With the advent of AI techniques, the analysis of biomarker data has become more efficient and accurate, enabling the delivery of personalized healthcare services to patients. In the following section, we will elaborate how various AI techniques work on these critical medical areas. In the domain of early disease diagnosis, SVM-based methods are notably adept at dealing with high-dimensional data, like gene expression profiles. Their proficiency in extracting complex patterns from this data allows them to discern distinct gene expression signatures that could separate cancer cells apart from healthy cells. This distinguishing capability is pivotal in facilitating the early detection of cancer. Given the prominence of high-dimensional gene expression data in cancer research, the application of SVMs might be more prevalent in this field than in others.114,115 However, their performance depends on the correct selection of a kernel function and adjustment of parameters, which may require specialized domain knowledge. Further research in this area could greatly enhance the applicability of SVMs. Moving on to the monitoring of treatment responses, Random Forest models play a central role in this domain.116-119 These models are ensemble learning methods that combine multiple decision trees. This approach could provide an understanding of the intricate interactions between various biomarkers and treatment outcomes. For instance, when managing chronic diseases like diabetes, Random Forest models can effectively handle diverse Figure 9. AI-assisted strategies for disease diagnosis (A) Schematic illustration of workflow and principles of CV-enabled image processing for simultaneous detection.113 (copyright American Chemical Society, 2023). (B) Schematic illustration of machine learning-based SERS detection by PCA-LDA.125 (copyright American Chemical Society, 2023). REVIEW 12 The Innovation Medicine 1(2): 100023, September 21, 2023 www.the-innovation.org/medicine
  • 13. data, ranging from glucose levels and insulin doses to lifestyle factors like diet and physical activity.120 This capability could help develop a more personal- ized and effective treatment strategies. However, the complexity of the model can increase as the number of features or variables grows, making it chal- lenging to interpret the results and understand the underlying patterns effec- tively. Therefore, future research may explore more effective ways to opti- mize the number of decision trees in the ensemble or propose new data dimensionality reduction techniques. Linear Discriminant Analysis (LDA) plays an important role in tracking disease progression.121-124 Its capability to simplify high-dimensional biomarker data makes it instrumental in identifying longitudinal changes in biomarker levels, paving the way for a deeper understanding of disease trajectories over time. For instance, when monitoring heart disease progres- sion, LDA can be applied to unravel dynamic alterations in multiple biomark- ers, such as cholesterol levels, blood pressure readings, and inflammation markers. This analytical power aids in understanding the fluid nature of disease progression and enabling timely interventions. For example, Jin group 125 employed a combined approach of principal component analysis (PCA) and Linear Discriminant Analysis (LDA) to extract features from high- dimensional Surface-Enhanced Raman Scattering (SERS) spectra (Figure 9B). First, they performed dimensionality reduction on the SERS spectra using PCA, reducing them to 46 principal components that accounted for 95% of the total variance. Next, they applied LDA to further reduce the dimensionality of these 46 principal components to just two LDA factors. Remarkably, this combined approach resulted in a clear separation and accurate identification of exosomes derived from normal cells and cancerous cells. In the field of disease subtyping, linear and logistic regression models serve as potent tools for grouping.126,127 These models can shed light on the associ- ations between biomarkers and specific disease subtypes. In addition, these models can integrate an array of biomarkers, such as gene expression profiles and metabolic markers, to estimate disease subtypes, enabling more effective and personalized management plans. In conclusion, AI techniques have demonstrated their effectiveness in analyzing biomarker data, facilitating a deeper understanding and diagnosis of diseases. These techniques contribute to early disease detection, treat- ment response monitoring, disease subtyping, ultimately providing more accurate and tailored healthcare services for patients. However, it is also essential to be aware of potential challenges. AI models heavily rely on the quality of the training data. Therefore, ensuring the accuracy, completeness, and standardization of biomarker data is critical to yield reliable results. In addition, these models often involve complex mathematical computations and require domain knowledge for optimal parameter tuning, highlighting the need for interdisciplinary collaborations between clinicians, data scientists, and bioinformaticians. Another potential challenge in embracing AI advance- ments is overfitting due to sample size. Overfitting occurs when a model is excessively complex and learns noise or specific patterns from the training data that do not generalize well to real data. Another area that warrants attention is the interpretability of these models. While AI models can make highly accurate predictions, the underlying reasoning can be opaque, a phenomenon often termed as the "black box" problem. This lack of transparency can hinder physicians' trust in the model predictions, thereby limiting their adoption in clinical practice. Lastly, ethical and legal considerations surrounding data privacy and security, and algorith- mic bias should also be considered when deploying AI in healthcare. As we move towards this future, it is imperative to address these challenges head- on to fully harness the potential of AI in transforming patient care and outcomes. The amalgamation of AI with medical science is truly an exciting frontier. It promises not only advancements in disease understanding and management but also a new era of personalized medicine. With careful navi- gation, we stand on the cusp of a healthcare revolution. CONCLUSION AND PERSPECTIVE This review provides a comprehensive view of confinement-based ultra- sensitive optical assays (Table 1), enabling the (digital) measurement of analytes without relying on advanced imaging systems such as total internal reflection fluorescence (TIRF)43,44 microscopy or photo-activated localization microscopy (PALM).128,129 As the investigation of novel and uncommon disease markers expands, along with the need to decipher multifactorial diseases, there is a rising demand for the development of unsophisticated, ultrasensitive, and multiplexed detection platforms to address these complex challenges. Luminescent systems, either through self-illuminating compounds or through the products of enzymatically induced luminescence or color systems, can be confined to internal or external spaces. These systems can cooperate with numerous signal amplification approaches, such as incorporating surface plasmon enhancement and nucleic acid amplifica- tion, to realize ultra-bright and ultra-strong signal readouts. The core aim is to harmonize the cooperation of signal amplification with noise reduction, reaching a balance that is crucial to achieving a distinguishable signal from the background at a single-entity level. This balance plays a key role in enabling early diagnosis and treatment. However, this cutting-edge detection approach still faces significant obstacles and challenges, highlighting the need for further investigation and resolution to fully exploit its potential in diagnostic and therapeutic applications. Ultrasensitive optical detection requires specific probes to avoid false results. Enhancing probe specificity can be achieved through unique molecu- lar design, directed evolution, high-throughput screening, multi-domain probes, and computational modeling. These steps can substantially improve the accuracy and reliability in applications like disease diagnosis. Various signal amplification strategies aim for ultrasensitivity, with the reduction of background noise being paramount. Incorporating anti-fouling techniques, such as PEGylation and zwitterionic coatings, can minimize contaminants, enhancing detection accuracy.130 With the rise of automation and AI, their synergy with ultrasensitive detec- tion is gaining traction. As the detection process requires sample purification to eliminate interference, automation offers improved efficiency and accu- racy over traditional methods. AI is particularly adept at rapidly interpreting images obtained through microscopy. Moreover, training AI on vast patient datasets allows for predicting disease trajectories and crafting tailored treat- ment plans. By fusing AI with particle-guided optical assays, we pave the way for innovative disease diagnostics and treatments. While many of these tech- nologies remain in their nascent stages, leveraging larger sample sizes in the future could lead to significant breakthroughs. This evolution symbolizes the shift from mere big data to its profound mining, optimizing the union of AI and detection hardware.131 The intersection of biotechnology (BT) and information technology (IT) is undeniably shaping our present and future. To achieve impactful outcomes, strategies in confinement, signal amplification, and encoding must seam- lessly integrate. The overarching goal is to craft miniaturized, automated, and smart systems for monitoring evolving disease threats with validated mark- ers, a venture that underscores the importance of interdisciplinary teamwork. The next decade promises transformative advancements, especially in single- entity detection technology.132 With potential applications spanning molecu- lar biology, biomedicine, to nanotechnology, it is essential to address any challenges and champion continued innovation for its broader acceptance. Such progression is vital for proactive disease management, improving human health, and realizing novel technological leaps. REFERENCES Cheung, A.H.K., Chow, C., and To, K.F. (2018). Latest development of liquid biopsy. J.Thorac. Dis. 10, S1645-S1651. 1. Miller, D.B., and O’Callaghan, J.P. (2015). Biomarkers of Parkinson’s disease: present and future. Metabolism 64, S40-S46. 2. Counts, S.E., Ikonomovic, M.D., Mercado, N., et al. (2017). Biomarkers for the early detection and progression of Alzheimer’s disease. Neurotherapeutics 14, 35-53. 3. Gangi, S., Fletcher, J., Nathan, M.A., et al. (2004). Time interval between abnormalities seen on CT and the clinical diagnosis of pancreatic cancer: retrospective review of CT scans obtained before diagnosis. Am. J. Roentgenol. 182, 897-903. 4. Metintas, M., Ak, G., Dundar, E., et al. (2010). Medical thoracoscopy vs CT scan- guided Abrams pleural needle biopsy for diagnosis of patients with pleural effusions: a randomized, controlled trial. Chest 137, 1362-1368. 5. Helpern, J.A., Jensen, J., Lee, S.P., and Falangola, M.F. (2004). Quantitative MRI assessment of Alzheimer's disease. J. Mol. Neurosci. 24, 45-48. 6. Morrow, M., Waters, J., and Morris, E. (2011). MRI for breast cancer screening, diagnosis, and treatment. Lancet 378, 1804-1811. 7. Gao, Q., Asthana, A., Tong, T., and Rueckert, D. (2014). Multi-scale feature learning 8. Medicine REVIEW The Innovation Medicine 1(2): 100023, September 21, 2023 13
  • 14. on pixels and super-pixels for seminal vesicles MRI segmentation. Medical Imaging 2014: Image Processing. Spie, 2014, 9034, 36-41. Braum, L.S., McGonagle, D., Bruns, A., et al. (2013). Characterisation of hand small joints arthropathy using high-resolution MRI —limited discrimination between osteoarthritis and psoriatic arthritis. Eur radiol 23, 1686-1693. 9. Zarovni, N., Corrado, A., Guazzi, P., et al. (2015). Integrated isolation and quantitative analysis of exosome shuttled proteins and nucleic acids using immunocapture approaches. Methods 87, 46-58. 10. Chi, X., Huang, D., Zhao, Z., et al. (2012). Nanoprobes for in vitro diagnostics of cancer and infectious diseases. Biomaterials 33, 189-206. 11. Zhang, Z., and Chan, D. W. (2010).. The Road from discovery to clinical diagnostics: lessons learned from the first FDA-cleared in vitro diagnostic multivariate index assay of proteomic biomarkers. Cancer Epidem. Biomar. 19, 2995-2999. 12. Pritchard, C.C., Cheng, H.H., and Tewari, M. (2012). MicroRNA profiling: approaches and considerations. Nat. Rev. Genet. 13, 358-369. 13. St John, A., and Price, C.P. (2014). Existing and emerging technologies for point-of- care testing. Clin Biochem. Rev. 35, 155-167. 14. Whitesides, G.M. (2006). The origins and the future of microfluidics. Nature 442, 368- 373. 15. Han, F., Wang, T., Liu, G., et al. (2022). Materials with tunable optical properties for wearable epidermal sensing in health monitoring. Adv. Mater. 34, e2109055. 16. Kaur, J., Jiang, C., and Liu, G. (2019). Different strategies for detection of HbA1c emphasizing on biosensors and point-of-care analyzers. Biosens. Bioelectron. 123, 85-100. 17. Liu, G., Jiang, C., Lin, X., and Yang, Y.J.V. (2021). Point‐ of‐ care detection of cytokines in cytokine storm management and beyond: significance and challenges. View 2, 20210003. 18. Fu, Y., Jiang, C., Tofaris, G.K., and Davis, J.J. (2020). Facile impedimetric analysis of neuronal exosome markers in Parkinson’s disease diagnostics. Anal. Chem. 92, 13647-13651. 19. Chen, R., Mias, G.I., Li-Pook-Than, J., et al. (2012). Personal omics profiling reveals dynamic molecular and medical phenotypes. Cell 148, 1293-1307. 20. Sharafeldin, M., Yan, S., Jiang, C., et al. (2023). Alternating magnetic field-promoted nanoparticle mixing: the on-chip immunocapture of serum neuronal exosomes for Parkinson’s disease diagnostics. Anal. Chem. 95, 7906–7913. 21. Jiang, C., Alam, M.T., Silva, S.M., et al. (2016). Unique sensing interface that allows the development of an electrochemical immunosensor for the detection of tumor necrosis factor α in whole blood. ACS Sens. 1, 1432-1438. 22. Cao, C., Zhang, Y., Jiang, C., et al. (2017). Advances on aryldiazonium salt chemistry based interfacial fabrication for sensing applications. ACS Appl. Mater. Interfaces 9, 5031-5049. 23. Vasilieva, A., Yurina, L., Azarova, D.Y., et al. (2022). Development of a diagnostic approach based on the detection of post-translation modifications of fibrinogen associated with oxidative stress by the method of high efficiency liquid chromatography. J Phys Chem B.16, 118-122. 24. Chiki, A. (2020). Development of novel methods and tools to decipher the huntingtin post-translation modifications code. EPFL. 25. Jiang, C., Hopfner, F., Katsikoudi, A., et al. (2020). Serum neuronal exosomes predict and differentiate Parkinson’s disease from atypical parkinsonism. J Neurol Neurosurg Psychiatry 91, 720-729. 26. Mitchell, P.S., Parkin, R.K., Kroh, E.M., et al. (2008). Circulating microRNAs as stable blood-based markers for cancer detection. PNAS 105, 10513-10518. 27. Weber, J.A., Baxter, D.H., Zhang, S., et al. (2010). The microRNA spectrum in 12 body fluids. Clin. Chem. 56, 1733-1741. 28. Chen, X., Ba, Y., Ma, L., et al. (2008). Characterization of microRNAs in serum: a novel class of biomarkers for diagnosis of cancer and other diseases. Cell Res. 18, 997- 1006. 29. Lu, X., Hu, C., Jia, D., et al. (2021). Amplification-free and mix-and-read analysis of multiplexed microRNAs on a single plasmonic microbead. Nano Lett. 21, 6718- 6724. 30. Wang, G., Tian, W., Liu, X., et al. (2020). New CRISPR-derived microRNA sensing mechanism based on Cas12a self-powered and rolling circle transcription- unleashed real-time crRNA recruiting. Anal. Chem. 92, 6702-6708. 31. Goetzl, L., Merabova, N., Darbinian, N., et al. (2018). Diagnostic potential of neural exosome cargo as biomarkers for acute brain injury. Ann. Clin. Transl. Neurol. 5, 4- 10. 32. Singh, K., Nalabotala, R., Koo, K.M., et al. (2021). Separation of distinct exosome subpopulations: isolation and characterization approaches and their associated challenges. Analyst 146, 3731-3749. 33. Picciolini, S., Gualerzi, A., Vanna, R., et al. (2018). Detection and characterization of different brain-derived subpopulations of plasma exosomes by surface plasmon resonance imaging. Anal. Chem. 90, 8873-8880. 34. Yousif, G., Qadri, S., Haik, M., et al. (2021). Circulating exosomes of neuronal origin as potential early biomarkers for development of stroke. Mol Diagn Ther 25, 163- 180. 35. Yan, S., Jiang, C., Davis, J.J., and Tofaris, G.K. (2023). Methodological considerations in neuronal extracellular vesicle isolation for α-synuclein biomarkers.Brain, awad169. 36. Kang, Y.T., Hadlock, T., Lo, T.W., et al. (2020). Dual‐ isolation and profiling of circulating tumor cells and cancer exosomes from blood samples with melanoma using immunoaffinity‐based microfluidic interfaces. Adv. Sci. 7, 2001581. 37. Słomka, A., Wang, B., Mocan, T., et al. (2022). Extracellular vesicles and circulating tumour cells-complementary liquid biopsies or standalone concepts? Theranostics 12, 5836. 38. Schwarzenbach, H., Hoon, D.S., and Pantel, K.J. (2011). Cell-free nucleic acids as biomarkers in cancer patients. Nat. Rev. Cancer 11, 426-437. 39. Jahr, S., Hentze, H., Englisch, S., et al. (2001). DNA fragments in the blood plasma of cancer patients: quantitations and evidence for their origin from apoptotic and necrotic cells. Cancer Res. 61, 1659-1665. 40. Zhang, H.Y., Wang, S., and Fang, G.Z. (2011). Applications and recent developments of multi-analyte simultaneous analysis by enzyme-linked immunosorbent assays. J. Immunol. Methods 368, 1-23. 41. Lequin, R.M. (2005). Enzyme immunoassay (EIA)/enzyme-linked immunosorbent assay (ELISA). Clin. Chem. 51, 2415-2418. 42. Steyer, J.A., and Almers, W.J.N.r.M.c.b. (2001). A real-time view of life within 100 nm of the plasma membrane. Nat. Rev. Mol. Cell Biol. 2, 268-275. 43. Axelrod, D. (2001). Total internal reflection fluorescence microscopy in cell biology. Traffic 2, 764-774. 44. Hartmann, F.J., and Bendall, S.C. (2020). Immune monitoring using mass cytometry and related high-dimensional imaging approaches. Nat. Rev. Rheumatol. 16, 87-99. 45. Robinson, J.P., Rajwa, B., Patsekin, V., and Davisson, V.J. (2012). Computational analysis of high-throughput flow cytometry data. Expert Opin Drug Discov 7, 679- 693. 46. Lu, Y., Cheng, H., Li, G.C., et al. (2022). Dynamic cryptography through plasmon‐ enhanced fluorescence blinking. Adv. Funct. Mater. 32, 2201372. 47. Zheng, Y., Jiang, C., Ng, S.H., et al. (2016). Unclonable plasmonic security labels achieved by shadow‐mask‐lithography‐assisted self‐assembly. Adv. Mater. 28, 2330-2336. 48. Su, Q., Jiang, C., Gou, D., and Long, Y. (2021). Surface plasmon-assisted fluorescence enhancing and quenching: from theory to application. ACS Appl. Bio Mater. 4, 4684-4705. 49. Zong, H., Wang, X., Mu, X., et al. (2019). Plasmon-enhanced fluorescence resonance energy transfer. Chem. Rec. 19, 818-842. 50. Min, X., Cao, B., Huang, S., et al. (2023). Bioorthogonal chemistry-based high- efficient quantum dots binding boosts the detection sensitivity of plasmon- enhanced fluorescence platform for immunoassay. Sens. Actuators, B 382, 133516. 51. Luan, J., Seth, A., Gupta, R., et al. (2020). Ultrabright fluorescent nanoscale labels for the femtomolar detection of analytes with standard bioassays. Nat. Biomed. Eng. 4, 518-530. 52. Fan, Z., Jiang, C., Wang, Y., et al. (2022). Engineered extracellular vesicles as intelligent nanosystem for next-generation of nanomedicine. Nanoscale Horiz. 7, 682-714. 53. Jiang, C., Fu, Y., Liu, G., et al. (2022). Multiplexed profiling of extracellular vesicles for biomarker development. Nano-Micro Lett. 14, 3. 54. Jiang, C., Hopfner, F., Berg, D., et al. (2021). Validation of α‐synuclein in L1CAM‐ immunocaptured exosomes as a biomarker for the stratification of Parkinsonian syndromes. Movement. Disord. 36, 2663-2669. 55. Fan, W., Ren, W., and Liu, C. (2023). Advances in optical counting and imaging of micro/nano single-entity reactors for biomolecular analysis. Anal. Bioanal. Chem. 415, 97-117. 56. Zheng, Y., Soeriyadi, A.H., Rosa, L., et al. (2015). Reversible gating of smart plasmonic molecular traps using thermoresponsive polymers for single-molecule detection. Nat. Commun. 6, 8797. 57. Zheng, Y., Rosa, L., Thai, T., et al. (2015). Asymmetric gold nanodimer arrays: electrostatic self-assembly and SERS activity. J. Mater. Chem. A 3, 240-249. 58. Lu, Y., Chen, H., Cheng, H., et al. (2022). Plasmonic physical unclonable function labels based on tricolored silver nanoparticles: implications for anticounterfeiting applications. ACS Appl. Nano Mater. 5, 9298-9305. 59. Lu, Y., Cheng, H., Francis, P.S., and Zheng, Y. (2023). Nanomaterials and artificial intelligence in anti-counterfeiting. Intelligent Nanotechnology, (Elsevier), 361-398. 60. Zhao, J., Liu, C., Li, Y., et al. (2020). Thermophoretic detection of exosomal microRNAs by nanoflares. J. Am. Chem. Soc. 142, 4996-5001. 61. Wu, X., Zhao, H., Natalia, A., et al. (2020). Exosome-templated nanoplasmonics for multiparametric molecular profiling. Sci. Adv. 6, eaba2556. 62. Lu, Y., Cheng, H., Li, G.C., et al. (2022). Dynamic cryptography through plasmon‐ enhanced fluorescence blinking. Adv. Funct. Mater. 32, 2201372, 63. Lu, Y., Chen, H., Cheng, H., et al. (2022). Plasmonic physical unclonable function labels based on tricolored silver nanoparticles: implications for anticounterfeiting applications. Adv. Funct. Mater. 5, 9298-9305. 64. Im, H., Shao, H., Park, Y.I., et al. (2014). Label-free detection and molecular profiling of exosomes with a nano-plasmonic sensor. Nat. Biotechnol. 32, 490-495. 65. Liang, K., Liu, F., Fan, J., et al. (2017). Nanoplasmonic quantification of tumour- derived extracellular vesicles in plasma microsamples for diagnosis and treatment monitoring. Nat. Biomed. Eng. 1, 0021. 66. Sriram, M., Markhali, B. P., Nicovich, P. R., et al. (2018) A rapid readout for many single plasmonic nanoparticles using dark-field microscopy and digital color 67. REVIEW 14 The Innovation Medicine 1(2): 100023, September 21, 2023 www.the-innovation.org/medicine
  • 15. analysis. Biosens. Bioelectron. 117, 530-536. Zhang, Y., Shuai, Z., Zhou, H., et al. (2018). Single-molecule analysis of microRNA and logic operations using a smart plasmonic Nanobiosensor. J. Am. Chem. Soc. 140, 3988-3993. 68. Lee, K., Fraser, K., Ghaddar, B., et al. (2018). Multiplexed profiling of single extracellular vesicles. ACS Nano 12, 494-503. 69. Craw, P., and Balachandran, W. (2012). Isothermal nucleic acid amplification technologies for point-of-care diagnostics: a critical review. Lab Chip 12, 2469- 2486. 70. Zhao, Y., Xiang, J., Cheng, H., et al. (2021). Flexible photoelectrochemical biosensor for ultrasensitive microRNA detection based on concatenated multiplex signal amplification. Biosens. Bioelectron. 194, 113581. 71. Fu, R., and Xianyu, Y. (2023). Gold nanomaterials‐ implemented CRISPR‐ Cas systems for biosensing. Small 2300057. 72. Long, Y., Zhou, X., and Xing, D. (2011). Sensitive and isothermal electrochemiluminescence gene-sensing of Listeria monocytogenes with hyperbranching rolling circle amplification technology. Biosens. Bioelectron. 26, 2897-2904. 73. Long, Y., Zhou, X., and Xing, D. (2013). An isothermal and sensitive nucleic acids assay by target sequence recycled rolling circle amplification. Biosens. Bioelectron. 46, 102-107. 74. Wu, C., Garden, P.M., and Walt, D.R. (2020). Ultrasensitive detection of attomolar protein concentrations by dropcast single molecule assays. J. Am. Chem. Soc. 142, 12314-12323. 75. Wang, W., Wu, J., Zhao, Z., et al. (2022). Ultrasensitive automatic detection of small molecules by membrane imaging of single molecule assays. ACS Appl. Mater. Interfaces 14, 54914-54923. 76. Jiang, C., Alam, M.T., Silva, S.M., et al. (2016). Unique sensing interface that allows the development of an electrochemical immunosensor for the detection of tumor necrosis factor α in whole blood. ACS Sens. 1, 1432-1438. 77. Jiang, C., Wang, G., Hein, R., et al. (2020). Antifouling strategies for selective in vitro and in vivo sensing. Chem. Rev. 120, 3852-3889. 78. Sun, J., Wang, G., Cheng, H., et al. (2022). An antifouling electrochemical aptasensor based on hyaluronic acid functionalized polydopamine for thrombin detection in human serum. Bioelectrochemistry 145, 108073. 79. Yan, H., Wen, Y., Tian, Z., et al. (2023). A one-pot isothermal Cas12-based assay for the sensitive detection of microRNAs. Nat. Biomed. Eng., 1-19. 80. Zhu, Z., Guo, Y., Wang, C., et al. (2023). An ultra-sensitive one-pot RNA-templated DNA ligation rolling circle amplification-assisted CRISPR/Cas12a detector assay for rapid detection of SARS-CoV-2. Biosens. Bioelectron. 228, 115179. 81. Wu, J., Lin, Z., Zou, Z., et al. (2022). Identifying the phenotypes of tumor-derived extracellular vesicles using size-coded affinity microbeads. J. Am. Chem. Soc. 144, 23483-23491. 82. Akama, K., Iwanaga, N., Yamawaki, K., et al. (2019). Wash- and amplification-free digital immunoassay based on single-particle motion analysis. ACS Nano 13, 13116- 13126. 83. Zhou, Y., Zhao, W., Feng, Y., et al. (2023). Artificial intelligence-assisted digital immunoassay based on a programmable-particle-decoding technique for multitarget ultrasensitive detection. Anal. Chem. 95, 1589-1598. 84. Gong, F., Yang, Y., Shan, X., et al. (2023). A microchamber-free and enzyme-free digital assay based on ultrabright fluorescent microspheres. Sens. Actuators, B 380, 133358. 85. Zhang, J., Li, Y., Chai, F., et al. (2022). Ultrasensitive point-of-care biochemical sensor based on metal-AIEgen frameworks. Sci. Adv. 8, eabo1874. 86. Wang, X., and Yan, X.P. (2018). Analyte-driven self-assembly of graphene oxide sheets onto hydroxycamptothecin-functionalized upconversion nanoparticles for the determination of type I topoisomerases in cell extracts. Anal. Bioanal. Chem. 410, 6761-6769. 87. Li, Y.F., Lin, Z.Z., Hong, C.Y., and Huang, Z.Y. (2021). Histamine detection in fish samples based on indirect competitive ELISA method using iron-cobalt co-doped carbon dots labeled histamine antibody. Food Chem. 345, 128812. 88. Yang, H., Liu, Y., Guo, Z., et al. (2019). Hydrophobic carbon dots with blue dispersed emission and red aggregation-induced emission. Nat. Commun. 10, 1789. 89. Sun, C., Liu, L., Pérez, L., et al. (2022). Droplet-microfluidics-assisted sequencing of HIV proviruses and their integration sites in cells from people on antiretroviral therapy. Nat. Biomed. Eng. 6, 1004-1012. 90. Tian, T., Shu, B., Jiang, Y., et al. (2021). An ultralocalized Cas13a assay enables universal and nucleic acid amplification-free single-molecule RNA diagnostics. ACS Nano 15, 1167-1178. 91. Mou, L., Hong, H., Xu, X., et al. (2021). Digital hybridization human papillomavirus assay with attomolar sensitivity without amplification. ACS Nano 15, 13077-13084. 92. Liu, C., Xu, X., Li, B., et al. (2018). Single-exosome-counting immunoassays for cancer diagnostics. Nano Lett. 18, 4226-4232. 93. Yelleswarapu, V., Buser, J.R., Haber, M., et al. (2019). Mobile platform for rapid sub–picogram-per-milliliter, multiplexed, digital droplet detection of proteins. PNAS 116, 4489-4495. 94. Chen, Z.P., Yang, P., Yang, Z.Z., et al. (2022). One-step digital droplet auto-catalytic nucleic acid amplification with high-throughput fluorescence imaging and droplet 95. tracking computation. Anal. Chem. 94, 9166-9175. Wang, Y., Shah, V., Lu, A., et al. (2021). Counting of enzymatically amplified affinity reactions in hydrogel particle-templated drops. Lab Chip 21. 96. Song, H., Chen, D.L., and Ismagilov, R.F. (2006). Reactions in droplets in microfluidic channels. Angew Chem Int Ed Engl 45, 7336-7356. 97. Garstecki, P., Fuerstman, M.J., Stone, H.A., et al. (2006). Formation of droplets and bubbles in a microfluidic T-junction —scaling and mechanism of break-up. Lab Chip 6, 437-446. 98. Rissin, D.M., Kan, C.W., Campbell, T.G., et al. (2010). Single-molecule enzyme-linked immunosorbent assay detects serum proteins at subfemtomolar concentrations. Nat Biotechnol 28, 595-599. 99. Leirs, K., Dal Dosso, F., Perez-Ruiz, E., et al. (2022). Bridging the gap between digital assays and point-of-care testing: automated, low cost, and ultrasensitive detection of thyroid stimulating hormone. Anal Chem 94, 8919-8927. 100. Sun, J.J., Hu, J.M., Gou, T., et al. (2019). Power-free polydimethylsiloxane femtoliter- sized arrays for bead-based digital immunoassays. Biosens. Bioelectron. 139, 111339. 101. Li, Q., Bencherif, S.A., and Su, M. (2021). Edge-enhanced microwell immunoassay for highly sensitive protein detection. Anal. Chem. 93, 10292-10300. 102. Song, Y., Zhao, J., Cai, T., et al. (2021). Machine learning-based cytokine microarray digital immunoassay analysis. Biosens. Bioelectron. 180, 113088. 103. Jeng, M.-J., Sharma, M., Sharma, L., et al. (2019). Raman spectroscopy analysis for optical diagnosis of oral cancer detection. J. Clin. Med. 8, 1313, 104. Alkhuder, K. (2023). Raman scattering-based optical sensing of chronic liver diseases. Photodiagn. Photodyn. Ther. 42, 103505. 105. Lü, G., Zheng, X., Lü, X., et al. (2021). Label-free detection of echinococcosis and liver cirrhosis based on serum Raman spectroscopy combined with multivariate analysis. Photodiagn. Photodyn. Ther. 33, 102164. 106. Wang, C., Zhang, T., Wang, P., et al. (2021). Bone metabolic biomarker-based diagnosis of type 2 diabetes osteoporosis by support vector machine. Ann. Transl. Med. 9. 107. Eke, C.S., Jammeh, E., Li, X., et al. (2020). Early detection of Alzheimer's disease with blood plasma proteins using support vector machines. IEEE J. Biomed. Health. Inf. 25, 218-226. 108. Javeed, A., Zhou, S., Yongjian, L., et al. (2019). An intelligent learning system based on random search algorithm and optimized random forest model for improved heart disease detection. IEEE Access 7, 180235-180243. 109. Wang, S., Wang, Y., Wang, D., et al. (2020). An improved random forest-based rule extraction method for breast cancer diagnosis. Appl. Soft Comput. 86, 105941. 110. Kumar, A., and Sodhi, S.S. (2020). Comparative analysis of gaussian filter, median filter and denoise autoenocoder. 2020 7th International Conference on Computing for Sustainable Global Development (INDIACom). IEEE, 45-51. 111. Noor, A., Zhao, Y., Khan, R., et al. (2020). Median filters combined with denoising convolutional neural network for Gaussian and impulse noises. Multimed. Tools. Appl. 79, 18553-18568. 112. Feng, N., Wang, S., Wei, L., et al. (2023). Artificial intelligence-based imaging transcoding system for multiplex screening of viable foodborne pathogens. Anal. Chem. 95, 8649-8659. 113. Zhang, F., Kaufman, H.L., Deng, Y., and Drabier, R. (2013). Recursive SVM biomarker selection for early detection of breast cancer in peripheral blood. BMC Med. Genomics 6, 1-10, 114. Zheng, R., Su, R., Xing, F., et al. (2022). Metabolic-dysregulation-based iEESI-MS reveals potential biomarkers associated with early-stage and progressive colorectal cancer. Anal. Chem. 94, 11821-11830. 115. Li, Y., Pan, J., Zhou, N., et al. (2021). A random forest model predicts responses to infliximab in Crohn’s disease based on clinical and serological parameters. Scand. J. Gastroenterol 56, 1030-1039. 116. Wager, S., and Athey, S. (2018). Estimation and inference of heterogeneous treatment effects using random forests. J. Am. Stat. Assoc. 113, 1228-1242. 117. Tabib, S., and Larocque, D. (2020). Non-parametric individual treatment effect estimation for survival data with random forests. Bioinform. 36, 629-636. 118. Waljee, A.K., Joyce, J.C., Wang, S., et al. (2010). Algorithms outperform metabolite tests in predicting response of patients with inflammatory bowel disease to thiopurines. CGH 8, 143-150. 119. Ellahham, S. (2020). Artificial intelligence: the future for diabetes care. AM. J. Med. 133, 895-900. 120. Blanco, F.C., Bigi, F., and Soria, M.A. (2014). Identification of potential biomarkers of disease progression in bovine tuberculosis. Vet. Immunol. Immunopathol. 160, 177- 183. 121. Rizk-Jackson, A., Stoffers, D., Sheldon, S., et al. (2011). Evaluating imaging biomarkers for neurodegeneration in pre-symptomatic Huntington's disease using machine learning techniques. NeuroImage 56, 788-796. 122. Lin, C.-H., Chiu, S.-I., Chen, T.-F., et al. (2020). Classifications of neurodegenerative disorders using a multiplex blood biomarkers-based machine learning model. Int. J. Mol. Sci. 21, 6914. 123. Ghiti Moghadam, M., Lamers-Karnebeek, F.B., Vonkeman, H.E., et al. (2018). Multi- biomarker disease activity score as a predictor of disease relapse in patients with rheumatoid arthritis stopping TNF inhibitor treatment. PLoS One 13, e0192425. 124. Medicine REVIEW The Innovation Medicine 1(2): 100023, September 21, 2023 15
  • 16. Diao, X., Li, X., Hou, S., et al. (2023). Machine learning-based label-free SERS profiling of exosomes for accurate fuzzy diagnosis of cancer and dynamic monitoring of drug therapeutic processes. Anal. Chem. 19, 7552-7559. 125. Guinney, J., Dienstmann, R., Wang, X., et al. (2015). The consensus molecular subtypes of colorectal cancer. Nat. Med. 21, 1350-1356. 126. Menyhárt, O., and Győrffy, B. (2021). Multi-omics approaches in cancer research with applications in tumor subtyping, prognosis, and diagnosis. Comput. Struct. Biotechnol. J. 19, 949-960. 127. Khater, I.M., Nabi, I.R., and Hamarneh, G. (2020). A review of super-resolution single- molecule localization microscopy cluster analysis and quantification methods. Patterns 1, 100038. 128. Scalisi, S., Pisignano, D., Cella Zanacchi, F. (2023). Single‐ molecule localization microscopy goes quantitative. Microsc Res Tech., 86(4), 494-504. 129. Mahmoudpour M, Jouyban A, Soleymani J, et al. (2022) Rational design of smart nano-platforms based on antifouling-nanomaterials toward multifunctional bioanalysis. Adv. Colloid Interface Sci. 302, 102637. 130. Dou, B., Zhu, Z., Merkurjev, E., et al. (2023). Machine learning methods for small data challenges in molecular science. Chem. Rev. 123, 8736-8780. 131. Fan, W., Dong, Y., Ren, W., and Liu, C. (2023). Single microentity analysis-based ultrasensitive bioassays: Recent advances, applications, and perspectives. TrAC, Trends Anal. Chem. 162, 117035. 132. Liang, K., Liu, F., Fan, J., et al. (2017). Nanoplasmonic quantification of tumour- derived extracellular vesicles in plasma microsamples for diagnosis and treatment monitoring. Nat. Biomed. Eng. 1, 0021. 133. Wang, F., Li, Y., Han, Y., et al. (2019). Single-particle enzyme activity assay with spectral-resolved dark-field optical microscopy. Anal. Chem. 91, 6329-6339. 134. Min, X., Cao, B., Huang, S., et al. (2023). Bioorthogonal chemistry-based high- efficient quantum dots binding boosts the detection sensitivity of plasmon- enhanced fluorescence platform for immunoassay. Sens. Actuators B: Chem. 382, 133516. 135. Cohen, L., Cui, N., Cai, Y., et al. (2020). Single molecule protein detection with attomolar sensitivity using droplet digital enzyme-linked immunosorbent assay. ACS Nano 14, 9491-9501. 136. Schmidt, K., Hageneder, S., Lechner, B., et al. (2022). Rolling circle amplification tailored for plasmonic biosensors: from ensemble to single-molecule detection. ACS Appl. Mater. Interfaces 14, 55017-55027. 137. Chen, D., Zhang, X., Zhu, L., et al. (2022). All on size-coded single bead set: a modular enrich-amplify-amplify strategy for attomolar level multi-immunoassay. Chem. Sci. 13, 3501-3506. 138. You, M., Peng, P., Xue, Z., et al. (2021). A fast and ultrasensitive ELISA based on rolling circle amplification. Analyst 146, 2871-2877. 139. Cui, J.Q., Liu, F.X., Park, H., et al. (2022). Droplet digital recombinase polymerase amplification (ddRPA) reaction unlocking via picoinjection. Biosens. Bioelectron. 202, 114019. 140. Xu, S., Wu, J., Chen, C., et al. (2021). A micro-chamber free digital biodetection method via the “sphere-labeled-sphere” strategy. Sens. Actuators B: Chem. 337, 129794. 141. Wang, F., Han, Y., Wang, S., et al. (2019). Single-particle LRET aptasensor for the sensitive detection of aflatoxin B(1) with upconversion nanoparticles. Anal. Chem. 91, 11856-11863. 142. Farka, Z., Mickert, M.J., Hlavacek, A., et al. (2017). Single molecule upconversion- linked immunosorbent assay with extended dynamic range for the sensitive detection of diagnostic biomarkers. Anal. Chem. 89, 11825-11830. 143. Guan, Z., Zou, Y., Zhang, M., et al. (2014). A highly parallel microfluidic droplet method enabling single-molecule counting for digital enzyme detection. Biomicrofluidics 8, 014110. 144. Zhou, W. (2018). Development of immunomagnetic droplet-based digital immuno- PCR for the quantification of prostate specific antigen. Anal. Methods 10, 3690- 3695. 145. Sun, J., Hu, J., Gou, T., et al. (2019). Power-free polydimethylsiloxane femtoliter- sized arrays for bead-based digital immunoassays. Biosens. Bioelectron. 139, 111339. 146. Maley, A.M., Garden, P.M., and Walt, D.R. (2020). Simplified digital enzyme-linked immunosorbent assay using tyramide signal amplification and fibrin hydrogels. ACS Sens. 5, 3037-3042. 147. Hu, J., Gou, T., Wu, W., et al. (2019). Proximity ligation assays for precise quantification of femtomolar proteins in single cells using self-priming microfluidic dPCR chip. Anal. Chim. Acta 1076, 118-124. 148. Morasso, C., Ricciardi, A., Sproviero, D., et al. (2022). Fast quantification of extracellular vesicles levels in early breast cancer patients by Single Molecule Detection Array (SiMoA). Breast Cancer Res Treat. 192, 65-74. 149. ACKNOWLEDGMENTS The work is supported by the Major Research Plan of the National Natural Science Foundation of China (Grant No.92259102), Dr Cheng Jiang appreciates the financial support from the Chinese University of Hong Kong (Shenzhen) startup funding (K10120220253), Shenzhen-Hong Kong Cooperation Zone for Technology and Innova- tion (HZQBKCZYB-2020056), Shenzhen Science and Technology Program (2022A1515110206), Shenzhen International Science and Technology Cooperation Project (GJHZ20220913144002005). AUTHOR CONTRIBUTIONS C.J., K.G. and C.X. supervised and revised the manuscript. W.Z. and Y.L. wrote and edited the manuscript. All authors contributed to the article and approved the submitted version. DECLARATION OF INTERESTS The authors declare no competing interests. LEAD CONTACT WEBSITE https://lhs.cuhk.edu.cn/en/teacher/326 REVIEW 16 The Innovation Medicine 1(2): 100023, September 21, 2023 www.the-innovation.org/medicine