You are on page 1of 54

Review

pubs.acs.org/CR

SERS-Activated Platforms for Immunoassay: Probes, Encoding


Methods, and Applications
Zhuyuan Wang, Shenfei Zong, Lei Wu, Dan Zhu, and Yiping Cui*
Advanced Photonics Center, Southeast University, Nanjing 210096, Jiangsu, China

ABSTRACT: Owing to their excellent multiplexing ability, high sensitivity, and large
dynamic range, immunoassays using surface-enhanced Raman scattering (SERS) as the
readout signal have found prosperous applications in fields such as disease diagnosis,
See https://pubs.acs.org/sharingguidelines for options on how to legitimately share published articles.

environmental surveillance, and food safety supervision. Various ever-increasing


demands have promoted SERS-based immunoassays from the classical sandwich-type
ones to those integrated with fascinating automatic platforms (e.g., test strips and
Downloaded via CHUNGNAM NATL UNIV on March 11, 2022 at 08:05:10 (UTC).

microfluidic chips). As recent years have witnessed impressive progress in SERS


immunoassays, we try to comprehensively cover SERS-based immunoassays from their
basic working principles to specific applications. Focusing on several basic elements in
SERS immunoassays, typical structures of SERS nanoprobes, productive optical spectral
encoding strategies, and popular immunoassay platforms are highlighted, followed by their representative biological applications
in the last 5 years. Moreover, despite the vast advances achieved to date, SERS immunoassays still suffer from some annoying
shortcomings. Thus, proposals on how to improve the SERS immunoassay performance are also discussed, as well as future
challenges and perspectives, aiming to give brief and valid guidelines for choosing suitable platforms according to particular
applications.

CONTENTS 4. SERS Encoding for Multiplex Detection 7926


4.1. Design and Fabrication of SERS Encoders 7926
1. Introduction 7911 4.2. SERS Spectral Encoding Strategy 7928
1.1. Historical View of Immunoassay Protocols 7911 4.2.1. Raman-Frequency-Based Encoding 7928
1.2. SERS-Based Immunoassay (History, Design, 4.2.2. Signal-Intensity-Based Encoding 7928
and Advantages) 7912 4.3. SERS-Included Joint Encoding 7929
2. Principles of SERS-Based Immunoassay 7912 4.3.1. SERS−Fluorescence Joint Spectral En-
2.1. Construction of SERS-Based Immunoassays 7912 coding 7929
2.2. Design and Fabrication of SERS Probes 7915 4.3.2. Spectral−Spatial Joint Encoding 7930
2.2.1. Optical Label: Organic and Inorganic 5. Applications of SERS-Based Immunoassay 7930
Raman Reporters 7915 5.1. Analysis of IgGs 7931
2.2.2. Signal Amplifiers: Metal Nanoparticles 7916 5.2. Study of Disease Biomarkers 7931
2.2.3. Protection Layer: Silica, Polymers, Etc 7917 5.2.1. Detection of Protein Biomarkers 7932
2.2.4. Targeting Molecules: Antibodies and 5.2.2. Detection of miRNAs and Circulating
Aptamers 7917 DNAs 7934
3. Platforms for SERS-Based Immunoassay 7917 5.2.3. Detection of Telomerase Activity and
3.1. SERS-Based Immunoassay on Solid Sub- Telomere Length Measurement 7935
strate 7918 5.3. Identification of Bacteria and Viruses 7936
3.1.1. Nonmetallic Substrate 7918 5.4. Detection of Ions and Toxins 7936
3.1.2. Metallic Substrate 7918 5.5. Sorting of Cells 7938
3.2. SERS-Based Immunoassay in Liquid Phase 5.6. Multiplex Biochemical Analysis Using SERS
(Suspension Array) 7919 Encoders 7939
3.2.1. Nonmagnetic Substrate 7920 5.6.1. Multiplex Immuno-Detection of Biomo-
3.2.2. Magnetic Substrate 7921 lecules and Cells 7939
3.3. Immunoassays on a SERS−Microfluidic Chip 7922 5.6.2. Multiplex Ion Detection 7943
3.3.1. Microfluidic Channels 7922 6. Challenges and Perspectives 7943
3.3.2. Droplet Microfluidics 7923 6.1. Improving the Reproducibility of SERS-
3.4. SERS-Based Immunoassay on Optical Fibers 7924 Based Immunoassay 7943
3.5. Paper-Based SERS Platforms 7924 6.1.1. Uniformity of the Immune Substrates 7943
3.6. Hydrophobic SERS Substrates 7925
3.7. Comparison between Different Platforms for
SERS-Based Immunoassay 7926 Received: January 12, 2017
Published: May 23, 2017

© 2017 American Chemical Society 7910 DOI: 10.1021/acs.chemrev.7b00027


Chem. Rev. 2017, 117, 7910−7963
Chemical Reviews Review

6.1.2. Stability of SERS Nanoprobes 7945 SERS-based immunoassays. The design and fabrication of SERS
6.2. Real Sample Detection 7945 nanoprobes are introduced in section 2. As this part has been
6.2.1. Serum and Plasma 7945 extensively reviewed previously,7,12−14 we briefly summarize the
6.2.2. Whole Blood 7946 typical design and structure of a SERS nanoprobe. In section 3,
6.2.3. Other 7947 different platforms in SERS-based immunoassay are summar-
6.3. Point-of-Care Testing (POCT) 7947 ized, ranging from solid substrates to liquid microspheres to
6.4. Practical SERS-Encoding Technique 7947 microfluidic chips, etc. The advantages and disadvantages of
6.5. Combining SERS with Other Techniques for each platform will be compared and discussed, which is
Multimodal Immunoassay 7948 expected to provide a brief guideline for choosing a suitable
6.5.1. SERS−SPR Immunoassay 7948 platform for specific applications. Aside from the nanoprobes
6.5.2. SERS−Fluorescence Immunoassay 7948 and platforms, spectral-encoding techniques are usally
6.5.3. SERS−Electrochemistry Immunoassay 7949 employed in an immunoassay system for multiplex detec-
6.5.4. Immunoassay with Hyperbolic Metama- tion.15−17 Therefore, in section 4, the typical SERS-encoding
terials 7949 techniques are summarized, including those based on Raman
7. Conclusion 7949 frequency or intensity, as well as several SERS-involved
Author Information 7949 multimodal-encoding strategies. We then provide considerable
Corresponding Author 7949 details on the biochemical applications of SERS-based
ORCID 7949 immunoassay in analyzing various targets in section 5. Finally,
Notes 7950 the challenges and future perspective of designing next-
Biographies 7950 gerneration SERS immunoassays are presented in sections 6
Acknowledgments 7950 and 7.
Abbreviations Used 7950
1.1. Historical View of Immunoassay Protocols
References 7950
An immunoassay is defined as a test for the quantitative
determination of chemical substances, such as hormones, drugs,
1. INTRODUCTION and specific proteins, that utilizes the highly specific binding
Immunoassay is one of the most famous tools for the between antibodies and antigens or haptens.18,19
quantitative detection of biochemical targets (e.g., proteins The development of immunoassay starts from the detection
and toxins). The fine accuracy and operability of immunoassays of insulin by Yalow and Berson.20 They found out that the
have promoted thorough investigations of stringent issues, such globulins bound to insulin in patients treated with exogenous
as disease diagnosis, food safety, and environmental protection. animal insulin were actually antibodies. Afterward, insulin
Recent years have witnessed significant progress in developing antibodies were purified and the first radio-immunoassay (RIA)
more efficient immunoassay protocols. Immunoassays have was established to detect insulin using a radioactive iodine
evolved from the traditional ones performed on microplates to label.21 Later, together with two other scientists, Yalow won the
those conducted with highly automized platforms using various Nobel Prize in Medicine/Physiology due to her pioneering
kinds of readout signals (e.g., colorimetry, electrochemistry, work on devising RIA. Since its discovery, RIA has paved the
surface plasmon resonance, fluorescence, and surface-enhanced way for the rapid development of immunoassays.
Raman scattering). To date, immunoassay is no longer In the late 1960s, immunoassay techniques were developed
regarded as just a simple test. It is more appropriate to for chemically linking enzymes to antibodies. Soon after that, in
consider immunoassay as a sophisticated analyte-detecting 1971, the enzyme immunoassay (EIA) and enzyme-linked
system composed of both biochemical and electrical or optical immunosorbent assay (ELISA) were developed by Engvall and
techniques. Since other immunoassay strategies have been well- Perlmann22 and van Weeman and Schuurs,23 respectively. The
reviewed elsewhere,1−3 here we only focus on optical replacement of radiolabels with enzymes made immunoassay
immunoassays, especially those using surface-enhanced much simpler and more popular. Today, most of the detection
Raman scattering (SERS) as the readout signal. SERS is well- of clinical protein biomarkers is performed with ELISA kits.
known for its high sensitivity, fine specificity, and excellent Since the 1970s, various detection techniques have been
multiplexing ability, leading to the blooming use of SERS-based employed to develop the formats of immunoassays. For
immunoassays.4−9 The generation of Raman enhancement example, immunoassays by electrochemical techniques can
needs a nanoscopic metallic surface;10,11 consequently, SERS- determine the level of analytes by the changes in electrical
based immunoassays have some special requirements on the signals such as potential, current, resistance, and capacitance.24
layout of the assay. SERS immunoassays usually contain several As a label-free method, electrochemical immune detection
basic elements (i.e., nanoprobes, SERS spectra, and assay offers the advantages of fast response, low cost, and simplicity,
platforms), each of which requires an elaborate design. Despite which has a wide range of applications in biosensing. On the
its rapid development, up to now, there is no review covering other hand, with the rapid development of optical materials and
SERS immunoassay in detail. methods, the optical detection technique has become one of
Hence, in this review, we try to comprehensively cover the most popular methods for immunoassays in recent years. In
SERS-based immunoassays from basic principles to abundant the beginning, chemiluminescence immunosensors have been
applications. Typical structures of SERS nanoprobes, produc- employed for routine clinical diagnosis or biomedical research
tive optical-encoding strategies, and popular immunoassay due to their advantages such as no radioactive waste, the
platforms are highlighted, followed by the introduction of relatively simple instrumentation, and high sensitivity. In
representative biological applications using SERS immuno- principle, they employ chemical reactions to produce optical
assays. Specifically, we start with the history, principles, and signals for immunoassays.48 Later, fluorescence labels, including
classifications of immunoassays in section 1. Then, we focus on fluorescent dyes and quantum dots, were used to develop
7911 DOI: 10.1021/acs.chemrev.7b00027
Chem. Rev. 2017, 117, 7910−7963
Chemical Reviews Review

optical immunoassays.49,50 The advantages of fluorescence screening, and environmental monitoring.9 Additionally and
immunoassays include simplicity of system design, improved importantly, the SERS-based optical-encoding technique has
sensitivity, and fast readout process. More recently, immuno- been well-employed for high-throughput detection, in which
assays based on SERS, which utilize Raman reporters as the multiple targets could be distinguished simultaneously by the
optical labels, have attracted much attention due to their Raman frequency or intensity.65 Further, SERS has also been
ultrahigh sensitivity, excellent multiplexing ability, and high combined with fluorescence17 or spatial coordinate66 for joint
photostability.8 Aside from these labeled optical techniques, encoding. The combination of SERS with other techniques
surface plasmon resonance (SPR) immunoassays provide a greatly improved the encoding capacity, which is of great
label-free optical sensing platform,51 in which the specific importance for achieving high-throughput analysis.
interactions between antibodies and antigens could be In brief, SERS-based immunoassays mainly have three
determined by the changes in reflective index in real time. unique advantages. First, due to the large enhancement factor
Another kind of immunosensor employs the quartz crystal up to ∼1010, SERS-based detection could reach a high
microbalance (QCM) technique, which measures the mass sensitivity to a single molecule level, making it extremely useful
change through a quartz crystal resonator,52 to perform the for trace analysis.67−69 Second, the narrow Raman bands could
label-free, noninvasive, and real-time detection. Summarily, the be employed for highly efficient spectroscopic encoding, which
diversity of detection techniques makes it easy and flexible to can facilitate high-throughput detection.9,70 Third, SERS is
design efficient immunoassays. Table 1 shows some typical insensitive to photobleaching and quenching.71 The high
examples of immunoassays using the above-mentioned stability of SERS signals allows repetitive signal measurements
techniques, in which the comparisons among different to reduce test errors. Owing to these advantages, SERS-based
techniques are also summarized. immunoassays have found an enormous number of usages in
both research and practical applications.
1.2. SERS-Based Immunoassay (History, Design, and
Advantages)
2. PRINCIPLES OF SERS-BASED IMMUNOASSAY
Among the various detection techniques employed for
immunoassays, SERS has gained increasing popularity in recent Typically, the development of SERS-based immunoassay
decades. In this review, we specially focus on the development requires two critical components: the immune substrate and
of immunoassays based on SERS technique. SERS-based the SERS immunoprobe. Basically, the immune substrates are
immunoassays refer to those that using surface-enhanced modified with targeting molecules (antibodies, aptamers, etc.)
Raman scattering as the readout signal. As is well-known, to capture the specific analytes from the samples. Afterward, the
Raman scattering is the inelastic scattering of a photon, which SERS probes are employed to quantify the concentration of
was discovered by Raman and Krishnan in liquids53 and by analytes. There are two basic functions of SERS probes: (1)
Landsberg and Mandelstam in crystals,54 respectively. Raman specifically recognizing and binding to the analytes captured by
scattering can provide rich structural and quantitative the immune substrate and (2) providing SERS signals for the
information on nearly all kinds of molecules and materials. quantitative detection of analytes. In accordance with the
However, the weak intensity of Raman spectroscopy limits its universal classification for immunoassays, generally, SERS-
application in biomedical and chemical studies. In 1973, based immunoassays could be classified into the heterogeneous
Fleischmann et al. discovered that pyridine molecules adsorbed assays and homogeneous ones, noncompetitive assays and
competitive ones, and labeled and label-free ones. In this
onto the rough surface of silver exhibited a dramatically
section, we first introduce the basic principles of SERS-based
enhanced Raman scattering light signal.55 The enhancement
immunoassay in different formats. Then, the design and
factor could reach from 103 to 1010,56−58 which has then been
fabrication of SERS probes are summarized. The discussion
widely used for unltrasensitive biochemical analysis.
on different immune substrates (platforms) is concretely
The first typical SERS-based immunoassay was developed in
described in section 3.
1999 using a “sandwich” scheme (Figure 1).59 In that protocol,
capture antibodies are bound to a flat gold surface to form an 2.1. Construction of SERS-Based Immunoassays
immune substrate. Gold nanoparticles were labeled with Raman According to the different separation and detection processes,
reporters and conjugated with detection antibodies to form SERS-based immunoassays could be classified into heteroge-
SERS probes. Then, the target antigens could be captured by neous and homogeneous immunoassays (Figure 2). The
the antibodies on the substrate and subsequently be recognized heterogeneous immunoassay requires the separation of bound
by the SERS probes. The presence and concentration of a SERS probes from free ones and the concentration of antigens
specific antigen is determined by the characteristic SERS is measured through the bound SERS probes. The separation
spectrum of the Raman reporters. Importantly, the authors also step can be fulfilled using solid substrates or liquid microbeads.
demonstrated that the method could be used for multiplex On the contrary, in homogeneous SERS immunoassays, the
detection through the SERS-encoding method. binding between SERS nanoprobes and target molecules could
In the following decades, the platforms for SERS-based induce the changes in SERS intensity, which are used for the
immunoassay have experienced a rapid development, ranging qualitive and quantitative analysis. In general, the homogeneous
from solid substrates (metallic59 and nonmetallic60) to liquid immunoassay is rapid, straightforward, and more adaptable to
phase (magnetic61 and nonmagnetic62) to microfluidic chips,63 automated systems. However, it is also more vulnerable toward
paper devices,64 and optical fibers.28 The diversity of SERS- nonspecific antibody−antigen cross-reactions. In contrast,
active platforms offers sufficient choices for analyzing proteins, heterogeneous immunoassay is excellent in detection perform-
disease biomarkers, ions, toxins, bacteria, viruses, cells, etc. On ance, including sensitivity and reproducibility, although it
the other hand, there is a growing demand for multiplex, high- requires repetitive separation procedures.
throughput analysis of large quantities of analytes in a single When considering the binding manner of the immunocom-
sample, especially in the fields of clinical diagnosis, drug plex, the SERS-based immunoassays can be classified into
7912 DOI: 10.1021/acs.chemrev.7b00027
Chem. Rev. 2017, 117, 7910−7963
Table 1. Some Typical Examples of Immunoassays Using Different Techniquesa
label- operation read-out
methods analyte dynamic range limit of detection free time read-out speed instrument multiplexing ability ref
electrochemistry CEA 10.0 pg/mL to 50.0 ng/mL 4.4 ± 0.1 pg/mL no real-time yes parallel multiplexing 25
PSA 500 pg/mL to ca. 100 ng/mL; in 0.5 ng/mL; in serum, 1 ng/mL yes <30 min yes 26
serum, 1−100 ng/mL
Chemical Reviews

H1N1 split influenza vaccine 5−500 ng/mL 5 ng/mL yes <30 min yes 26
simultaneous detection of 500 pg/mL to ca. 10 μg/mL; 1 ng/mL 500 pg/mL; 1 ng/mL yes <30 min yes 26
AFP/ PSA to ca. 10 μg/mL
CA 125 0−0.1 U/mL 0.0016 U/mL yes yes 27
AFP 10.0 pg/mL to 10.0 ng/mL 3 pg/mL yes yes 28

QCM HIgG 5.0 pg/mL to 20.0 ng/mL 5.0 ± 0.18 pg/mL no >1 h real-time yes parallel multiplexing 29
thrombomodulin 10−5000 ng/mL 2 ng/mL yes >1 h yes 30

chemiluminescence CRP 1−1000 ng/mL 0.93 ng/mL no real-time/a few yes parallel multiplexing 31
AFP 1.0 × 10−3−100 ng/mL 2.7 × 10−5 ng/mL no >1 h seconds yes 32
β-hCG 2.0 × 10−4−20 IU/L 1.1 × 10−5 IU/L no >1 h yes
CA125 (1.0 × 10−4)−10 U/mL 1.7 × 10−5 U/mL no >1 h yes
CEA (1.0 × 10−4)−10 ng/mL 2.0 × 10−5 ng/mL no >1 h yes
human prealbumin 0.05−1000 μg/L 0.01 μg/L no 35 min yes 33

fluorescence NSE 75.0 pM no 45 min real-time/a few yes simultaneous and 34

7913
CEA 0.5 pM no 45 min seconds yes parallel multiplexing
Cyfra21-1 14.3 pM no 45 min yes
SCC 6.7 pM no 45 min yes
CA15-3 0.03 U/mL no 45 min yes
PSA 1.6 ng/mL no yes 35
EGFR 0.12 nM or 23 ng/mL in buffer, 0.18 no 1h yes 36
nM or 34 ng/mL in serum

colorimetry pathogen of Salmonella, 6.7 aM no real-time no parallel multiplexing 37


Listeria, and E. coli O157
IgG/hepatitis B virus IgG, 1 ng/mL to 10 μg/mL; HBsAg, IgG, 1.6 ng/mL; HBsAg, 1.3 ng/mL no 1h no 38
0.34 ng/mL to 340 μg/mL

SERS AFP, GPC3 0.1 pM no 4h 10−60s (usual yes simultaneous and 39


EGFR 1 ng/mL cetuximab no <1 h acquisition time) yes parallel multiplexing 40
SEB 2−100 pg/mL 1.3 pg/mL no 1h yes 41
NT-proBNP 1 fg/mL to 1 ng/mL 0.75 fg/mL no >1 h yes 42

SPR lysozyme dimer 1.7−35 nM 1.4 nM yes 10 min real-time yes parallel multiplexing 43
gluten peptides 1.12−19.2 ng/mL 0.33 ng/mL yes 20 min yes 44
CA125 0.26 U/mL 1h yes 45
β2-M 0.55 ng/mL 1h yes 45
ApoA1 7.7 ng/mL 1h yes 45
Review

DOI: 10.1021/acs.chemrev.7b00027
Chem. Rev. 2017, 117, 7910−7963
Chemical Reviews Review

staphylococcal enterotoxin B (SEB), N-terminal pro-brain natriuretic peptide (NT-proBNT), β2-microglobulin (β2-M), apo-lipoprotein A1(ApoA-I), Plasmodium falciparum histidine-rich protein 2 (Pf
antigen (CA15-3), α-fetoprotein (AFP), C-reactive protein (CRP), human chorionic gonadotrophin-β (β-hCG), carcinoma antigen 125 (CA125), hepatitis B virus (HBsAg), glypican-3 (GPC3),
Carcinoembryonic antigen (CEA), epidermal growth factor receptor (EGFR), prostate specific antigen (PSA), neuron-specific enolase (NSE), squamous cell carcinoma antigen (SCC), carbohydrate
ref

46
47
multiplexing ability

parallel multiplexing
instrument
read-out

yes
yes
read-out speed

real time/a few


seconds
operation
time

>2 h
>2 h
label-
free

no
no

Figure 1. Schematic illustration of the SERS-based immunoassay


developed by Ni et al. in 1999. Reprinted with permission from ref 59.
Copyright 1999 American Chemical Society.
limit of detection

competitive and noncompetitive ones (the latter is also known


as the “sandwich” or “two-site” immunoassays) (Figure 3).
Typical noncompetitive immunoassays are performed using a
sandwich structure, in which the targets are sandwiched
between the capture antibodies and the SERS probes. Thus,
4 ng/mL

the concentration of targets is quantified by the SERS probes


2.5 pM

bound to the detection antibodies, which is positively


correlated to the intensity of the readout signal. However, in
competitive SERS immunoassays, the antigens are labeled to
act as the immunoprobes, in which the targets compete with
the SERS probes for binding to the antibodies. Hence, the
intensity of detected signal is negatively correlated to the
dynamic range

concentration of the targets. Generally, the competitive


immunoassays are suitable for the detection of small molecules
with only one binding site to antibodies (such as peptides, ions,
10−1000 ng/mL
0.001−70 nM

and hormones), while noncompetitive immunoassays are


usually employed for analyzing macromolecules (e.g., proteins)
with at least two binding sites for antibodies. Compared to the
competitive immunoassays, the noncompetitive ones usually
perform better in consideration of sensing sensitivity, dynamic
range, and reproducibility.
On the other hand, both the labeled and label-free SERS
techniques could be used to develop immunoassays (Figure 4).
analyte

While labeled SERS immunoassays require the Raman reporter-


labeled immunoprobes to detect the target molecules, label-free
vancomycin

SERS immunoassays directly measure the intrinsic Raman


Pf HRP2

spectra of the target analytes. Compared to labeled SERS


immunoassays, label-free SERS detection greatly simplifies the
Table 1. continued

detection procedures. However, it requires highly efficient


SERS substrates and strong nanoparticle−molecule binding.
One major obstacle is that not all kinds of analytes could be
methods

discriminated from their intrinsic SERS spectra. It is not easy to


analyze chemicals with a small scattering cross-section. On the
HRP2).
ELISA

contrary, labeled SERS immunoassay is a universal technique


that could be used to analyze almost all kinds of analytes. The
a

7914 DOI: 10.1021/acs.chemrev.7b00027


Chem. Rev. 2017, 117, 7910−7963
Chemical Reviews Review

Figure 2. Principles of the heterogeneous and homogeneous SERS immunoassay.

Figure 3. Principles of noncompetitive and competitive SERS immunoassay.

high sensitivity and standardized protocol make it popular in nanoparticles needs to be carefully designed, and Raman
clinical diagnosis, biochemical analysis, and environmental reporters with intrinsic high-scattering cross sections are usually
monitoring. selected as the labels. Additionally, a protection layer wrapped
2.2. Design and Fabrication of SERS Probes around the metal nanoparticles is often introduced to avoid the
In a SERS-based immunoassay, the immunoprobes play a loss of Raman reporters, which also facilitates the conjugation
critical role in specific and quantitative detection. Thus, the of targeting molecules. Then, the analytes can be specifically
structure of SERS nanoprobes should be carefully designed to recognized by the targeting molecules. Generally, the aim for
improve the performance of immunoassays. Typical SERS designing SERS probes is to improve their sensitivity, chemical
nanoprobes contain four main parts: metal nanoparticles, and physical stability, and targeting efficiency, as well as their
Raman reporters, protection layers, and targeting molecules multiplexing ability. Since there are already some wonderful
(Figure 5). In such nanoprobes, the Raman signal could be reviews focusing on the design and fabrication of SERS
enhanced by attaching the reporter molecules to the surfaces of probes,7,72 we will only briefly provide some basic points here.
metal nanoparticles. To obtain a large enhancement factor and 2.2.1. Optical Label: Organic and Inorganic Raman
high stability of Raman signals, the structure of metal Reporters. Raman reporters act as the signal source in SERS
7915 DOI: 10.1021/acs.chemrev.7b00027
Chem. Rev. 2017, 117, 7910−7963
Chemical Reviews Review

Figure 4. Principles of label-free SERS detections: (a) adsorption-mediated label-free SERS detection and (b) antibody/ligand-mediated label-free
SERS detection.

Figure 5. General procedure for the fabrication of a typical SERS probe.

probes. There are several general requirements for selecting the nanoprisms could further enhance SERS signals. In addition,
reporter molecules: (1) The reporter molecules can be easily it has also been reported that closely spaced nanowires could
and strongly anchored to the surfaces of metal nanoparticles. also generate hot spots, resulting in an intense SERS
(2) The scattering cross-section of the reporters should be enhancement. In addition to selecting metal nanoparticles
intrinsically high enough to obtain a strong intensity of Raman with different shapes, another important method to improve
signals. (3) Their Raman spectra should have few characteristic the SERS activity is to create “hot spots” through controlled
peaks, which can minimize the band overlap with other aggregation of nanoparticles.83,84 Previously, we have fabricated
reporters in multiplex detection. Up to now, various molecules a kind of SERS probe based on aggregated Au nanoparticles
have served as Raman reporters, ranging from small organic with greatly enhanced SERS signals, in which the Raman
molecules [such as 4-mercaptobenzoic acid (4MBA)73 and molecules were used to induce the aggregation of gold
rhodamine 6G (R6G)74] to inorganic materials (such as nanoparticles. Afterward, a layer of silica shell was coated
graphene oxide75 and carbon nanotube76). The diversity of onto the Au aggregates to improve the stability of the
Raman spectra offers wide choices for fabricating SERS nanoaggregates. Alternatively, SERS hot spots could be
nanoprobes. generated through the formation of “nanogaps”.85 These
2.2.2. Signal Amplifiers: Metal Nanoparticles. As is nanogaps can be created as nanosphere dimers,86 nanorod
well-known, metal nanoparticles can enhance Raman signals of dimers,87 nanowire dimers,88 nanostar dimers,89 nanosphere
reporter molecules through the local electromagnetic field and trimers,90 nanopheres/nanodisk trimers,91 etc. The greatly
electron transfer. Traditionally, Au or Ag nanopheres are the enhanced local electromagnetic field in the nanogap leads to
most commonly used SERS substrates.77,78 In recent decades, strongly enhanced Raman signals. Especially, several groups
metal nanoparticles with different sizes and shapes were have reported the fabrication of SERS nanoparticles containing
synthesized. Technologies have been developed for the interior nanogaps.92−94 For example, Lim et al. presented a type
fabrication of nanorods,79 nanowires,80 nanostars,81 and of DNA-tailorable nanoparticle with interior nanogaps. This
nanoprisms,82 which show a higher SERS enhancement factor kind of nanoparticle with an excellent SERS enhancement is
compared to that of the nanospheres. Unlike nanospheres with able to provide reproducible SERS signals.94 Recently, core−
only one SPR band, the nanorods show a longitudinal SPR shell-type metal nanoparticles have also been widely used for
band and a transverse SPR band. The tunable longitudinal SPR designing SERS probes. In one of our previous works, Au@Ag
band and strong enhancement at the tips of the nanorods make nanorods (NRs) were used to fabricate SERS nanoprobes for
them one of the excellent candidates for SERS applications. immunoassays.17,73 By combining the high stability of gold and
Besides, the multibranched metal nanostars and nanoprisms the large enhancement factor of silver, Au@Ag NRs were found
have also attracted much attention as SERS substrates due to to be very useful in the applications of SERS-based immuno-
the following two reasons. One is that they own increased assay,61,95 cell imaging,96 label-free chemical detection,79,97 and
surface-to-volume ratios, allowing for more Raman reporters to drug delivery.98 More recently, graphene-enhanced Raman
attach onto their surfaces. The other reason is that the scattering (GERS) using graphene as the substrate for Raman
hybridization of the individual tips of the nanostars or enhancement has been presented.99 The novel enhancing
7916 DOI: 10.1021/acs.chemrev.7b00027
Chem. Rev. 2017, 117, 7910−7963
Chemical Reviews Review

Figure 6. (a) High-throughput protein detection on glass slides using OMP NPs. Reprinted with permission from ref 17. Copyright 2012 American
Chemical Society. (b) Multiplex DNA detection on glass slides using encoded SERS nanoprobes. Reprinted with permission from ref 60. Copyright
2012 Royal Society of Chemistry. (c) Multiplex detection of serological cancer markers on a quartz array. Reprinted with permission from ref 112.
Copyright 2015 Royal Society of Chemistry. (d) Glycoprotein detection on an array of boronate-affinity molecularly imprinted polymers (MIPs)
using SERS probes. Reprinted with permission from ref 113. Copyright 2014 Wiley-VCH Verlag GmbH & Co. KGaA.

substrate could produce clean, reproducible, and increased molecules specifically. Among these biomolecules, antibodies
Raman signals. Compared to conventional Raman enhance- are the most commonly used ones to recognize specific
ment techniques, GERS mainly relies on a chemical mechanism antigens. The purity, affinity, and orientation of antibodies
and therefore shows unique molecular selectivity. It has been attached to the nanoparticles significantly influence the
employed for analytical applications from graphene-veiled targeting efficiency. Even though antibodies are relatively
SERS to graphene-based SERS for quantitative analysis.100 expensive and not quite stable, the mature fabrication and
2.2.3. Protection Layer: Silica, Polymers, Etc. A large
modification technique still make them the most popular
percentage of SERS probes contain one or more protection
layers, which aim to protect Raman reporters from the outside targeting molecules used in SERS probes.
environment and provide anchor spots for targeting molecules. Except antibodies, aptamers have become attractive mole-
Silica,61 titanium dioxide,101 polymers [such as polyethylene cules in recent years, due to the properties such as high affinity
glycol, poly(acrylic acid), polyallylamine hydrochlor- and specificity to their targets, easy chemical synthesis and
ide],73,102,103 mesoporous silica,98 and liposomes104 are modification, and rapid tissue penetration. Aptamers are
commonly introduced materials to form the protection layers, peptides or oligonucleotides that can bind to their specific
which improve the stability of SERS probes. In 2010, Tian’s molecules by folding into distinct secondary and tertiary
group developed a novel SERS method named shell-isolated structures. The aptamer is usually selected from a large pool of
nanoparticle-enhanced Raman spectroscopy, in which the random sequences. Although aptamers generally have a lower
Raman signal amplification was provided by gold nanoparticles affinity to target molecules as compared to antibodies, the high
coated with an ultrathin silica or alumina shell.105 The use of stability, low cost, high reproducibility, and small molecular
the shell coating around the Au nanoparticle protects the SERS-
weight still make them an excellent alternative to antibod-
active nanostructure from contacting with whatever is being
probed and serves as “smart dust” over the surface that is to be ies.107−109
probed. Recently, Jiang et al. synthesized a Ag@mesoporous
silica (MSiO2)@Ag core−shell architecture,106 in which the 3. PLATFORMS FOR SERS-BASED IMMUNOASSAY
silica shell improved the stability while the inner Ag core and SERS-based immunoassay has been performed on various
outer Ag shell can form SERS hot spots. Here, the silica shell platforms, ranging from solid substrates to liquid microbeads,
not only serves as a protection layer but also acts as a spacer to
microfluidic chips, optical fibers, papers, optical waveguides, etc.
control the plasmonic coupling effects for an improved SERS
enhancement. Each of these platforms has its own characteristics regarding the
2.2.4. Targeting Molecules: Antibodies and Aptamers. fabrication, human operation, detection performance, and
In general, the specificity of SERS-based immunoassay is overall costs. In this section, the design, fabrication, and
achieved through the specific antibody (or aptamer)−antigen characteristics of each platform are highlighted, together with
interactions. Hence, SERS probes should be conjugated with the comparison and discussion of the advantages and
antibodies, peptides, or aptamers for binding to the target disadvantages of these platforms.
7917 DOI: 10.1021/acs.chemrev.7b00027
Chem. Rev. 2017, 117, 7910−7963
Chemical Reviews Review

Figure 7. (a) Tuberculosis antigen detection on 1D magnetic plasmonic nanoparticles substrate using grapheme quantum dots. Reprinted with
permission from ref 115. Copyright 2015 American Chemical Society. (b) Multiplex Raman shift immunoassay on silver nanofilms using
microcontact printing. Reprinted with permission from ref 117. Copyright 2016 American Chemical Society.

3.1. SERS-Based Immunoassay on Solid Substrate DNA detection with Au@Ag nanoparticles on DNA-attached
In the early years, SERS-based immunoassays were usually glass slides (Figure 6b).60 The benefits of using glass slides as
performed on solid surfaces, including metallic and nonmetallic the immune substrates include facile preparation, high
substrates. In general, antibodies or aptamers are first uniformity, and low cost. Alternatively, Zhang and Du used a
immobilized onto the solid substrates through chemical quartz slide to develop an immunoassay with carbon-nanodot-
bonding. Then the target molecules are captured by the decorated Ag@SiO2 SERS probes.110 Li et al. performed a
antibodies or aptamers on the substrate. Finally, SERS SERS-based sandwich immunoassay on a quartz array to
nanoprobes are used for a quantitative analysis of the analytes achieve simultaneous detection of multiple serological cancer
captured by the substrate. The solid substrates serve as sensing markers in real serum samples (Figure 6c).112 Another high-
platforms for specific biomolecular interactions, target analyte throughput solid substrate for immunoassays is commercially
separation, and signal transduction. available multiwell plates. Liang et al. employed 96-well
3.1.1. Nonmetallic Substrate. Nonmetallic immune polystyrene plates to create a novel platform for high-
substrates include glass slides,60 quartz slides,110 multiwell throughput analysis, in which Raman scattering light was
plates,111 etc. These commercially available solid substrates used as the signal-generating system of an ELISA.111
could be modified with antibodies or aptamers and used as the Interestingly, Ye et al. developed an array of boronate-affinity
immune substrates. Although most of them are flat substrates molecularly imprinted polymers (MIPs) for the determination
without special nanostructures, their ease of use still makes of glycoproteins with a sandwich structure (Figure 6d).113 The
them very popular among researchers, medical doctors, and MIP ensures a high specificity while the SERS probes improve
scientists. the sensitivity. The boronate-affinity SERS assay exhibited
Among the above-mentioned substrates, chemical-modified significant advantages over a conventional immunoassay in
glass slides are one of the most popular substrates for antibody terms of cost efficiency, stability, and speed. The method can be
immobilization. Previously, our group has performed SERS- easily extended to the analyses of other glycoprotein
based immunoassays on glass slides using SERS nanoprobes biomarkers.
composed of different kinds of nanoparticles, including gold@ 3.1.2. Metallic Substrate. Except the above-mentioned
silver nanorods,73 and organic−metal−quantum dots compo- nonmetallic substrates, metallic substrates have also been
site nanoparticles (Figure 6a).17 Li et al. realized multiplex employed widely for SERS-based immunoassays, in which the
7918 DOI: 10.1021/acs.chemrev.7b00027
Chem. Rev. 2017, 117, 7910−7963
Chemical Reviews Review

Figure 8. (a) Homogeneous immunoassay on 3D ordered silver nanoshell silica photonic crystal beads using SERS nanotags. Reprinted with
permission from ref 125. Copyright 2016 Royal Society of Chemistry. (b) “Lab on a bubble” immunoassay developed with silica microspheres.
Reprinted with permission from ref 118. Copyright 2012 American Chemical Society. (c) Phage−SERS sensing platform developed with silica
microparticle immune substrates and filamentous M13 bacteriophage-assisted SERS probes. Reprinted with permission from ref 126. Copyright 2014
Wiley-VCH Verlag GmbH & Co. KGaA. (d) SERS-based biomarker immunoassay developed with core−satellite nanoassembly. Reprinted with
permission from ref 127. Copyright 2015 Royal Society of Chemistry.

coupling of surface and resonance enhancements can further which greatly reduced the interferences when sensing multiple
improve the detection sensitivity. Genrally, the metallic SERS biomarkers. Recently, Liu et al. presented a plasmonic
substrates are fabricated either by microelectronics fabrication immunosandwich assay for detecting low-copy-number protein
technology or through the assembly of nanoparticles. The in single living cells.116 The combination of gold layer substrate
former route requires sophisticated instruments and laborious with SERS probes enhanced the Raman signal by 9 orders of
human work, but the obtained SERS substrates are usually magnitude, which enabled unltrasensitive detection of intra-
highly uniform and reproducible. The nanoparticle-assisted cellular proteins. Interestingly, different from the typical
fabrication is sometimes easy to perform, but it requires sandwich immunoassay format, a SERS quantitative bioassay
elaborate design and careful operation. was created on aptamer-functionalized nanopillars.117 As shown
In a study by Zheng et al., an Au nanohole array was in Figure 7c, an ultrasensitive and specific detection of 5-
prepared by using polystyrene microbeads as the template,114 carboxytetramethylrhodamine (TAMRA)-labeled vasopress in
which was then used in combination with gold nanostar-based molecules was achieved by collecting highly enhanced SERS
SERS nanoprobes for the quantitative detection of Hg2+ and signals emanating from nanojunctions formed between the
Ag+ in human saliva. In this platform, the coupling between the leaning nanopillars and aptamer-functionalized surfaces. The
local surface plasmon resonance of the nanohole and nanostar SERS hot spots created by aptamer-target binding on a solid
enhanced the local electromagnetic field, subsequently substrate provide an alternative tool for immunoassays.
improving the detection sensitivity. Besides, Zou et al.
3.2. SERS-Based Immunoassay in Liquid Phase (Suspension
developed a sensing platform based on linearly aligned
Array)
magnetoplasmonic nanoparticles (NPs) (Figure 7a).115 The
one-dimensional alignment of magneto-plasmonic NPs sim- To create a SERS-based immunoassay, the solid substrates can
plified the immunoassay process and enabled a fast, enhanced be replaced by microbeads in liquid phase. The homogeneous
signal transduction. They used graphene quantum dots as the environment in liquid immunoassays greatly reduces the
SERS−fluorescence dual-mode labels to detect tuberculosis diffusion distance between antibodies and antigens, resulting
antigen. Similarly, a silver nanoparticle assembled film was in faster immune reactions. In general, microspheres are
produced to develop an ultrasensitive, multiplex Raman employed for the enrichment of target molecules in liquid
frequency shift immunoassay (Figure 7b).39 In this method, immunoassays, including silica beads,118 polystyrene beads,119
microcontact printing was employed to define the ordered magnetic beads,120,121 and so on. For high-throughput
domains of chemisorbed Raman reporters on the substrate, detections, multiple microbeads modified with specific anti-
7919 DOI: 10.1021/acs.chemrev.7b00027
Chem. Rev. 2017, 117, 7910−7963
Chemical Reviews Review

Figure 9. (a) A multiplex and straightforward aqueous immunoassay using silica-coated magnetic nanobeads. Reprinted with permission from ref 61.
Copyright 2013 Elsevier B.V. (b) Multiplex cancer cell detection and separation with silica-coated magnetic beads and SERS−fluorescence dual-
mode nanoprobes. Reprinted with permission from ref 135. Copyright 2014 Wiley-VCH Verlag GmbH & Co. KGaA. (c) S. aureus detection with a
MnFe2O4@Ag nanoparticle-based immunoassay. Reprinted with permission from ref 139. Copyright 2015 American Chemical Society. (d) High-
performance CTC enrichment with biomimetic immuno-magnetosomes and magnetic nanoclusters. Reprinted with permission from ref 140.
Copyright 2016 Wiley-VCH Verlag GmbH & Co. KGaA.

bodies or aptamers are used for simultaneous detection of technique, these immune substrates are capable of conducting a
multiple analytes in the liquid phase. As a large crowd of simultaneous detection of multiple biomarkers or cells in the
microbeads are readily suspended and drifted around in liquid phase. Interestingly, a kind of “lab on a bubble”
solutions, this high-throughput platform is also known as a immunoassay platform was developed by Schmit et al. using
suspension array.122,123 Moreover, magnetic beads are usually buoyant silica microspheres (Figure 8b).118 In such an
introduced in such arrays for a more easily achieved separation immunoassay structure, the analyte could be rapidly detected
or enrichment of the nanoprobes. Here, we will discuss some without an additional magnetic field or a centrifugation
examples of the substrates, like nonmagnetic and magnetic procedure. In addition, there are some studies focusing on
ones, respectively. signal amplification by using novel biomaterials in immuno-
3.2.1. Nonmagnetic Substrate. Frequently used non- assays. Lee et al. utilized filamentous M13 bacteriophage with a
magnetic beads include silica spheres, 118 polystyrene high surface area as a signal amplifier. As shown in Figure 8c,
spheres,119 metal nanoparticles124 and photonic crystal SERS nanoparticles were deposited on a single phage via a
beads,62 which are then modified with antibodies or aptamers layer-by-layer approach in their experiments, leading to
to form the immune substrate. Through the specific antibody− exponential gains in Raman intensities.126 The phage−SERS
antigen binding, SERS immunoprobes can be captured by the sensing platform was demonstrated as a highly sensitive, facile,
microbeads. The presence and concentration of target and rapid antigen detection system.
molecules can be characterized by SERS spectra. Another kind of homogeneous immunoassay is achieved
For example, SERS-based homogeneous immunoassay with a through the self-assembly of nanoparticles. The self-assembly of
sandwich structure could be created by antibody-functionalized nanostructures exploits the enhancement of the local electro-
polystyrene (PS) microbeads.119 SERS nanoprobes can be magnetic field by creating SERS hot spots, greatly improving
collected onto the PS microspheres through the recognition of the sensitivity of immunoassays. On the basis of this concept,
the target molecules, forming a typical sandwich immunocom- Feng et al. developed satellite nanoassemblies for the detection
plex for quantitative analysis. Yan et al. employed aptamer- of mucin-1 (MUC1) (Figure 8d),127 prostate-specific antigen
functionalized Au@Ag nanostars (NSs) for ultrasensitive (PSA), 128 aflatoxin B1 (AFB1),129 folic acid (FA),130
determination of chloramphenicol in milk samples.124 Chlor- biomarkers,131 etc. The self-assembly of metal nanostructures
amphenicol could bind to the aptamers on Au@Ag NSs, which assisted by target biomarkers created “SERS hot spots” with
greatly reduced SERS signals. Here, the metal nanoparticles enhanced Raman signals, which can be used for sensitive
served simultaneously as the immune substrate and the SERS quantitative analyses.
signal amplifier. Besides, as shown in Figure 8a, 3D ordered In terms of high-throughput detection, the microbeads-based
silver nanoshell silica photonic crystal beads or other kinds of immunoassay is compatible with the flow cytometer. Walker’s
photonic crystal beads have also been used for creating such group developed a SERS-flow cytometry for triplex detection of
SERS arrays.16,62,125 When combined with the SERS-encoding leukemia and lymphoma cells in combinaion with SERS-
7920 DOI: 10.1021/acs.chemrev.7b00027
Chem. Rev. 2017, 117, 7910−7963
Chemical Reviews Review

Figure 10. (a) A gold-array-embedded gradient microfluidic chip for SERS-based immunoassay. Reprinted with permission from ref 63. Copyright
2012 Royal Society of Chemistry. (b) A magnetic-assisted immunoassay on a microfluidic chip. Reprinted with permission from ref 145. Copyright
2015 Elsevier B.V. (c) “Magnetic focusing” microfluidic immunoassay for the detection of biomarkers. Reprinted with permission from ref 146.
Copyright 2015 American Chemical Society. (d) A SERS-assisted 3D barcode immunoassay for high-throughput immunoassay. Reprinted with
permission from ref 66. Copyright 2015 Wiley-VCH Verlag GmbH & Co. KGaA.

encoded nanoprobes.132 This platform is expected to promote antigen binding. This kind of nanocomposite is useful in
the development of SERS immuno-phenotyping for improving enormous applications in SERS-based immunoassays.
the sensitivity and specificity of blood cancer diagnosis. Previously, we have developed a multiplex and straightfor-
3.2.2. Magnetic Substrate. The excellent properties of ward aqueous immunoassay using silica-coated magnetic
magentic nanoparticles, such as high surface-to-volume ratio, nanobeads and SERS nanoprobes (Figure 9a).61 The magnetic
easy fabrication, low toxicity, and flexible bioconjugation, make beads were coated with a silica nanoshell to provide anchor
them popular in immunoassays. Magnetic nanoparticles with spots for antibodies. After the sandwich immunocomplex was
sizes from nanometers to micrometers can be synthesized using formed through the specific interactions between biomolecules,
various chemical methods. In a typical magnetic immunoassay, the concentration of analytes can be quantified by SERS
the repetitive washing processes can be easily and rapidly intensities. The use of magnetic beads avoids tedious washing
fulfilled with a magnet without tedious human work. Addition- steps during immune reactions, while the SERS nanoprobes
ally, magnetic beads help to enrich the target molecules in provide excellent optical properties (Figure 9b).135 Similarly,
samples, which improves the sensitivity of SERS-based work by Wang et al. used the magnet-assisted SERS-based
immunoassays. immunoassay to achieve a highly sensitive detection of the
Commonly used magnetic beads are Fe3O4 and Fe2O3 hormone estradiol E2.136 Li et al. fabricated multifunctional
magnetic beads [Fe3O4@polymethacrylic acid (PMAA)-SS-FA]
nanospheres.120,121 Although these magnetic nanoparticles
to capture, detect, and release circulating tumor cells.27 The FA
can be directly used for biomodification due to the rich
molecules can target cancer cells while the disulfide bond is
functional groups on their surfaces, it is recommended that a
sensitive to glutathione. Hence, the platform could be used for
protective layer (e.g., silica, metal or polymer) is first coated on the separation and recovery of cancer cells. On the other hand,
the magnetic beads. For instance, the presence of hydroxy as mentioned above, metal-nanoshell-coated magnetic beads
groups on the surfaces of these magnetic nanospheres enables were usually used for SERS enhancement. Many structures
the easy reaction with silane reagents. Hence, a silica shell can have been proposed to obtain such nanoparticles, among which
be coated onto the surfaces of magnetic nanoparticles.133 The Fe3O4@Au is one of the popular employed nanocomposites.137
coating of a silica nanoshell not only improves the stability and Recently, Jun et al. prepared a multimodal tagging material (M-
biocompatibility of magnetic beads but also makes it more SERS dots, Fe3O4@SiO2@Ag NPs) which has strong magnetic
convenient for the bioconjugation on magnetic beads. As a properties as well as Raman signatures for imaging, separating,
result, silica-coated magnetic nanoparticles are widely used in and targeting specific cancer cells.138 Wang et al. developed a
various research fields. In the case of SERS-based immuno- MnFe 2O4 @Ag nanoparticle-based immunoassay for the
assays, magnetic beads are usually wrapped with a gold or silver detection of Staphylococcus aureus using aptamers as the
nanoshell to form the magnetic/metal nanocomposites.134 The recognition units (Figure 9c).139 The presence of a metal
metal shell helps to enhance the SERS signal upon antibody− nanoshell helps to enhance the Raman signals and improves the
7921 DOI: 10.1021/acs.chemrev.7b00027
Chem. Rev. 2017, 117, 7910−7963
Chemical Reviews Review

Figure 11. (a) A microfluidic SERS sensor based on nanopillar forests that are produced through the oxygen-plasma-stripping-of-photoresist
technique. Reprinted with permission from ref 148. Copyright 2014 Wiley-VCH Verlag GmbH & Co. KGaA. (b) A plasmonic nanoarray embedded
microfluidic chip fabricated by combining photolithography and nanosphere lithography. Reprinted with permission from ref 149. Copyright 2015
Royal Society of Chemistry.

sensitivity for SERS immunoanalysis. Very recently, Xiong et al. microfluidic chips to assist SERS immunoassays for the fast and
fabricated biomimetic immuno-magnetosomes for the enrich- sensitive detection of an anthrax biomarker (Figure 10b).145 All
ment of circulating tumor cells (CTCs) from blood samples of the immunoassay processes can be automatically performed
(Figure 9d).140 Coating of a leukocyte membrane on the in the microfluidic channels, avoiding tedious human operation
prepared magnetic nanoclusters ensured the high specificity of and control. Recently, Li et al. developed a “magnetic focusing”
CTC detection. microfluidic immunoassay (Figure 10c).146 The enrichment of
3.3. Immunoassays on a SERS−Microfluidic Chip hot spots in microfluidic channels for SERS detection enabled a
high sensitivity. Besides, Kaminska et al. employed an Au−Ag-
With the rapid development of microfluidic (lab-on-a-chip)
coated GaN substrate as the novel immune solid platform to
technologies in the 21st century, SERS has been combined with
increase the efficiency of SERS-based microfluidic immuno-
microfluidics to create optofluidic immunoassays with an
assay. They realized the detection of hepatitis B virus antigen in
improved performance.141,142 The microfluidic platform
exhibits numerous advantages, including fast response, low human blood.147
sample consumption, high-throughput screening ability, and The microfluidic chip could be combined with the SERS-
portability.143,144 The high surface-to-volume ratio of micro- encoding technique for high-throughput biosensing. Previously,
meter-sized channels could accelerate the antibody−antigen a kind of SERS-assisted barcode chip for immunoassays was
binding. Consequently, the reaction rate of immunoassays developed by our group (Figure 10d).66 Multiple parallel
performed on microfluidic devices can be further accelerated microfluidic channels were used for the patterning of antibody
compared to those on solid substrates or in the aqueous phase. barcodes. Further, by combining the SERS-spectral-encoding
In addition, the continuous flowing environment helps to technique with the microfluidic spatial encoding, a simulta-
reduce nonspecific adsorption of SERS nanoprobes, which in neous detection of multiple targets in multiple samples could
turn increases the sensitivity and accuracy for a quantitative be achieved within 30 min.
detection. Therefore, SERS-based immunoassay has become In addition, many research groups have reported that
more and more popular in the last 5 years. Typically, there are plasmonic arrays with high SERS activity could be implemented
two basic kinds of SERS−microfluidic platforms: the micro- into microfluidic chips for a highly sensitive and reproducible
fluidic channels and the droplet microfluidics. Here, the biochemical analysis. For example, Mao et al. introduced
characteristics of each of the two types will be discussed, nanopillar forests into a microfluidic SERS sensor with a high
respectively. sensitivity (Figure 11a).148 In their study, the SERS substrate
3.3.1. Microfluidic Channels. The most common micro- was prepared by employing noble-metal-covered Si nanopillar
fluidic immunoassays are performed in microfluidic channels forests as a substrate, in which the nanopillar forests were
with a dimension ranging from 10 to 1000 μm. The produced on the basis of the oxygen-plasma-stripping-of-
microfluidic channels enable a parallel analysis of multiple photoresist technique. Under optimized fabrication parameters,
samples simultaneously. Combined with the SERS technique, the enhancement factor could reach an order of 1.5 × 106 and
the microfluidic immunoassays can improve the performance of the SERS signal variation was within the range of ±13%, which
immunoassays in terms of sensitivity, response time, makes the devices more reliable for chemical analysis. Similarly,
throughput, and overall costs. a plasmonic nanopillar array embedded microfluidic chip was
In the past 5 years, a variety of SERS-based immunoassays developed by Zhao et al. for in situ SERS analysis (Figure
have been performed in microfluidic channels. Importantly, the 11b).149 By combining photolithography and nanosphere
bottoms of the microfluidic channels are designed to contain a lithography, the chip is well integrated with highly efficient
film of metal or metal nanoparticles to generate SERS signals. SERS substrates. Highly reproducible SERS signals can be
Choo’s group reported a programmable and fully automatic obtained in the presence of microfluids. The wide enhancement
gold-array-embedded gradient microfluidic chip for SERS-based field and sufficient inter-nanopillar spacing allow the reprodu-
immunoassay, in which sandwich SERS immunoassays were cible SERS detection of dsDNA, which indicates the potential
performed within microfluidic channels embedded with gold of this platform for the detection of biomolecules. Yick et al.
arrays. The platform was able to simultaneously detect multiple fabricated Au-nanodot-decorated carbon nanotube arrays using
α-fetoprotein (AFP)-containing samples within 60 min (Figure a cold atmospheric-pressure microplasma jet, which required no
10a).63 Later, they also implanted magnetic nanoparticles into expensive vacuum equipment and could be operated in the
7922 DOI: 10.1021/acs.chemrev.7b00027
Chem. Rev. 2017, 117, 7910−7963
Chemical Reviews Review

Figure 12. (a) Multiplex bacteria detection with SERS−microfluidic platform. Reprinted with permission from ref 155. Copyright 2016 American
Chemical Society. (b) A wash-free droplet SERS immunosensor for PSA detection. Reprinted with permission from ref 156. Copyright 2016 Royal
Society of Chemistry.

Figure 13. (a) Multiplex cancer biomarker detection combining SERS with hollow core photonic crystal fiber. Reprinted with permission from ref
160. Copyright 2014 Wiley-VCH Verlag GmbH & Co. KGaA. (b) A tip-coated multimode fiber with a double-substrate “sandwich” structure for
protein detection. Reprinted with permission from ref 438. Copyright 2011 American Chemical Society. (c) Intracellular pH sensing in a single cell
with a plasmonic SERS optical fiber. Reprinted with permission from ref 161. Copyright 2009 Springer-Verlag. (d) Quantitative molecular
phenotyping with topically applied SERS nanoparticles for intraoperative guidance of breast cancer lumpectomy. Reprinted with permission from ref
162. Copyright 2016 Nature Publishing Group.

open air and at room temperature.150 The 3D hybrid approach can be used to investigate biochemical processes
nanostructure could serve as a reliable SERS sensing platform. occurring on the cell membrane152 and to perform broadband
In addition, De Angelis et al. showed that hollow plasmonic SERS analysis.153
nanostructures can accumulate the plasmonic field into 3.3.2. Droplet Microfluidics. Unlike the continuous
nanofluidic channels, thus providing a straightforward approach microfluidic flow systems, the droplet microfluidics focuses
to combine microfuidics with plasmonics.151 The same on creating discrete volumes by utilizing immiscible phases.
7923 DOI: 10.1021/acs.chemrev.7b00027
Chem. Rev. 2017, 117, 7910−7963
Chemical Reviews Review

Figure 14. (a) A plasmonic paper-based analytical platform with functional versatility and a sub-attomolar detection limit using SERS as a
transduction method. Reprinted with permission from ref 64. Copyright 2013 American Chemical Society. (b) A lateral flow assay biosensor for
highly sensitive detection of HIV-1 DNA. Reprinted with permission from ref 165. Copyright 2013 Elsevier B.V.

Droplet microfluidic systems enable miniaturized reactions in biomarkers (Figure 13a).160 In this platform, immunoassay was
compartmentalized droplets of femtoliters to microliters. It performed in the hollow core of the fiber. HCPCF provides
enables rapid and high-throughput analysis without increasing better light confinement and a larger interaction length for the
the complexity and size of the microfluidic chip. This kind of guided light and the analyte, resulting in an improved sensitivity
platform provides advantages including rapid mixing of the of bioanalytes at extremely low sample volume. Gu and
reagents, controlled timing of the reaction, and the ability to coauthors fabricated two structures for SERS−optical fiber
synthesize highly uniform micro/nanostructures.154 Droplet biosensors. One is based on a tip-coated multimode fiber with a
microfludics could be combined with the SERS technique for double-substrate “sandwich” structure, which achieved a limit of
rapid and reproducible analysis. The turbulent flow inside each detection (LOD) of 0.2 μg/mL in protein detection (Figure
droplet greatly accelerates the mixing between nanoparticles 13b).438 The other one is based on a liquid core photonic
and analytes, which improves the efficiency of reactions. To crystal fiber with a detection limit of 106 cells/mL for the
date, the droplet SERS platform has been used for various detection of bacteria. Both of their SERS biosensors showed
biochemical applications. Previously, our group developed a great potential for highly sensitive biosensing. Vo-Dinh’s group
droplet SERS−microfluidic chip for the rapid and sensitive focued on a SERS-based plasmonic biosensor using an optical
detection of thiocyanate in human saliva and serum.79 Au@Ag fiber, in which the pH-sensitive 4MBA molecules were attached
NRs were mixed with saliva and serum samples that contain onto silver nanoisland films coated onto the fiber (Figure
SCN− in microdroplets. The peak intensity at 2100 cm−1, 13c).161 This sensor can be used to measure the intracellular
which originates from the −CN bond, was used to pH in human mammary epithelial cells and PC-3 human
characterize the concentration of thiocyanate. Muhlig et al. prostate cancer cells.
employed a lab-on-a-chip (LOC) platform for the differ- Recently, Wang et al. combined SERS nanoprobes with
entiation of six species of mycobacteria using SERS spectros- optical fibers for a rapid (15 min) quantitative molecular
copy (Figure 12a).155 The spectral information was derived prototyping (QMP) (Figure 13d).162 The results of QMP
from the vibrational signals of the cell-wall component mycolic imaging of excised tissues from fresh human breast specimens
acid. This platform was successfully applied to identify agree well with those of flow cytometry and immuno-
Mycobacterium tuberculosis complex and non-tuberculous histochemistry, which may find potential applications for
mycobacteria. Moreover, similar to the SERS immunoassay in guiding breast-conserving surgeries.
the liquid phase, droplet microfluidics have also been combined 3.5. Paper-Based SERS Platforms
with magnetic immunoassays for the ease of separation or wash
of SERS probes. For example, Choo and coauthors presented a More recently, paper has emerged as a potential replacement
novel wash-free immunosensor for PSA detection (Figure for conventional substrates. It is quite simple and cheap to
12b).156 The magnetic bars enabled the separation of free and produce, easy to take, and has an intrinsic capacity for fluid
bound SERS tags, so that the SERS intensity could be used to transportation.163,164 Generally, the paper-based devices
characterie the concentration of PSA. provide the advantages of extremely low cost, high flexibility,
and ease of use. Importantly, some of these devices provide an
3.4. SERS-Based Immunoassay on Optical Fibers excellent detection sensitivity comparable to traditional
While for immuno-detection, SERS-based immunoassays can substrates, making them a promising choice for SERS-based
also be conducted based on optical fibers, especially in the immunoassays.
applications using portable SERS spectrometers and endos- As an example, a plasmonic paper-based analytical platform
copies, due to their long-distance optical conduction ability and for a quantitative SERS detection was developed by
flexibility.157−159 Combining the SERS technique with a hollow Singamaneni and coauthors (Figure 14a).64 The analytical
core photonic crystal fiber (HCPCF), Dinish et al. developed platform was prepared using a simple cut and drop method
an ultrasensitive platform for multiplex detection of cancer without lithography. The fluids could transport through the
7924 DOI: 10.1021/acs.chemrev.7b00027
Chem. Rev. 2017, 117, 7910−7963
Chemical Reviews Review

Figure 15. A SERS sensing platform combining a plasmonic array with a super-hydrophobic substrate for unltrasensitive detection. Reprinted with
permission from ref 168. Copyright 2011 Nature Publishing Group.

paper with a rapid capillary-driven flow. There is no need for hydrophobic surfaces with nanoplasmonics for detection
microchannel patterning. The combination of this concept with (Figure 15).168,169 They achieved an ultrasensitive SERS
SERS analysis pushes the detection limit down to the attomolar detection under an attomolar concentration. Besides, a method
level, making microfluidic paper-based analytical devices using Ag/ZnO surface modified with organic stearic acid and
competitive with the traditional sensors. Commercial inkjet polyvinylpyrrolidone was demonstrated by Meng and co-
printers can also be employed for fabricating SERS substrates. authors.170 The hydrophobic substrate enhanced the SERS
Hoppmann et al. developed an inkjet-printed paper-based signals by 3 times compared to the hydrophilic surfaces, which
SERS devices for 1,2-bis(4-pyridyl)ethylene (BPE) detection improved the sensitivity of SERS-based detections. Leiterer et
(Figure 14b).165 The low-cost fabrication, ease of use, and high al. presented a straightforward way to obtain the monolayer
sensitivity make it a potentially practical SERS substrate. assembled by silver nanoparticles, in which Ag NPs were coated
Another kind of novel SERS substrate is fabricated through the with a hydrophobic shell to induce self-assembly without any
in situ synthesizing method. Liu et al. was able to synthesize other surfactants.171 The SERS hot spots generated with the
gold nanoparticles on flexible silk fabrics directly through redox assistance of hydrophobic nanoparticles could enhance the
reaction.166 The simple SERS substrates were used for trace SERS signals. Similarly, Hou et al. synthesized hydrophobic Ag
analysis of p-aminothiophenol (pATP), 4-mercaptopyridine (4- NPs in the presence of tri-n-octylphosphine sulfide.172 They
MPy), and crystal violet (CV) molecules. One limitation of the demonstrated that the hydrophobic AgNPs had a higher SERS
inkjet printing is that the functional inks must be suitable for enhancement factor than that of the hydrophilic ones. On the
printing. To overcome this shortcoming, Große et al. combined basis of the above results, the sensitivity of label-free SERS
laser printing with paper substrates for peptide patterning. They detection could be improved with the phase transfer from
exploited material-specific adsorption of peptides and enabled hydrophobic to hydrophilic. As another interesting example,
the selective functionalization of laser-printed patterns and Wallace et al. realized the super-hydrophobic analyte
nonprinted paper regions.167 In this device, no specific toners concentration utilizing SERS substrates made of colloid-pillar
are needed for printing. The peptides could be specifically arrays.173 The pillars provide a route to concentrate target
captured on the printed or nonprinted areas. This kind of molecules via super-hydrophobic droplet evaporation effects. It
substrate could potentially be used for the patterning of other was estimated that the concentration was over 100 times
biomolecules, like antibodies and aptamers, which could be increased, resulting in a detection limit down to 2.9 × 10−12 M
used as the immune substrates for SERS-based immunoassays. for mitoxantrone dihydrochloride. Despite the tremendous
advancement, it is still challenging to create a platform for
3.6. Hydrophobic SERS Substrates
detecting analyte molecules in all phases with a high specificity
The detection of a few molecules in diluted solutions has and sensitivity. To overcome this limitation, Yang et al.
attracted much attention in the field of biomedicine, security, developed a universal platform named slippery liquid-infused
and environmental monitoring. To achieve ultrasensitive porous SERS.174 The platform enables the enrichment and
detections, various hydrophilic SERS substrates have been delivery of analytes originating from solid, liquid, and gas
developed. The hydrophobic SERS substrates also belong to phases. The analyte molecules could be delivered to SERS-
the solid substrate. However, due to their unique properties of active sites for a highly sensitive detection at a concentration
enriching the hydrophilic target molecules, we specially discuss down to the sub-femtomolar level. Tabakman et al. fabricated
these kinds of novel solid substrates here. plasmonic substrates for multiplex protein microarrays with a
In general, most of the traditional SERS substrates are femtomolar sensitivity and a broad dynamic range.175 The gold
hydrophilic; hence, the analyte molecules are easily spread over on gold nanoisland film, prepared by uniform, solution-phase
the surfaces (since most bioanalytes are also hydrophilic). One growth method, affords near-infrared fluorescence enhance-
of the most promising routes to improve the sensitivity is to ment (NIR-FE) of up to 100-fold, which is useful for the
fabricate super-hydrophobic SERS substrate to overcome the significant improvement of protein microarray detection assays.
diffusion limit of analyte molecules. To overcome the limit Later, Zhang et al. employed the developed plasmonic sensor
dictated by diffusion, De Angelis et al. combined super- for biomarker discovery and diagnosis of type 1 diabetes, which
7925 DOI: 10.1021/acs.chemrev.7b00027
Chem. Rev. 2017, 117, 7910−7963
Chemical Reviews Review

Table 2. Comparison of Different Immunoassay Platforms on Fabrication, Human Operation, Sample Consumption, Time
Consumption, and Detection Sensitivity
solid
liquid: nonmagnetic and
nonmetallic metallic magnetic microfluidic optical fiber paper hydrophobic
fabrication commercially microfabrication commercially available/ microfabrication commercially commercially microfabrication
available and self-assembly chemical synthesis available available
human operation repeated washing and reagent adding repeated washing and highly automated highly repeated repeated washing and
mixing automated reagent reagent adding
adding
sample and reagent 10−30 10−30 0.1−5 0.01−5 2−20 10−30
consumption (μL)
time consumption (h) 2−4 2−4 0.25−1 2−3 0.25−1 2−3
sensitivity level μM to pM nM to fM μM to fM nM to fM nM to fM μM to pM nM to aM

showed a comparable sensitivity and specificity to radio- realize the detection of multiple analytes in an immumoassay,
immunoassays.176 fabrication of SERS-encoded nanoparticles (i.e., the SERS
3.7. Comparison between Different Platforms for encoders) is a prerequisite. Up to now, increasing attention has
SERS-Based Immunoassay been paid to the creation of hybrid nanoparticles for generating
SERS encoders with a high encoding accuracy and capacity.
In summary, different platforms have their own characteristics.
Here, we list the advantages and disadvantages of each platform 4.1. Design and Fabrication of SERS Encoders
in Table 2 for a clear comparison. Efficient SERS-encoding strategies depend directly on the
design and fabrication of SERS encoders. Although SERS
4. SERS ENCODING FOR MULTIPLEX DETECTION probes for immunoassay have already been introduced in the
In practical biomedical applications (e.g., clinical diagnosis), previous section, the specific requirements of encoded SERS
high-throughput sensing technologies have become increasingly probes (denoted as SERS encoders) still deserve to be
important with the growing demands in which millions of demonstrated. Up to now, various kinds of SERS encipherors
synthetic reactions and biological tests are performed have been reported. To simplify the following discussion and
simultaneously. One of the most popular ways to conduct a better demonstrate the spectral-encoding technique, we
multiplex assay is to use encoding microbeads or nanoprobes propose a concise classification method for SERS encryptors
with a unique code to identify the attached ligand according to their structures. Usually, in a SERS encoder,
molecules.177,178 Up to now, various encoding strategies have Raman reporters are assembled onto metal nanoparticles to
been proposed and demonstrated, including spectrometric produce SERS signals, forming a signal-generating layer
encoding,179−181 graphical enciphering,182,183 electronic en- composed of reporter molecules. Accordingly, if the SERS
crypting,184,185 physical encoding,186,187 chemical encipher- encipherors contain only one signal-generating layer, they can
ing,188,189 and magnetic encrypting,190,191 generating a great be classified as monolayer ones. Meanwhile, if the SERS
number of accurate codes for multiplex detections. Since encryptor contains more than one signal-generating layer, they
nonoptically based enciphering methods have been well can be referred to as multilayer ones (Figure 16).65,198,199 Here,
described elsewhere,192,193 we only discuss the optical (i.e., the “signal” is not limited to SERS, as other kinds of optical
spectroscopic) encoding methods with a focus on the SERS- signals (e.g., fluorescence) can also be included.
encoding technique, which is widely used owing to its As shown in the left panel of Figure 16, typical monolayer
convenient enciphering and noninvasive deciphering operation. SERS encoders are obtained by assembling a layer of Raman
Optical encoding, also known as spectrometric encoding, can reporters on the surfaces of metal nanoparticles. Since the size
be divided into four main families according to the light- of the reporter molecules is quite small as compared with that
emitting mechanisms: fluorescence, SERS, reflection, and of the inner metallic nanoparticles (typically around 10−100
luminescence lifetime.194,195 Among these, fluorescence- and nm), the overall size of such monolayer SERS encipherors is
SERS-based encoders occupy a dominant role owing to their determined by the metal nanoparticles (i.e., on the nanoscopic
relatively simple encoding process and high encoding scale). The nanoscale monolayer SERS encryptor can be
capacity.195 Fluorescence is popular for its high brightness denoted as nanoencoders for simplicity.196 By choosing
and easy readout, while SERS is advantageous for its narrow different kinds of Raman reporters assembled in the signal-
spectral bands and high resistance toward fluorescence generating layer, nanoencoders with distinguished SERS codes
backgrounds.70 However, when typical fluoresent dyes are can be acquired. However, the limited nanoscale surface area
used, the limited numbers of available fluorescent agents with and the complexity of the reporter adsorption process severely
the same excitation wavelength and the overlapping of the hinder the realization of reproducible nanoencoders, especially
emission bands caused by broad emission profiles restrict the when several Raman reporters are involved together. Thus, the
number of available distinct codes. In the SERS encrypting biggest obstacle in fabricating nanoencoders is how to produce
method, distinct emission signatures can be obtained by strong and reproducible SERS signals.
selecting different Raman reporters or their combinations.196 To solve this problem, another kind of monolayer SERS
Additionally, ratiometric codes can be created by tuning the encoder is proposed, which is illustrated in the middle pannel
intensity ratio between different characteristic SERS peaks, of Figure 16. This type of SERS encoder (denoted as
which can greatly increase the enciphering capacity.197 Further, microencoders according to their overall sizes) also contains
by combining SERS with other optical-encoding methods, the one signal-generating layer. The difference as compared with
encoding capacity can even be increased exponentially.135 To the SERS nanoencoders is that, in a nanoencoder, Raman
7926 DOI: 10.1021/acs.chemrev.7b00027
Chem. Rev. 2017, 117, 7910−7963
Chemical Reviews Review

decorated with one kind of Raman reporter to form the raw


SERS nanoprobes/dots. Different SERS nanoprobes/dots can
be produced by changing the Raman reporters. Then, the as-
prepared SERS nanoprobes/dots with different codes self-
assemble onto the microscale scaffold, forming a larger
microencoder. Commonly employed scaffolds are silica and
polystyrene microspheres due to their abundant size choices
and easy surface modification.201,202 This kind of microencoder
exhibits a high feasibility in achieving different codes, which can
be straightforwardly accomplished by changing the type or the
amount of SERS nanoprobes/dots on their surfaces. The SERS
nanoprobes/dots can be fabricated in realitively large scale with
acceptable signal variations, thereby microencoders usually
exhibit a better reproducibility as compared with nanoencoders.
In addition to the monolayer SERS encoders discussed
above, another type of SERS encoder is the multilayer one,
which refers to the encoders containing more than one signal-
generating layer (see the right pannel of Figure 16). In the
Figure 16. Classification of SERS encoders regarding their monolayer encoders, signal generators are all incorporated in a
composition and structure. For clarity, only two or three coding single layer, making the encoding procedures sometimes
elements (Raman reporters or quantum dots) are shown. Images are chaotic since the signal generators can compete with each
not to scale. other when adsorbing to the cores. The advantage of multilayer
encoders is that different layers contain different kinds of signal
molecules are directly used as the signal generators and generators, which are separated from each other. Thus, the
assembled on a nanoscale metallic core. However, in the interference between different signal generators can be greatly
microencoder, individual SERS nanoprobes (or SERS dots) are reduced and the encoding procedures become more control-
used as the SERS-generating units and assembled on a larger lable.
microscale scaffold (e.g., polystyrene spheres).92,200 To sum up, A representative multilayer encoder is the SERS−fluores-
both SERS nanoencoders and microencoders contain a single cence dual-encoded nanoprobe presented by our group. The
signal-encoding layer, but their overall sizes and signal- nanoprobe contains two signal-generating layers, a SERS layer
generating units are different. consisting of Raman reporters and a fluorescent layer
In microencoders, since the signal-generating layer consists composed by quantum dots (QDs). To be specific, the
of SERS nanoprobes/dots, various SERS codes can be facilely nanoprobe was fabricated by a layer-by-layer assembling
obtained by changing the SERS nanoprobes/dots. Typically, to strategy using metal nanoparticles, organic molecules, and
fabricate such microencoders, metal nanoparticles are only semiconductor QDs. Raman reporters and fluorescent QDs

Figure 17. Typical TEM image of SERS labels (AuNPs−TFMBA@SiO2). (b) SEM image of a single PS bead encoded with SERS nanoparticle
labels. (c) SERS spectra of AuNPs−TFMBA@SiO2 (left) and molecular structures of Raman reporters employed in this study (right). (d) Typical
Raman spectra (left) of microbeads with different types of SERS label using different Raman reporters (10000, 01000, 00100, 00010, 00001, and
11111) and their corresponding spectral barcodes (right). Reprinted with permission from ref 204. Copyright 2015 The Royal Society of Chemistry.

7927 DOI: 10.1021/acs.chemrev.7b00027


Chem. Rev. 2017, 117, 7910−7963
Chemical Reviews Review

Figure 18. (a) Schematic illustration of the preparation of the encoded SERS nanoprobes. (b) Color-coded SERS spectra of individual Raman
reporters: BDT (red), 2-NAT (green), and 4MT (blue). (c) Codes, structures, and measured spectra of the synthesized 19 SERS encoders. The
spectra are only shown from wavenumber 600 to 900 for clarity. Reprinted with permission from ref 65. Copyright 2014 The Royal Society of
Chemistry.

were assembled onto two different layers and separated with an which can be expressed with an easy combination calculus as
inert isolating layer. Such a structure can well-prevent the signal follows
cross-talk and guarantee a high stability. The two-layer-type
n
nanoprobe can fulfill SERS−fluorescence joint spectral
encoding (SFJSE), which is dicussed in detail in section 4.3.1. N= ∑ Cni = 2n − 1
i=1
4.2. SERS Spectral Encoding Strategy
4.2.1. Raman-Frequency-Based Encoding. So far, SERS- where N is the total possible number of codes and n is the
encoding approachs depending on either Raman frequency or number of different kinds of Raman reporters.204 According to
signal intensity have both been reported. Due to the narrow the equation, more than 100 barcodes can be generated with 7
bandwidths in Raman spectra, the SERS-encoding method different Raman reporters.
using Raman frequency (i.e., Raman wavelength) has attracted 4.2.2. Signal-Intensity-Based Encoding. According to
great attention. Su et al. proposed spectroscopically distin- the above-mentioned encoding method using Raman fre-
guished SERS codes by using three different Raman tags in quency, an enormous number of Raman reporters have to be
composite organic−inorganic nanoparticles (COINs).203 Re- involved with the increasing number of required codes. This
cently, with the investigation of wavelength-based-encoding makes the fabrication procedures rather complicated and limits
strategy, practically available codes with more Raman reporters the actual encoding capacity. Although the amount of
were accomplished in reality. For example, Wang and co- commercially available Raman reporters is relatively abundant,
workers developed a kind of SERS microencoder that was their similar chemical structures make them difficult to be well
fabricated using five types of silica-encapsulated SERS nano- distinguished by Raman frequency when lots of reporters are
encoders (AuNPs-Ra@SiO2) (Figure 17).204 These five types used together. Thus, as one of the solution, another SERS-
of SERS nanoencoders were composed of Raman reporters encoding method has been developed by using signal intensity
such as 4MBA, 2,7-mercapto-4-methylcoumarin (MMC), 2- as an additional code element. As presented by our group, an
mercapto-4-methyl-5-thiazoleacetic acid (MMTAA), 5-thio-2- enormous number of codes could be generated using the
nitrobenzoic acid (MNBA), and 2,3,5,6-tetrafluoro-4-mercap- intensity-based SERS-encoding approach.65 We initially se-
tobenzoic acid (TFMBA), respectively. Thus, the correspond- lected three kinds of Raman reportersbenzenedithiol (BDT),
ing distinct peaks at 1080, 1172, 1290, 1337, and 1380 cm−1 2-naphthalenethiol (2NAT), and 4-methoxythiophenol
were used for encoding in the SERS microencoder (Figure (4MT)to fabricate SERS nanoencoders, which were
17c). Since the most intensive peak of each Raman reporter was commercially available aromatic compounds with different
separated by at least 35 cm−1, the SERS signature can be easily functional groups. Correspondingly, Raman peaks at 730, 767,
distinguished, resulting in a straightforward decoding process. A and 794 cm−1 were used as the encoding bands. A ternary
binary notation was introduced to characterize possible signal notation was introduced to describe the intensity of character-
combinations, where “1” represents the presence of the istic peaks, where “0” represented the absence of the SERS peak
corresponding SERS peaks while “0” represents the absence and “1” or “2” stood for a low- or high-intensity level of the
(Figure 17d). Consequently, 31 spectrally distinct SERS corresponding peak, respectively. According to the information
signatures (i.e., barcodes) can be obtained by mixing five on both the wavenumber and intensity of the SERS signals, a
different Raman reporters together. total of 19 codes were achieved experimentally with distinguish-
Definitely, the potential number of codes (denoted as N) can able spectral signatures. The components, structures, and
be increased simply by using more kinds of Raman reporters, measured SERS spectra of different codes are listed in Figure
7928 DOI: 10.1021/acs.chemrev.7b00027
Chem. Rev. 2017, 117, 7910−7963
Chemical Reviews Review

Figure 19. (a) Schematic illustration of the preparation of organic−metal−QD hybrid nanoparticles (OMQ NPs). (b) STEM image and the
corresponding EDX elemental distribution of Au, Ag, Si, and Cd in OMQ NPs. The merged mapping is also shown. (c) Composition, measured
spectra, and codes of the synthesized 15 nanoparticles. Reprinted with permission from ref 17. Copyright 2012 American Chemical Society.

18. It is worth noting that only 7 codes can be generated in a encoding capacity is limited by the spectral overlap when a sole
sole wavenumber-encoding system. SERS or fluorescence signal is used, which is far less than that
Similarly, the groups of Lee and Wu further explored the predicted theoretically. Specifically, the Raman fingerprint
feasibility of a joint SERS-encoding approach using Raman region of most organic SERS reporters is in the range of
shifts and intensities.197,200 For instance, two kinds of Raman 500−2000 cm−1 (i.e., only ∼100 nm for excitation wavelengths
reporters, such as 4-methylbenzenethiol (4MT) and 2- at visible region) and the commonly used SERS reporters have
naphthalenethiol (2NAT), were mixed at different molar ratios similar chemical structures. Therefore, decoding the Raman
(4MT:2NAT = 3:1, 7:1, 12:1, 19:1, 39:1) to generate different spectrum of a plurality of reporters may become difficult owing
codes. Raman shifts and intensities of representative peaks of to the spectral overlap. All of these factors restrict the number
each chemical can be clearly resolved. Under this circumstance, of codes that can be realized practically.
the maximum number of potential codes (N) was determined To solve this problem, we have presented a new concept for
by both the number of selected reporters (M) and the number optical encoding, the SERS−fluorescence joint spectral
of intensity levels (i) encoding method (SFJSE) (Figure 19).17 This method has
M
two characteristics to ensure a greatly enlarged encoding
L! capacity. First, the spectral range available for encoding is
N= ∑ (i k − i + 1)
(L − k) ! k! broadened through the utilization of both photoluminescence
k=1
(PL) and SERS spectral regions, allowing for more spectrally
where L is the number of available Raman reporters.200,203 For distinguishable codes. For example, when two kinds of Raman
example, when two SERS reporters and five intensity levels reporters (4MBA and DTNB) and two kinds of QDs (CdTe
were selected (in this case, L referred to 35), more than 10 000 515 and CdTe 591) were employed, a total of 15 codes were
types of SERS nanoencoders with specific signatures can be achieved experimentally (Figure 19c). In constrast, in an
created. encoding system based solely on fluorescence or SERS signals,
4.3. SERS-Included Joint Encoding only three codes can be generated using two fluorescence or
two SERS agents, which was far fewer than those based on
As is well-known, the spectral fingerprint region of most SFJSE. When more fluorescence (m) or SERS agents (n) are
organic Raman reporters is in the range of 500−2000 cm−1 (i.e., introduced, the potential encoding capacity of OMQ NPs (N)
only ∼100 nm with excitation wavelengths in the visible can be estimated using the following equations
region). Although Raman spectra have quite narrow bands,
such a narrow spectral window (∼100 nm) still gets quite m+n m+n
(m + n)!
crowded when multiple reporters are used simultaneously, N= ∑ Cm+ ni = ∑ = 2m + n − 1
resulting in inevitable spectral overlap. A brilliant solution is to i=1 i=1
i ! (m + n − i )!
introduce another kind of optical signal (e.g., fluorescence) to
SERS encoders. Such encoders based on joint optical signals N /N1 = (2m + n − 1)/(2m − 1)
can remarkably increase the encoding capacity compared with
those tagged solely with SERS signals.16,17 Besides, combining N /N2 = (2m + n − 1)/(2n − 1)
SERS signals with a spatial address for hybrid encoding offers
another choice for increasing the number of generated codes. where N1 and N2 are the number of codes in fluorescence and
4.3.1. SERS−Fluorescence Joint Spectral Encoding. As SERS-encoding systems, respectively.17 Moreover, the number
mentioned above, although multiplex optical sensing could be of available codes can be further increased when the intensity-
achieved with either SERS or fluorescence encoding, the encoding mode is also considered.
7929 DOI: 10.1021/acs.chemrev.7b00027
Chem. Rev. 2017, 117, 7910−7963
Chemical Reviews Review

Figure 20. (a) Label-free SERS immunoassay of IgGs. IgGs are captured and analyzed using antibody-modified Fe3O4@Ag nanocomposites.
Reprinted with permission from ref 239. Copyright 2014 American Chemical Society. (b) SERS-based gradient optofluidic sensor for fast and
sensitive immunoassay. Hollow gold nanospheres are used as the SERS nanoprobes. Reprinted with permission from ref 232. Copyright 2010
American Chemical Society. (c) SERS immunoassays based on frequency shifts. Binding of the target antigen can cause a frequency shift in the
Raman bands, which can subsequently be used to quantify the targets. Reprinted with permission from ref 238. Copyright 2012 American Chemical
Society. (d) Labeled SERS immunoassay of IgGs using gold mesoflowers as the SERS nanoprobes and gold-coated magnetic nanoparticles as the
capturing substrates. Reprinted with permission from ref 243. Copyright 2014 American Chemical Society.

Second, to create such an encoder, organic−metal−quantum 3D barcode. The SERS-assisted 3D barcode greatly enhances
dot hybrid nanoparticles (OMQ NPs) were achieved using a the information storage capacity. All necessary information,
core−shell structure with multiple layers, in which Raman including the species and amount of target analytes, can be
reporters and fluorescent QDs were assembled onto different simultaneously read out during the detection process. To
layers (Figure 19a).17 It makes the fabrication process easier summarize, the SERS-based joint encoding strategy is practical
and more controllable for increasing the number of involved and feasible for the easy and rapid identification of large
agents. Therefore, using the proposed SFJSE method, the chemical and biological libraries. A beneficial aspect of the joint
number of the codes that can be generated practically is much encoding strategy is the diversification of encoding elements.
more than that through a traditional fluorescence- or SERS- Besides fluorescent or spatial signals, extinction or other optical
based encoding method, which makes the SFJSE approach signals can also be combined with SERS. Moreover,
particularly promising for multiplex bioanalysis at high levels.
substantially more codes can be acquired when multimodes
Later, Li and co-workers further developed this SERS−
or the intensity-encoding mode is also considered.
fluorescence joint encoding system with material displacement.
They used bipyramid gold nanocrystals as the SERS substrates
instead of Au/Ag nanorods for a more sensitive detection. 5. APPLICATIONS OF SERS-BASED IMMUNOASSAY
Moreover, gold nanoclusters served as fluorescent agents for a So far, SERS-based immunoassays have been well-employed in
better biocompatibility.198 biochemical and biomedical applications for the detection of
4.3.2. Spectral−Spatial Joint Encoding. As discussed biological targets, such as proteins,110,205,206 nucleic acids,207,208
above, there are many different types of SERS-based encoding. virus,209 cells,210,211 and toxins.212 Up to now, there are a series
However, they are restricted to spectroscopic encoding, which of well-established traditional biological detection strategies for
also can be referred to as 1D encoding. Interestingly, by these targets [such as ELISA,213 Western blot,214 flow
combining SERS spectroscopic encoding (1D) with spatial
cytometry,215 mass spectroscopy,216 electrophoresis,217 and
encoding (2D), a 3D encoding strategy can be accomplished.
polymerase chain reaction (PCR)218]. They have played a quite
Our group proposed the concept of a “3D SERS barcode”,
which adds SERS fingerprints as the third dimension to the important role in the development and investigation of
widely used 2D barcode (Figure 12d).66 To make the 3D SERS sophisticated biological and physiological issues. However,
barcode easier to understand, a multiplex immunoassay was they usually suffer from some unsatisfactory shortcomings,
conducted on the basis of a microfluidic chip. First, multiple including limited sensitivity, complicated working procedures,
proteins were spatially separated using a patterned antibody and a high risk of artificial contaminants. Compared with
barcode substrate, forming a 2D hybridization array. Then traditional biological detection methods, SERS-based immuno-
SERS nanoencoders were used to identify and quantify the assays stand out due to some unique and excellent character-
proteins. As different SERS nanoencoders carry different kinds istics, for example, negligible background noise, low spectral
of Raman reporters, they can incorporate spectroscopic overlap, high sensitivity, large dynamic range, and fine
information (1D) into the 2D spatial barcode, resulting in a multiplexing ability.8,130,219,220
7930 DOI: 10.1021/acs.chemrev.7b00027
Chem. Rev. 2017, 117, 7910−7963
Chemical Reviews Review

Table 3. SERS-Based Immunoassays for IgG Detection


refs IgGs recognition unit detection platform dynamic range LOD
83 human IgG goat anti-human IgG Ag glass substrate 100 ng/mL to 100 fg/mL 100 fg/mL
239 human IgG anti-IgG antibody liquid phase 100 ng/L to 500 mg/L 600 fg/mL
246 human IgG anti-human IgG gold patterned substrate 1 μg/mL to 10 fg/mL 10 fg/mL
247 human IgG goat anti-human IgG fingerprints on glass slides 1 × 10−7 to 1 × 10−1 mg/mL 1 × 10−7 mg/mL
248 human IgG goat anti-human IgG gold triangle nanoarray 0.1 pg/mL to 10 ng/mL 7 fg/mL
81 human IgG goat anti-human IgG gold nanostar substrate (1 × 10−4)−10−14 g/mL 10 fg/mL
249 human IgG goat anti-human IgG nanoporous waveguide (PAA) film (1 × 10−10)−(1 × 10−6) g/mL 0.1 ng/mL
250 human IgG goat anti-human IgG bowl-shaped silver cavity array 0.5−5 ng/mL 0.1 ng/mL
206 human IgG anti-human IgG polystyrene substrate 45−500 pM 45 pM
251 mouse IgG goat anti-mouse IgG liquid phase 50 ng/mL to 1.9 pg/mL 1.9 ng/mL
252 mouse IgG goat anti-mouse IgG Ag + diatom biosilica substrates 1 pg/mL to 10 μg/mL 10 pg/mL
253 mouse IgG goat anti-mouse IgG antibody Ag colloid/PVP/paper 1−500 ng/mL 0.33 ng/mL
244 mouse IgG goat anti-mouse IgG liquid phase 1−(5 × 105) fg/mL 1 fg/mL
61 mouse IgG goat anti-mouse IgG liquid phase 0.1 pg/mL to 1 μg/mL 0.1 pg/mL
254 mouse IgG goat anti-mouse IgG gold-plated filters 10−1−(1 × 105) ng/mL 0.84 ng/mL
84 mouse IgG goat anti-mouse IgG glass substrate 100 pg/mL to 100 μg/mL 100 pg/mL
110 mouse IgG anti-β-actin mouse antibody quartz slide 0.01−1000 μg/mL 2.5 ng/mL
255 rabbit IgG goat anti-rabbit IgG bowl-shaped silver cavity thin film 0.1−20 ng/mL 5 pg/mL
232 rabbit IgG polyclonal antibody microfluidic chips 0−100 ng/mL 1−10 ng/mL
66 mouse IgG goat anti-mouse IgG microfluidic chips 10−8−10−14g/mL 10 fg/mL
human IgG goat anti-human IgG
242 mouse IgG anti-mouse IgG Ag substrate 5 ng/mL to 5 fg/mL 800 ag/mL
human IgG anti-human IgG 5 ng/mL to 500 fg/mL 5 fg/mL
238 H1 protein anti-H1 antibody Au/Ag bimetallic glass 0−100 nM 2.2 nM
p53 protein anti-human p53 substrate 2.5 nM

5.1. Analysis of IgGs the analysis of mouse IgG.244 The detection limit is 1 fg/mL
IgGs play a vital role in the immune system of an organism, and the dynamic range is from 1 to 5 × 105 fg/mL, which is 5
helping to protect the organism from external invasions of orders of magnitude. In their work, the SERS nanoprobe was
bacteria and viruses. Hence, detection of IgGs is important for fabricated by attaching antibodies onto gold-core−silver-shell
the determination of infectious or immune-system-related nanorods, and Raman reporter molecules were embedded in
diseases (e.g., rheumatoid arthritis, hepatitis, and lupus the silver shell to provide SERS signals. Mouse IgGs were
erythematosus). In addition to Western blot and mass anchored on the surfaces of the magnetic beads. Hence, the
spectrometry, the most representative method for analyzing immune recognition between mouse IgGs and the antibodies
the species and concentrations of IgGs is immunoassay, which on the SERS nanoprobes allows the formation of a sandwich-
is usually realized by the immune recognition between IgGs type immune complex. The fine performance of the presented
and their specific antibodies. According to different signal assay is attributed to the 3D structure of magnetic beads, which
can reduce the steric effect, as well as the structure of SERS
readout schemes, immunoassays for IgGs can be divided into
nanoprobes, which can generate strong SERS enhancement.
several categories, including ELISA,221−223 chemilumines-
Analyzing of IgGs can be largely automated and simplified
cence,224,225 colorimetry,226,227 fluorescence,228,229 SPR,230,231
with the assistance of microfluidic chips.142,245 Chon et al.
SERS,83,232 etc. The other immunoassays have been reviewed
developed a SERS-based gradient optofluidic sensor for fast and
well elsewhere.1,2,233−236 Here, we focus on SERS-based
sensitive immunoassay.232 The LOD for rabbit IgG was
immunoassays (Figure 20). SERS-based immunoassays can be
estimated to be 1−10 ng/mL, and the assay took less than
divided into two kinds. The first one uses the intensity of the
30 min to achieve the results because the experimental
SERS signal as the indicator for IgGs,232,237 while the second
procedures were automatically controlled with the microfluidic
one uses the frequency shift of SERS bands to detect the
chip. Recently, our group reported the detection of IgGs using
targets.39,238 The former kind can be subsequently divided into a 3D-barcode microfluidic chip.66 The LOD was found to be 10
two types, i.e., label-free SERS immunoassay239 and labeled fg/mL with a dynamic range from 10−8 to 10−14 g/mL. The
SERS immunoassay.83,240 Label-free SERS immunoassays are most outstanding advantage of this chip-based assay is its
relatively simple as compared with labeled SERS immuno- excellent multiplexing ability offered by spatial and SERS
assays, since they do not need a specially designed SERS encoding, which is well-discussed in detail in section 4. Typical
nanoprobe. However, labeled SERS immunoassays can offer a SERS-based immunoassays of IgGs are listed in Table 3.
higher sensitivity as compared with label-free SERS immuno-
assays due to the strong Raman signals offered by Raman 5.2. Study of Disease Biomarkers
reporter molecules. These features make SERS-based immuno- Early diagnosis of malignant disease (e.g., cancer) is important
assays suitable for IgG identification applications, such as for choosing appropriate therapeutic strategies and improving
immuno-phenotyping. the overall survival rates. Formerly, diagnosis of cancer had
Usually, SERS-based immunoassay can achieve quite low been strongly dependent on traditional approaches, such as X-
LODs and large dynamic ranges.241−243 For example, by using ray, computer tomography, endoscopy, ultrasonography,
magnetic beads and SERS nanobioprobes, Xiao et al. realized pathological section, and so on.256−261 However, these
7931 DOI: 10.1021/acs.chemrev.7b00027
Chem. Rev. 2017, 117, 7910−7963
Chemical Reviews Review

Figure 21. SERS immunoassay of disease biomarkers. (a) Rapid detection of HER2 using an ac-EHD microfluidic chip. Bifunctional fluorophore-
integrated gold/silver (F-SERS) nanoshells are used as the SERS probes. Reprinted with permission from ref 280. Copyright 2016 American
Chemical Society. (b) Aptamer-guided Ag−Au nanostructures for targeting of MUC1-overexpressing cancer cells. Reprinted with permission from
ref 287. Copyright 2012 American Chemical Society. (c) SERS-based immunoassay of tumor marker VEGF using DNA aptamers and silica-
encapsulated hollow gold nanospheres. Reprinted with permission from ref 271. Copyright 2013 Royal Society of Chemistry.

approaches are mainly focused on the anatomic form of tissues, including efficient Raman signal enhancing provided by
which usually lacks sensitivity toward early-stage tumors. aggregated Ag NPs. Wang et al. utilized an alternating current
Besides, these traditional diagnosis approaches are often electro-hydrodynamic (ac-EHD) microfluidic chip and SERS
accompanied by surgical procedures, which are harmful to the nanoprobes to realize the fast and sensitive detection of
patients and hinder repetitive and timely examinations. With HER2.279 Interestingly, in their design, ac-EHD-induced
the development of molecular and medical biology, researchers nanoscaled surface shear forces enhanced the capture kinetics
have found that tumor cells (or cells sensitive to tumors) can and thus greatly shortened the overall reaction time. The LOD
secret special biomarkers that can enter body fluids (e.g., blood, was found to be 10 fg/mL and the detection only took 40 min.
urine, ascites, and saliva). The presence of biomarkers in body Similarly, Zhou et al. also employed a typical sandwich-type
fluids is a valuable and prompt warning of health conditions. SERS immunoassay for the detection of cancer biomarkers.280
Hence, analyzing and screening of biomarkers is a powerful tool The silica-coated silicon NPs were used as the SERS
for early-stage diagnosis. Up to now, the discovered biomarkers nanoprobes, which were functionalized with specific antibodies.
related to cancers include proteins (e.g., receptors, cancer The immune substrate was fabricated by depositing Ag film
antigens), microRNAs (miRNAs), circulating DNA, and onto SiC sandpapers. Capturing antibodies were immobilized
CTCs.262−267 The most well studied biomarker species are onto the Ag films. With the presence of target biomarkers, the
proteins. Typical protein biomarkers include carcinoembryonic SERS nanoprobes linked to the Ag film via the formation of
antigen (CEA), AFP, human epidermal growth factor receptor sandwich-type immune complexes. Hence, the Raman signal of
2 (HER2), vascular endothelial growth factor (VEGF), PSA, the silicon NPs was enhanced and thus can be used as the
tumor suppressor p53, and epidermal growth factor receptor indicator of biomarkers. Separate detection of three biomarkers
(EGFR). Similar to IgG analysis, SERS-based immunoassays in human serum sample was realized, including PSA, AFP, and
have found prosperous applications in the detection of disease carbohydrate antigen 19-9 (CA19-9). The LODs were found to
biomarkers63,125,158,248,268−276 (Figure 21 and Table 4). More- be 1.79 fg/mL (PSA), 0.46 fg/mL (AFP), and 1.3 × 10−3 U/
over, the detection of biomarkers using SERS-based immuno- mL (CA19-9), respectively.
assay platforms only requires a little amount of body fluids and Aside from antibodies, the recognition of biomarkers can also
thus strongly reduces the injury toward patients and allows be accomplished by aptamers. Since their discovery, aptamers
repetitive examinations during therapy. This is crucial for have been well-known for excellent binding affinity toward their
prognosis evaluations and adjusting therapeutic strategies in targets (e.g., thrombin, peptides, and cells).281 DNA aptamers
time. provide a powerful candidate for the design of novel immune
5.2.1. Detection of Protein Biomarkers. The detection sensors with high performance. Up to now, aptamers have been
of protein biomarkers follows a similar principle as compared employed as the recognition unit in immunoassays based on
with the detection of IgGs, that is, using immuno-functionalized electrochemistry, SPR, and SERS. Electrochemistry- and SPR-
SERS nanoprobes to recognize these biomarkers.16,145,277,278 By based assays using aptamers have been reviewed else-
combining ELISA with SERS, Liang et al. presented a novel where.282−284 For SERS-based immune reactions using
immunoassay protocol for the evaluation of PSA in whole aptamers, the detection scheme is quite similar to those using
serum and urine.111 The introduction of aggregated Ag NPs antibodies.285,286 Wu et al. fabricated an aptamer-guided Ag−
resulted in a quite impressive sensitivity in the ultralow Au nanostructure using photoreduction.287 To generate SERS
concentration range (10−9−10−6 ng/mL). The high sensitivity signals, Rh6G is attached to the nanostructure. The aptamer
of this protocol is attributed to multistage signal amplifications, they used is S2.2, which is a 25 base oligonucleotide specific for
7932 DOI: 10.1021/acs.chemrev.7b00027
Chem. Rev. 2017, 117, 7910−7963
Chemical Reviews Review

Table 4. SERS-Based Immunoassays for Biomarkers


recognition
refs biomarkers unit detection platform dynamic range LOD
305 A1AT antigen antibody 10−1000 ng/mL 10 ng/mL
129 aflatoxin B1 (AFB1) DNA Au NS 1−1000 pg/mL 0.48 pg/mL
63 α-fetoprotein (AFP) antibody gold array 0−10 ng/mL 0.1 ng/mL
269 angiogenin antibody gold-patterned microarray chip 10−4−10−12g/mL 0.1 pg/mL
145 anthrax biomarker poly-γ-D- antibody magnetic beads 100 pg/mL to 100 μg/mL 100 pg/mL
glutamic acid (PGA)
137 antigen gp51 anti-gp51 MNP-Au 0.95 mg/mL
112 CA15-3 antibody 0.1−500 U/mL for CA15-3 antigens 0.99 U/mL
CA27-29 0.1−500 U/mL for CA27-29 antigens 0.13 U/mL
CEA 0.1−500 ng/mL for CEA antigen 0.05 ng/mL
40 cetuximab antibody MDA-MB-468 breast cancer cells 0−2000 ng/mL 1 ng/mL
115 CFP-10 antibody 1 pg/mL to 1 μg/mL 0.0511 pg/mL
118 cholera toxin (CT) antibody silica microspheres 1100 ng
303 ctDNA DNA single-walled carbon nanotubes 10 fM to 1 nM 0.3 fM
306 detection of polar antibiotics Ag−graphene/electrode
304 DNA DNA magnetic NPs
307 dopamine aptamer 0.01−10pM 0.006 pM
308 EGF, ErbB2, IGF-1 antibody
309 EGFR antibody photonic crystal fiber (PCF) 100 pg in 10 nL
310 EGFR antibody
279 EGFR2 antibody gold-coated glass chips 1 fg/mL to 1 ng/mL 10 fg/mL
130 folic acid antibody Ag NPs 0.005−1 ng/mL 0.86 pg/mL
113 glycoproteins boronate- 0.1 ng/mL to 100 μg/mL (HRP), 1 3 nM (HRP), 0.7 nM
affinity ng/mL to 10 μg/mL (AFP) (AFP)
311 heparin protamine 0.5−150 ng/mL 0.5 ng/mL
147 hepatitis B virus anti-HBsAg SERS-active substrate 0−625 IU/mL 0.01 IU/mL
312 HIV-1 DNA DNA lateral flow assay 64 ng/mL ∼ 8 pg/mL 0.24 pg/mL
136 hormone estradiol E2 anti-E2 magnetic NPs 0.1−1000 pg/m 0.65 pg/mL
antibody
248 human IgG antibody gold triangle nanoarray 0.1 pg/mL to 10 ng/mL 7 fg/mL
313 IL6 antibody Au 10 ng to 1 μg 10 ng
270 IL6/TNFα antibody nitrocellulose membrane 0−5 ng/mL 7.15 pg/mL
0−14 ng/mL 10.7 pg/mL
288 latent fingerprints aptamer glass substance
39 liver cancer biomarkers α- antibody Ag island film 5.0 pM
fetoprotein and glypican-3
314 lung cancer markers antibody
315 microcystin-LR (MC-LR) DNA Au NFs 0.01−100 nM 8.6 ± 0.4 pM
300 miR-203 DNA 10−14 −10−7 M 6.3 fM
294 mir-21 miRNA gold slide 4.44−1480 nM 0.36 nM
7.40−1480nM 0.85 nM
291 miRNA miRNA silver nanorod arrays
297 miRNA miRNA Silica microbeads 1 fM to 10 pM 0.3 fM
268 MMP-7 and CA19-9 antibody Au/glass and mask to generate 2.28 and 34.5 pg/mL
array
316 mouse double minute peptide 1.5 nM
(MDM2) protein
287 MUC1 aptamer breast cancer cell
274 N-terminal pro-brain antibody CoFe2O4 1 fg/mL to 1 ng/mL 0.75 fg/mL
natriuretic peptide
273 platelet-derived growth factor DNA Au film (1.0 × 10−12)−10−8 M 0.42 pM
B-chain (PDGF-BB)
317 prostate-specific antigen (PSA) DNA 0−4 ng/mL 0.5 ng
318 proteases peptides AuNPs 0−100 nM 85 fM
319 protein antibody 0−(3.4 × 105) ng/mL 10 zM
320 protein antibody 146 ng/L
74 PSA antibody silicon (microcontact printing) 1 pg/mL 1 pg/mL
111 PSA and the adrenal stimulant antibody 96-well plates 10−9 and 10−6 ng/mL
ractopamine (Rac)
280 PSA/AFP/CA19-9 antibody silver NPs 1.79 fg/mL, 0.46 fg/mL,
1.3 × 10−3 U/mL
276 RGG9 domain of nucleolin AS1411 G- split-ring resonators 10−13−10−5 M 10−13 M
quadruplex

7933 DOI: 10.1021/acs.chemrev.7b00027


Chem. Rev. 2017, 117, 7910−7963
Chemical Reviews Review

Table 4. continued
recognition
refs biomarkers unit detection platform dynamic range LOD
321 telomerase DNA electrodes 5−1000 Hela cells 5 cells
322 telomerase DNA sensor surface 1 tumor cell/1000
background cells
323 telomerase DNA TS-modified AuNPs 0−100 Hela cell/μL 1 Hela cell/μL
324 telomerase DNA porphyrin graphene nanocomposite 0−5000 cells/mL 10 Hela cells/mL
electrode
78 telomerase TS primers 1 cell/mL
325 telomerase TS primers MB@Au@TS 0−1000 cells/mL 1 cell/mL (SERS), 10 cell/
mL (colorimetry)
326 telomerase DNA Ag substrate
327 telomerase DNA
328 telomerase DNA silica microbeads (SiMBs) single HeLa cell
329 thrombin peptides 160 fM
330 thrombin ATP MB 0.27 pM
117 vasopressin molecules ATP gold-decorated silicon nanopillars 1 pM to 1 nM 1 pm/mL
271 VEGF ATP/VEGF gold microarray 0.1−1000 ng/mL 1−10 pg/mL
antibody
331 vitellogenin (Vg) antibody nanosculptured thin films (nSTFs) 5 pg/mL to 25 ng/mL 5 pg/mL
of silver on Si substrates
332 WNV and RVFV antigen antibody magnetic NPs 5 fg/mL (PBS), 25 pg/mL
(FBS)

MUC1 mucin. MUC1 is a transmembrane glycoprotein and is divided into three kinds: label-free detection,291−293 direct
overexpressed in primary and metastatic breast cancers. By hybridization detection,294−296 and signal amplification detec-
measuring the SERS signals on cells, it was found that the tion.297−299 Label-free detection of miRNAs is conducted by
aptamer-guided nanostructure can specifically bind to MUC1- enhancing the Raman signals of RNA strands via SERS
overexpressing cancer cells. Moreover, the nanostructures substrates, which is relatively simple and thus will not be
bound on cancer cell membranes can be subsequently used discussed in detail here. Direct hybridization detection of
as the NIR absorber and heat generator for photothermal miRNAs is realized by using complementary DNA capturing
therapy. Ko et al. reported a SERS-based immunoassay of strands. Signal amplification detection of miRNAs is accom-
tumor marker VEGF using DNA aptamers.271 They used a plished by the addition of enzymes or complementary strands
gold-patterned microarray chip as the immune substrate, and to exaggerate the signals.
capturing antibodies were immobilized on the chip. They also Guven et al. reported direct hybridization detection methods
fabricated silica-encapsulated hollow gold nanospheres as the for mir-21.294 They employed two detection schemes. The first
SERS nanoprobes. VEGF-specific aptamers were decorated on one is to immobilize the target mir-21 on a gold slide, then
the SERS nanoprobes. Consequently, a sandwich-type immune SERS nanoprobes decorated with mir-21 complementary
complex can be formed by the capturing antibody, target strands can hybridized with the mir-21. The second detection
VEGF, and SERS nanoprobes. As compared with conventional scheme is a sandwich-type assay. A DNA strand that is
ELISA, the dynamic range of the aptamer-based assay was 3−4 complementary to one end of mir-21 is immobilized on the
orders of magnitude larger and the detection sensitivity was 2− gold slide. This DNA strand is used to capture mir-21. Then a
3 orders of magnitude higher. Zhao et al. fabricated a SERS SERS nanoprobe decorated with another DNA strand that is
nanoprobe for the imaging of fingerprints.288 Aptamers were complementary to the rest of the mir-21 can hybridize with it.
decorated on the surface of the nanoprobes to recognize and Thus, a sandwich-type structure is formed by the capturing
bind lysozyme in the fingerprints. Owing to the fine sensitivity strand, mir-21, and SERS nanoprobe, which is similar to the
of SERS and the excellent selectivity of aptamers, high- immune complex formed in sandwich-type immunoassays. In
resolution imaging of fingerprints was realized. Moreover, such both detection schemes, the SERS intensity of the hybridized
an aptamer-based SERS imaging technique does not require nanoprobes can be used to report the concentration of mir-21.
complicated pre- or post-treatment and can be used to detect The LOD of the first scheme was 0.36 nM and that of the
various types of fingerprints. second scheme was 0.85 nM. Besides, the assay only took less
5.2.2. Detection of miRNAs and Circulating DNAs. than 40 min with a final performance comparable to that of
miRNAs are short (19−25 nucleotides) noncoding RNAs that PCR. As an example of signal amplification assays of miRNAs,
are important for the regulation of genes. miRNAs are Zheng et al. presented an enzyme-free quadratic SERS signal
considered as an important kind of biomarker since they are amplification approach for the detection of circulating miRNAs
closely related with the development of cancerous dis- in human serum.297 Signal amplification was realized through
eases.289,290 Detection of miRNAs can be accomplished by the combination of a miRNA-triggered hybridization chain
traditional methods such as PCR, Northern blotting, and reaction and Ag+-mediated cascade amplification. An ultrahigh
microarrays. However, these methods suffer from some sensitivity (LOD is 0.3 fM) toward miRNA was achieved with
shortcomings, such as large sample amount, complicated this approach. Moreover, this approach can also be used for the
primer design, and cross hybridization. Recently, SERS has direct detection of miRNAs in serum collected from patients
also been applied to realize the facile and sensitive detection of with different stages of chronic lymphocytic leukemia. The
miRNAs. Generally, SERS-based detection of miRNAs can be results correspond well with those determined by qPCR. Zhang
7934 DOI: 10.1021/acs.chemrev.7b00027
Chem. Rev. 2017, 117, 7910−7963
Chemical Reviews Review

Figure 22. SERS-based telomerase activity detection and telomere length measurement. (a) TEC-SERS method for ultrasensitive detection of
telomerase. Reprinted with permission from ref 78. Copyright 2013 Wiley-VCH Verlag GmbH & Co. KGaA. (b) In situ sensing of cellular
telomerase using SERS. Reprinted with permission from ref 340. Copyright 2016 WILEY-VCH Verlag GmbH & Co. KGaA. (c) SERS based in situ
hybridization (SISH) for telomere length measurement. Reprinted with permission from ref 327. Copyright 2016 American Chemical Society.

et al. reported a cascade amplification and SERS-based targets for the design of anticancer drugs.333,334 Telomerase
detection method for miR-203, which is an important activity detection and telomere length measurement can
biomarker for tumor growth and progression.300 The method provide important information for the diagnosis and
was realized with the assistance of a specially designed therapeutics of cancerous diseases. Telomerase activity
multifunctional nucleic acid probe as well as the nicking detection methods include telomere repeat amplification
endonuclease and polymerases. The detection limit of miR-203 protocol (TRAP),335,336 electrochemical method,321 SPR,322
was found to be 6.3 fM. colorimetry,323 and chemiluminescence.324 Telomere length
Circulating DNAs are found in peripheral blood of patients measurement methods include Southern blotting, hybridization
and can reflect the genomic state of cancerous disease.301 In the protection assay,337 qPCR,338 and fluorescent in situ hybrid-
past year, SERS has just begun to be employed for the analysis ization (FISH).339
of circulating DNAs.302 Zhou et al. presented a SERS-based SERS-based detection of telomerase activity is usually based
approach for the detection of circulating DNAs in human on the fact that telomerase can enlongate telomere repeats and
blood.303 A triple-helix molecular switch (THMS) was used as thus induce a SERS signal variation. Our group was the first to
the recognition element for circulating DNAs. With the aid of use a SERS technique for the detection of telomerase activity
RNase HII, numerous T-rich strands of the THMS will be and telomere length measurement (Figure 22). We reported
released in the presence of target circulating DNAs and the telomerase elongation controlled SERS (TEC-SERS)
subsequently be absorbed on single-walled carbon nanotubes method for ultrasensitive detection of telomerase.78 TEC-
(SWNTs) via π−π stacking interactions. Then the T bases on SERS utilizes the fact that SERS is highly sensitive to the
SWNTs can facilitate the in situ growth of copper nano- aggregation state of metal nanoparticles. Telomerase-sensitive
particles, which can enhance the Raman signals of SWNTs. SERS nanoprobes were fabricated by decorating spherical gold
Hence, the intensity of SERS signals of SWNTs indicates the nanoparticles with telomerase substrate primer (TS primer)
concentration of target circulating DNAs. Wee et al. reported a and Raman molecules. Then the SERS nanoprobes were used
multiplex detection of mutations in circulating tumor DNA to detect the telomerase activity of tumor cells. The detection
using SERS.304 They first amplified three melanoma mutations limit of TEC-SERS is 2 orders of magnitude lower than that of
with PCR, and then mutation-specific SERS nanotags were other methods, which is beneficial for the ultraearly diagnosis of
hybridized with the PCR products. Finally, the SERS-nanotag- malignant tumors. Besides, no PCR amplification is needed in
labeled PCR products were captured by magnetic beads. The the TEC-SERS method, which greatly simplifies the detection
presence or absence of melanoma mutations in circulating procedure, avoids false negative results, and improves reliability.
DNAs can be determined by measuring the SERS spectra. This Later, we also presented a colorimetry- and SERS-based
SERS-assisted method can reproducibly detect as few as 0.1% telomerase activity screening method.325 Magnetic capturing
(10 copies, CV < 9%) of target sequences. nanoparticles with TS primer anchored on their surfaces and
5.2.3. Detection of Telomerase Activity and Telomere SERS nanoprobes with telomere complementary strand
Length Measurement. Aside from the above-mentioned attached to their surfaces were synthesized. By monitoring
common biomarkers, telomerase is another important bio- the color and SERS signal of the final sediments, both
marker. The overexpression of telomerase and abnormal colorimetry- and SERS-based telomerase activity detection was
variation in telomere length are closely related with carcino- achieved. This method simultaneously holds the merits of
genesis, which makes telomerase and telomere new therapeutic colorimetry and SERS, i.e., fast screening and high sensitivity.
7935 DOI: 10.1021/acs.chemrev.7b00027
Chem. Rev. 2017, 117, 7910−7963
Chemical Reviews Review

Figure 23. SERS assays of bacteria and viruses. (a) Label-free SERS assay of bacteria using vancomycin-coated Ag-nanoparticle arrays. Reprinted
with permission from ref 345. Copyright 2011 Macmillan Publishers Ltd. (b) Microfluidic dielectrophoresis device for rapid online detection of a
single bacterium. Bacterial cells are recognized by SERS nanoprobes decorated with antibodies. Reprinted with permission from ref 353. Copyright
2014 Wiley-VCH Verlag GmbH & Co. KGaA. (c) Schematic illustration of SERS enzyme-catalyzed immunoassay of respiratory syncytial virus.
Reprinted with permission from ref 355. Copyright 2016 Elsevier B.V.

Thus, efficacious and sensitive screening of numerous samples embedded beads. Hence, these SERS nanoprobes can be
can be realized. The detection limits for SERS and colorimetry bound to the target bacteria via the immune reaction between
were 1 tumor cell/mL and 10 tumor cells/mL, respectively. In the antibodies and proteins on the bacteria’s surface. With the
addition, two other research groups also started to work on assistance of a microfluidic chip, the total assay time was
SERS-based telomerase activity detection. Xu et al. reported in significantly reduced (<2 h). Besides, the amount of antibodies
situ sensing of cellular telomerase using SERS and multigap- used in the SERS-based assay was 100-fold less than that used
embedded nanoassemblies.340 Shi et al. presented a quadratic in the traditional ELISA. The LOD was found to be 70 cfu/mL.
SERS signal amplification based method for telomerase Neng et al. reported a multiplex immunoassay for the surface
detection with LOD down to the single-cell level.328 envelope and capsid antigens of the viral zoonotic viruses.332
For measuring telomere length, we presented two SERS- Detection was mediated by immune recognition using SERS
based strategies.326,327 The typical one is called SERS-based in nanoprobes and paramagnetic nanoparticles conjugated with
situ hybridization (SISH).327 The SISH method uses two kinds antibodies. The LOD was 5 fg/mL in PBS and 25 fg/mL in
of SERS nanoprobes to hybridize in situ with telomeres and fetal bovine serum. Li et al. presented a SERS enzyme-catalyzed
centromeres, respectively. Telomere length is evaluated using a immunoassay of respiratory syncytial virus.125 The combination
redefined telomere-to-centromere ratio (T/C ratio). The of ELISA and SERS produced an impressive LOD of 0.05 pg/
calculation method for the T/C ratio in the SISH method is mL, which is 20 times lower than that of a colorimetric assay.
more reliable than that in FISH. The SISH method can be used In addition to the identification of whole virus particles,
to analyze single telomeres. These features make SISH an researchers also focused on the investigation of virus DNAs
excellent alternative strategy for telomere length measurement. using SERS techniques. Here, the recognition of target virus
5.3. Identification of Bacteria and Viruses DNAs is fulfilled by their complementary DNA strands
modified on the surfaces of SERS nanoprobes or nanotags.354
Detection of bacteria and viruses is crucial in food safety and For example, Fu et al. developed a SERS-based lateral flow
surveillance of epidemic diseases.9,341,342 The complexity of the assay biosensor for a highly sensitive detection of HIV-1
samples makes SERS quite suitable for the identification of DNA312 (Figure 24). The SERS nanotags were fabricated by
bacteria and viruses, since SERS holds a fine endurance toward attaching Raman reporters and target complementary DNA
biological background interferences (Figure 23). Label-free strands onto gold nanoparticles. Capturing DNA strands were
SERS strategy has been well applied in the identification of immobilized on the lateral flow strip. With the target HIV-1
bacteria and viruses, which is realized by directly analyzing the DNA present, a sandwich structure was formed by the
SERS spectra of bacteria and viruses.102,343−345 Here, we mainly capturing strand, HIV-1 DNA, and SERS nanoprobe. Hence,
focus on labeled SERS immunoassays of bacteria and viruses, SERS signals can be used to identify the HIV-1 DNA. The
which usually rely on the specific recognition of proteins of detection limit of the target DNA was found to be 0.24 pg/mL.
bacteria and viruses by SERS nanoprobes.147,346−352 Lin et al.
reported a microfluidic dielectrophoresis device for a rapid 5.4. Detection of Ions and Toxins
online detection of single bacterium353 (Figure 23b). They SERS-based immunoassays can also be used to detect metal
fabricated SERS nanoprobes by immobilization of bacteria- ions and toxins (Figure 25). Especially, monitoring of heavy
specific antibodies onto the surfaces of nanoaggregate- metal ions and toxins is important for environmental
7936 DOI: 10.1021/acs.chemrev.7b00027
Chem. Rev. 2017, 117, 7910−7963
Chemical Reviews Review

or DNAs/aptamers.118,356−360 Li et al. presented a strategy for


the detection of aflatoxin B1.129 They constructed a SERS
nanosensor using aflatoxin B1 aptamer-modified Ag satellites
and complementary-strand-modified Au nanospheres. Aflatoxin
B1 can destroy the core−satellite structured nanosensor due to
its strong binding affinity toward the aptamer and induce a
significant reduction in SERS intensity. The linear detection
range was from 1 to 1000 pg/mL with a detection limit of 0.48
pg/mL. Yang et al. reported a magnetic-separation-assisted
competitive SERS immunoassay for chloramphenicol.369 A
competitive immune reaction happened between free chlor-
amphenicol and chloramphenicol-modified SERS nanotags.
The dynamic range of this assay was from 1 to 1 × 104 pg/mL
and the LOD was 1 pg/mL.
Up to now, SERS has been used to detect ions including
Hg2+, Ag+, Pb2+, and Cd2+. As a representative, mercury ion
(Hg2+) has been thoroughly investigated using SERS-based
immunoassays.361−364 It can be recognized with antibodies or
T−Hg2+−T base pairs. Wang et al. presented a SERS-based
competitive immunoassay of Hg2+ on a glass substrate.365 The
LOD was 8 × 10−5 ng/mL. Later, She et al. developed a similar
competitive immunoassay for an ultrasensitive detection of
Hg2+ in water, human serum, and urine samples.366 The assay
Figure 24. SERS-based lateral flow assay biosensor for a highly was conducted on an immuno-chromatographic test (ICT)
sensitive detection of HIV-1 DNA. (a) Schematic illustration of the strip. The immunoprobe was fabricated by attaching Raman
lateral flow biosensor. (b) Working principle of the SERS-based lateral reporters and Hg2+ antibodies onto the surfaces of gold
flow chip. Reprinted with permission from ref 312. Copyright 2016 nanoparticles. The competitive binding of immunoprobes to
Elsevier B.V.
free Hg2+ or immobilized Hg2+ can cause a difference in the
SERS signals on the ICT strip; thus, SERS signals can be used
maintenance and public health. Specific SERS-based immune to determine the concentration of Hg2+ with a LOD of 0.45 pg/
detection of ions and toxins is commonly realized by antibodies mL.

Figure 25. SERS immunoassays for ions and toxins. (a) SERS-based competitive immunoassay for mercury ions. Reprinted with permission from ref
365. Copyright 2014 The Royal Society of Chemistry. (b) Gold nanohole array based SERS biosensor for detection of silver and mercury ions in
human saliva. Reprinted with permission from ref 114. Copyright 2015 The Royal Society of Chemistry. (c) Competitive SERS immunoassay for
accurate and sensitive detection of chloramphenicol with the assistance of magnetic beads and SERS probes. Reprinted with permission from ref 369.
Copyright 2016 Elsevier B.V.

7937 DOI: 10.1021/acs.chemrev.7b00027


Chem. Rev. 2017, 117, 7910−7963
Chemical Reviews Review

Table 5. SERS-Based Immunoassay of Ions and Toxins


refs ions recognition unit detection platform dynamic range LOD
361 Hg2+ ATP Au@Ag 10 pM
362 Hg2+ DNA GNS dimer 0.002−1 ng/mL 0.8 pg/mL
364 Hg2+ DNA Au NR oligomer 5−5000 pM 4.3 pM
366 Hg2+ antibody nitrocellulose (NC) membrane 0.12 ng/mL to 0.45 pg/mL 0.45 pg/mL
370 Hg2+ DNA AuNPs/rGO/SiO2/Si 0.1 nM
371 Hg2+ DNA Ag@AuNPs 5 pM to 50 nM 5 pM
368 Hg2+ DNA AuNPs@SiNWs array 1 pM to 100 nM 7.3 × 10−13 M
365 Hg2+ antibody GNP 10−5−100 ng/mL 8 × 10−5 ng/mL
114 Hg2+, Ag+ DNA a gold nanohole array 1.7 pM
372 Hg2+, Ag+ DNA MSS@Au/Au 0.1−1000 nM (Hg2+), 10−1000 nM 2 nM (Hg2+), 10 nM
(Ag+) (Ag+)
363 Hg2+, Cu2+ DMT and TMT AuNR on fiber 10−2−10−6 M Hg2+, 1−100 μM Cu2+ 1 μM
373 Pb2+ DNA AuNP 16.7−666.7 nM 16.7 nM
374 Pb2+ none DNAzyme 20 nM to 1 μM 20 nM
375 Ag+ 2MNA AuNP (5 × 10−8)−(1.7 × 10−5) M 2.5 × 10−8 M
376 Cd2+ ATRP AuNP 1.0−25 μM 1.0 μM
377 Zn2+ 4- AgNP 0.1−10 mM 0.1 mM
mercaptopyridine
378 malachite green label free Fe3O4/Ag core/shell 10−350 ppb 10 ppb
379 aromatic molecules label free Au/AgNP-rGO 1−10 nM 1 nM
380 BPE pyridyl units paper substrate adsorbed with 0.1−5 nM 0.1 nM
nanorods
381 CV molecules label free wrinkled nanoporous gold films
129 aflatoxin B1 DNA AuNS 1−1000 pg/mL 0.48 pg/mL
369 chloramphenicol antibody magnetic NPs 1−(1 × 104) pg/mL 1.0 pg/mL
382 ochratoxin-A ATP gold nanotriangles
359 zearalenone antibody Au 1−1000 pg/mL 1 pg/mL
383 kanamycin antibody Fe3O4 2 pg/mL to 80 ng/mL 2 pg/mL
384 melamine DNA AuNP 1.0 pg/mL to 10 ng/mL 1.0 pg/mL
356 staphylococcal aptamer Fe3O4@Au 2.5 fM to 3.2 nM 224 aM
enterotoxin B

Besides antibodies, the detection of ions can also be achieved nanoprobes/nanotags, which are decorated with cell-specific
by DNA strands.367 For example, silver ion can be detected via recognition units (e.g., peptides, antibodies, or aptamers).
the formation of the C−Ag+−C pair, and mercury ion can be Considering aptamers, Tan’s group has performed serial studies
detected via the formation of the T−Hg2+−T pair. Zheng et al. on how to generate aptamers for cell recognition.108,109,395
reported a gold nanohole array based SERS biosensor to detect Usually, when selecting aptamers, purified proteins are used as
Ag+ and Hg2+.114 They fabricated SERS nanoprobes using the targets. Tan et al. found that it was more accurate to directly
silica-coated gold nanostars. Two different kinds of single- use the whole cell as the target when selecting aptamers for
strand DNAs were attached on the nanohole array and cells (cell-SELEX), since free proteins might take different
nanoprobe, respectively. If Ag+ or Hg2+ is present, the two steric configurations as compared to proteins on cell
kinds of single-strand DNAs will hybridize through the membranes.
formation of C−Ag+−C or T−Hg2+−T pairs. As a result, the With cell-specific ligands, the SERS nanoprobes/nanotags
SERS nanoprobe will get very close to the nanohole array, and help to determine the species and expression levels of proteins
a huge Raman enhancement will be generated, leading to a on the cell surfaces, which eventually contributes to the
strong SERS signal. Sun et al. used gold-nanoparticle-decorated identification of cells. For example, Wang et al. reported the
silicon nanowires as the SERS substrate for the detection of successful detection of CTCs in human peripheral blood using
Hg2+.368 A Raman-reporter-tagged single-strand DNA was used SERS nanoparticles.396 The CTCs specific SERS nanoparticles
as the Hg2+ recognition unit. Hg2+ can bend the single-strand were constructed by linking EGF peptides and Raman reporters
DNA via T−Hg2+−T pairs and pull the Raman reporter close onto gold nanoparticles. With EGF peptides on their surfaces,
to the SERS substrate, resulting in strong SERS signals. The
the SERS nanoparticles can bind to EGFR-overexpressing
LOD was 7.3 × 10−13 M. There are quite a few works using
CTCs via immune recognition. They found that the SERS
similar working principles to detect ions and they are
nanoparticles can rapidly detect CTCs from peripheral blood of
summarized in Table 5.
patients with squamous cell carcinoma of the head and neck.
5.5. Sorting of Cells Similarly, Bhana et al. fabricated an iron oxide−gold nano-
Sorting of cells is another prosperous application of SERS- particle for the detection of SKBR3 cells from whole blood.397
based immunoassays, especially in the detection and classi- The magnetic and SERS-active nanoparticle is decorated with
fication of tumor-derived cells (e.g., CTCs) (Figure SKBR3-cell-specific antibodies to allow capturing of these cells.
26).310,385−387 Similar to the detection of other targets, cells The LOD of this strategy was 1−2 cells/mL of blood. Song et
are differentiated by various proteins on their surfaces.388−394 A al. developed a SERS-encoded nanogapped plasmonic nano-
necessity for SERS-based immune sorting of cells is SERS particle for the identification of cancer cells.93 The nanoparticle
7938 DOI: 10.1021/acs.chemrev.7b00027
Chem. Rev. 2017, 117, 7910−7963
Chemical Reviews Review

Another kind of in vivo tumor targeting using nanoparticles


(with no specific ligands) is “passive” tumor targeting, which is
usually realized by the enhanced permeability and retention
(EPR) effect. Passive tumor targeting is well-reviewed else-
where and will not be discussed in detail in this review.408,409
Dinish et al. reported in vivo active targeting of three breast
cancer biomarkers (i.e., EGFR, CD44, and TGFβRII) using
SERS nanoprobes decorated with antibodies.410 Wang et al.
presented an endoscopic imaging strategy for detecting
gastrointestinal cancers. In their strategy, the molecular
phenotype of tumor tissues is visualized by SERS nanoprobes
decorated with anti-EGFR or anti-HER2 antibodies.411 Besides,
SERS nanoprobes (passively or actively targeted) can also
facilitate surgical tumor resection by identifying and imaging
millimeter/submillimeter-sized tumors using SERS sig-
nals.412−414 In vivo targeting of tumors using SERS nanoprobes
is quite beneficial, since the fingerprint characteristic of SERS
enables their precise tracking in complicated biological
environments. However, the complex in vivo circumstances
bring more rigorous requirements for the SERS nanoprobe’s
targeting ability. This is because proteins in blood or other
body fluids tend to adsorb on the SERS probes, forming a
protein “corona”, which can severely disrupt the recognition
between targeting ligands and biomarkers.415,416 To avoid the
formation of a protein corona, SERS nanoprobes for in vivo
applications are usually decorated with an outer passivation
layer using polyethylene glycol, which can effectively resist
protein adsorption as well as improve the stability of SERS
nanoprobes.417
5.6. Multiplex Biochemical Analysis Using SERS Encoders
Figure 26. SERS immunoassays for cells. (a) Detection of CTCs in
human peripheral blood using SERS probes. Reprinted with The most extraordinary advantage of the SERS technique is its
permission from ref 396. Copyright 2011 American Association for excellent multiplexing ability.7,70,418−422 The sharp bands of
Cancer Research. (b) Identification of rare cancer cells assisted by a Raman spectra make SERS an ideal candidate for optical
microfluidic channel and SERS. Reprinted with permission from ref encoding, which can further facilitate multiplex and high-
401. Copyright 2015 American Chemical Society. throughput immuno-detection. SERS-based optical encoding
has been explained in detail in section 4. Here, we will provide
possesses a sub-2-nm inner gap, which can generate a strong several typical immuno-detections based on SERS-encoded
Raman enhancement. By modifying the nanoparticle’s surfaces nanoprobes and platforms, which can better prove the excellent
with MUC-1 aptamers, the nanoparticle can target MUC-1- multiplexing ability of SERS (Figure 27, 28). Up to now, SERS-
overexpressing cancer cells and realize SERS imaging of these encoded nanoprobes (i.e., SERS encoders) have been
cancer cells. employed in the analyses of several different kinds of biological
By combining SERS-based immunoassays with microfluidic or chemical targets, including DNAs, proteins, bacteria, cells,
chips, efficient cell sorting can be realized.398−400 For instance, and ions.15,60,160,423−426
Pallaoro et al. reported a rapid identification of prostate cancer 5.6.1. Multiplex Immuno-Detection of Biomolecules
cells using microfluidic channel and SERS biotags.401 Neuro- and Cells. In the past few years, our group has been working
pilin-1-specific peptides were attached on the SERS biotags, on SERS-based multiplex bioanalyte detection. We introduced
making these biotags specific for neuropilin-1-overexpressing a SFJSE method using specially designed nanoencoders.17
cancer cells. When incubated with cancer cells, the SERS These SFJSE-based nanoencoders have been employed for the
biotags can bind to the target cells. These cancer cells together immuno-detection of proteins and cells.61,135 For example, we
with adsorbed biotags were flowed through a microfluidic fabricated SERS−fluorescence dual-encoded magnetic nano-
channel, and the cancer cells were identified by the SERS beads and used them to separate rare cancer cells from
signatures. This microfluidic-chip-based assay can detect one enormous normal cells.135 By decorating the surfaces of the
target cancer cell out of 100 control cells. dual-encoded nanobeads with cancer-cell-specific antibodies,
In addition to detecting protein biomarkers in solutions or we can efficiently capture four different kinds of cancer cells
on cell membranes, SERS probes can also be employed for in from phosphate-buffered saline or human blood samples. We
vivo targeting of tumors. Up to now, in vivo tumor targeting also developed a 3D SERS barcode microfluidic chip and
experiments were all performed on animals (e.g., mouse realized the simultaneous detection of multiple kinds of
xenografts).402−406 SERS probes are decorated with tumor- proteins.66
specific ligands (e.g., antibodies) and injected into the animal Zhao et al. fabricated SERS-encoded nanotags for the
models for “active” tumor targeting. That is, the injected SERS detection of three different kinds of virus genes.92 The
probes can target and bind to tumor sites via recognition nanotags that contain nanogaps can generate SERS enhance-
between the targeting ligands and tumor biomarkers.407 ment. Raman reporter molecules are placed inside these
7939 DOI: 10.1021/acs.chemrev.7b00027
Chem. Rev. 2017, 117, 7910−7963
Chemical Reviews Review

Figure 27. Multiplex SERS immunoassays. (a) Multiplex immuno-phenotyping and imaging of cancer cell receptors using Au@pNIPAM SERRS
tags. Reprinted with permission from ref 429. Copyright 2015 Wiley-VCH Verlag GmbH & Co. KGaA. (b) Sensitive multiplex biomarker detection
using SERS-encoded silver pyramids. Reprinted with permission from ref 131. Copyright 2015 Wiley-VCH Verlag GmbH & Co. KGaA. (c)
Multiplex virus DNA detection using gold nanostructures containing nanogaps. Reprinted with permission from ref 93. Copyright 2014 American
Chemical Society. (d) Multiplex protein detection using antibody-functionalized SERS-encoded nanoparticle assemblies. Reprinted with permission
from ref 430. Copyright 2014 Wiley-VCH Verlag GmbH & Co. KGaA.

nanogaps to provide SERS signals. By using different reporter


molecules, different SERS signatures can be obtained, thus
realizing SERS encoding. The surfaces of the SERS tags are
decorated with target-specific DNA strands. Hepatitis A virus
(HAV) Vall7 polyprotein gene, hepatitis B virus (HBV) surface
antigen gene, and human immunodeficiency virus (HIV)
antigen gene are simultaneously detected using the SERS
nanotags and magnetic microparticles. The LODs of HAV,
HBV, and HIV were 0.39, 0.18, and 0.51 pM with a good
dynamic range of 5 orders of magnitude. Similar applications in
multiplex DNA detections have also been reported by other
groups.
Xu et al. reported SERS-encoded silver pyramids for the
simultaneous and ultrasensitive detection of multiple protein
biomarkers, including MUC1, PSA, and thrombin.131 Recog-
nition units for these biomarkers are aptamers. With target
biomarkers presented, the specific biorecognition between the
biomarker and the aptamer will alter the 3D geometries of the
pyramids and cause Raman signal enhancement. LODs were
0.96, 85, and 9.2 aM with detection ranges of 1−500 aM, 0.1−
50 fM, and 0.01−5 fM for PSA, thrombin, and MUC1,
respectively. Wang et al. presented a multiplex homogeneous
immunoassay for the sensitive detection of proteins.427 The
SERS nanoparticles are attached with Raman reporters and
antibody fragments. The immune recognition between anti-
Figure 28. Multiplex cell imaging using SERS probes. The Raman body fragments and target proteins closely assembles the SERS
reporters are alkyne-containing molecules. The SERS probes are nanoparticles and results in strong SERS signals. Reproducible
decorated with ligands that target cancer cells. Reprinted with detection and quantification of three cytokines (i.e., interferon
permission from ref 431. Copyright 2016 American Chemical Society. γ, interleukin-2, and tumor necrosis factor α) are achieved.
Tang et al. presented a SERS-frequency-shift immunoassay for
7940 DOI: 10.1021/acs.chemrev.7b00027
Chem. Rev. 2017, 117, 7910−7963
Table 6. Multiplex SERS Assays
multiplexing recognition
refs targets ability detection platform unit dynamic range LOD
92 virus genes HAV, 3 magnetically separated DNA 5 orders of LOD 0.39 pM
HBV, (liquid phase) 0.18 pM
HIV 0.51 pM
Chemical Reviews

268 biomarkers MMP-7, 2 metal substrate antibody 2.28 pg/mL


CA19-9 34.5 pg/mL

434 biomarkers p53, p21 2 glass antibody 5 ng/mL to 500 fg/mL 1 pg/mL

131 biomarkers PSA, 3 Ag pyramids in liquid aptamer 1−500 aM 0.96 aM


thrombin, phase 0.1−50 fM 85 aM
mucin-1 0.01−5 fM 9.2 aM

435 thrombin, PDGF BB, Ig E 3 magnetically separated aptamer 10 pg/mL to 5.0 ng/mL 3.2 pg/mL
(liquid phase)

428 plant pathogens Botrytis cinerea, Pseudomonas 3 magnetically separated DNA


syringae, Fusarium oxysporum (liquid phase)

436 pathogens 2 magnetically separated antibody 103−106 cfu/mL 1 × 103 cfu/mL

7941
(liquid phase)

437 pathogen antigens EHI_115350, 2 metal substrate antibody 0−10 ng/mL 1 pg/mL
EHI_182030 10 pg/mL

429 proteins EGFR, EpCAM, CD44 3 cell membrane antibody

273 phenotypic markers 3 cell membrane antibody

438 proteins 2 liquid phase 0.2−2 μg/mL 0.2 μg/mL

17 IgG 2m+n − 1 glass antibody 1 μM to 1 fM 1 fM

61 IgG 3 magnetically separated antibody 1 μg/mL to 0.1 pg/mL 0.1 pg/mL


(liquid phase)

135 cancer cells 4 magnetically separated antibody 2 × 105 MRC-5 (SBR3/HeLa), 5 × 104 MRC-5 (Jurkat T), a single cancer cell
(liquid phase) 9 × 104 MRC-5 (LNCaP)

433 Hg2+ 2 liquid phase DNA 0.002−0.5 ng/mL 1.69 pg/mL


Ag+ 1.71 pg/mL

372 Hg2+ 2 magnetically separated DNA 0.1−1000 nM 2 nM


Review

(liquid phase)

DOI: 10.1021/acs.chemrev.7b00027
Chem. Rev. 2017, 117, 7910−7963
Table 6. continued
multiplexing recognition
refs targets ability detection platform unit dynamic range LOD
Ag+ 10−1000 nM 10 nM

114 metal ions (Ag+/Hg2+) 2 metal substrate DNA 1.7 pM for the Au nanohole array chip, 5 pM
Chemical Reviews

for the Au film chip

113 glycoproteins HRP, 2 liquid phase boronate 0.1 ng/mL to 100 μg/mL 0.7 nM
AFP affinity 1 ng/mL to 10 μg/mL 3 nM

112 CA15-3 3 glass antibody 0.1−500 U/mL 0.99 U/mL


CA27-29 0.1−500 U/mL 0.13 U/mL
CEA 0.1−500 ng/mL 0.05 ng/mL

125 CEA 2 liquid phase antibody 0.01 pg/mL to 1000 ng/mL 6.6 × 10−6 ng/mL
AFP 0.1 pg/mL to 1000 ng/mL 7.2 × 10−5 ng/mL

439 CEA 2 liquid phase antibody 1 ng/mL to 1 fg/mL 1.48 pg/mL


NSE 2.04 pg/mL

272 CA125 4 microfluidic antibody 1 aM to 1 pM 15 fM


HER2 17 fM

7942
HE4 21 fM
eotaxin-1 6.5 fM

304 melanoma DNA 3 magnetically separated DNA 10 copies


(liquid phase)

440 Staphylococcus aureus 2 magnetically separated aptamer 102−107 cfu/mL 35 cfu/mL


Salmonella typhimurium (liquid phase) 15 cfu/mL
Review

DOI: 10.1021/acs.chemrev.7b00027
Chem. Rev. 2017, 117, 7910−7963
Chemical Reviews Review

Figure 29. (a) Assembly of a single nanowire on graphene to be used as SERS substrate. Reprinted with permission from ref 441. Copyright 2016
Royal Society of Chemistry. (b) Ordered SERS microsphere array obtained by assembling Fe3O4@Ag@SiO2 microparticles. Reprinted with
permission from ref 442. Copyright 2016 American Chemical Society. (c) Evaporation-based assembly of silver nanowires. Reprinted with
permission from ref 444. Copyright 2016 Wiley-VCH Verlag GmbH & Co. KGaA.

multiplex detection of liver cancer biomarkers AFP and 6. CHALLENGES AND PERSPECTIVES
glypican-3.39 The sensitivity is down to sub-picomolar SERS has been reported to realize single-molecule detection
concentrations in saline solution. from more than 20 years. Despite the excellent optical-sensing
Lau et al. reported a multiplex point-of-care diagnostic capacity, SERS has not yet been used for practical applications
platform for plant pathogens.428 This platform employs SERS such as clinical diagnosis and biochemical detection. To
and recombinase polymerase amplification (RPA) techniques. promote SERS-based immunoassay from a research focus to
They utilized DNA-strand-modified SERS nanotags and practical detection method, future attention should be focused
magnetic nanoparticles. Multiplex detection of three pathogens on the improvement of reproducibility, the reduction of costs,
was successfully demonstrated. Experimental results proved the ease of use for point-of-care (POC) applications, etc. In this
that the RPA−SERS method was faster and more sensitive than section, we will focus on these issues by discussing practical
PCR. routes and future prospects toward the optimization of SERS-
Bodelon et al. synthesized surface-enhanced resonance based immunoassays.
Raman scattering (SERRS)-encoded colloids for the identi- 6.1. Improving the Reproducibility of SERS-Based
fication and imaging of proteins on cell membranes.429 In their Immunoassay
experiments, simultaneous imaging of three tumor biomarkers The relatively poor reproducibility of SERS-based immuno-
on the cancer cell membrane was realized, including EGFR, assay is a common problem that hinders the promotion of
epithelial cell adhesion molecule (EpCAM), and homing cell SERS to practical applications. To improve the reproducibility
adhesion molecule (CD44). The SERRS-encoded colloids can of SERS-based biosensing, the uniformity and stability of SERS
discriminate tumor and nontumor cells. substrates and nanoprobes should be improved.
5.6.2. Multiplex Ion Detection. Aside from the above- 6.1.1. Uniformity of the Immune Substrates. In
mentioned biological targets, platforms based on SERS can also addition to the previously mentioned plasmonic arrays (the
be used to analyze other chemical targets, such as metal ions last paragraph of section 3.3.1), there are some other substrates
and small molecules. For example, Kang et al. reported a for reproducible SERS analysis. Kim et al. assembled a single
simultaneous detection of three metal ions (i.e., Hg2+, Ag+, and nanowire on graphene as a novel SERS substrate (Figure
29a).441 Gold nanowires (Au NWs) with well-defined single-
Pb2+) using a gold-nanowire-on-chip SERS sensor.432 Our
crystal geometry were coated onto a monolayer graphene,
group has presented a magnetic-nanobead-assisted SERS-based forming a well-defined and continuous nanogap that provides
method for the simultaneous detection of Hg2+ and Ag+.372 Li extremely reproducible and stable SERS signals. The in-
et al. reported a SERS-encoded nanoplatform for the nanowire (in-NW) and nanowire−nanowire (NW−NW)
simultaneous detection of Hg2+ and Ag+.433 With the excellent reproducibility was verified through SERS mapping and
multiplexing ability, SERS holds a great potential in developing cumulative curves. Moreover, the platform showed an improved
highly efficient and informative biochemical detection platforms stability and long-term oxidation immunity. The high
(Table 6). reproducibility and stability of this substrate make it a
7943 DOI: 10.1021/acs.chemrev.7b00027
Chem. Rev. 2017, 117, 7910−7963
Chemical Reviews Review

Figure 30. (a) A 3D-printed supercapacitor-powered electrochemiluminescent protein immunoarray. Reprinted with permission from ref 450.
Copyright 2016 Elsevier B.V. (b) A portable and microcapsule array chip fabricated by 3D ice printing for visual target detection. Reprinted with
permission from ref 451. Copyright 2015 American Chemical Society.

potentially practical SERS substrate. Zheng et al. employed self- used the cost-effective 3D ice printing technology to fabricate
assembled polystyrene microbeads as templates to fabricate a ice structures for the rapid visual detection of nitrite and
gold nanohole array as a reproducible SERS substrate.114 Niu et glucose (Figure 30b).451 A variety of ice structures with
al. assembled monodisperse Fe3O4@Ag@SiO2 microspheres different geometries and sizes can be easily achieved with 3D
onto hierarchical surfaces (Figure 29b).442 Perfect integration ice printing. The platform has a potential for POC diagnosis
of ordered microsphere arrays has been achieved with the without the requirement of medical specialists. Currently, the
assistance of a magnetic field. The simple manipulation and low 3D printing technology has not been combined with SERS
cost make it a promising candidate for a SERS substrate. detection yet. The common materials for SERS substrates,
Another route to fabricate highly uniform substrate is including gold and silver, are seldom used as the printing
evaporation-based self-assembly. Self-aligned nanorods and materials due to the intrinsic properties of metal. However, 3D
nanowires could be attached onto the surface and used as the printing has already been used to fabricate various nanostruc-
SERS substrates (Figure 29c).443,444 Recently, Zhu et al. tures with nonmetallic materials. The combination of 3D
fabricated a hierarchically ordered array of silver nanorod printing with traditional metallic nanofabrication technologies
bundles with a large SERS enhancement factor (108) and a high might be promising for producing highly reproducible and
reproducibility of SERS-signal (RSD 10%).445 The great SERS SERS-active 3D substrates at low costs.
enhancement was due to the strong EM field coupling among On the other hand, there are some other uniform
those neighboring Ag nanorods with nanogaps of around 2 nm. nanosubstrates that broaden the dynamic range of detection.
Currently, there are also some commercial SERS substrates In a typical immunoassay, the intensity of readout signal often
with a high reproducibility, such as gold (or silver) coated undergoes a linear zone (commonly known as the dynamic
nanostructured silicon like Klarite or those by produced range) and a saturation plateau. So far, the majority of
Silmeco, nanosponge gold/silver SERS substrates developed immunoassays still suffer from a narrow dynamic range. Thus,
by Ocean Opotics, and gold nanorods substrates developed by ways to broaden the dynamic range are urgently required in
Horiba Scientific. It is expected that all the above substrates diagnostics applications. Since the concentration of target
might be used for SERS-based immunoassays to improve the molecules is usually very low in the practical applications, one
reproducibility. typical method for improving the dynamic range is to decrease
Another popular technique for substrate fabrication is 3D the LOD value. For example, Miele et al. fabricated the sensing
printing. Using this new technology, the desired materials could surfaces by using regular arrays of microdomains of different
be assembled under computer control to fabricate certain 3D wettabilities, self-assembling analyte molecules in the more
structures.446−448 Since the 1990s, remarkable progress has wettable domains.169 Thus, the detecting sensitivity was
been made in 3D printing technology. It enables a rapid increased to ∼10 aM due to the concentrated molecules in
fabrication of personalized products at very low cost. Up to the active domains, leading to a remarkable improvement in the
now, 3D printing have been employed for printing tissues and dynamic range as 6 orders of magnitude (10 aM to 1 pM).
organs, microfluidic chips,446 electrochemical devices,449 etc. Besides, Dai and co-workers presented protein microarrays on a
Recently, Kadimisetty et al. developed a low-cost 3D-printed novel nanostructured, plasmonic gold film with near-infrared
immunoarray for the detection of three proteins (Figure fluorescence enhancement of up to 100-fold, which shows a
30a).450 The system costs around ∼€0.50 for each protein and detecting limit to the 5 fM level with a dynamic range over 6
each detection can be completed in 35 min. This device is orders of magnitude.452 In the case of a high concentration of
suitable for resource-limited clinical environments. Zhang et al. targets, the dilution operation can be carried out to obtain
7944 DOI: 10.1021/acs.chemrev.7b00027
Chem. Rev. 2017, 117, 7910−7963
Chemical Reviews Review

Figure 31. (a) Multilayered shell nanotags with highly uniform single-particle Raman readout. Reprinted with permission from ref 458. Copyright
2012 Royal Society of Chemistry. (b) Ag shell−Au satellite hetero-nanostructure SERS probes. Reprinted with permission from ref 459. Copyright
2014 American Chemical Society.

accurate results. Thus, a lower detection limit and a broader intense and uniform SERS signals with an EF of 1.4 × 106
dynamic range are two key factors for developing SERS and a relative standard deviation of 11%. These novel
immunoassays for practical uses. nanostructures open an avenue for reliable SERS immunoa-
6.1.2. Stability of SERS Nanoprobes. On the other hand, nalysis.
the structure of the nanoprobes should be carefully designed to 6.2. Real Sample Detection
improve the stability of the nanoprobes.453,454 The famous
shell-isolated nanoparticle-enhanced Raman spectroscopy Commercialization of SERS-based immunoassay requires a
(SHINERS) developed by Li et al. employed an ultrathin silica precise quantification of biomolecules in real samples (e.g.,
or alumni shell to protect metal nanoparticles from the human blood). Currently, it is still challenging to minimize the
aggregation and direct contact with the surrounding environ- interference from other components in real sample and to
ment, while maintaining a high SERS activity.105 The versatility realize precise quantification of biomarkers.
6.2.1. Serum and Plasma. Human plasma is extracted
of SHINERS has been demonstrated through the detection of
from whole blood by removing the blood cells and clotting
yeast cells, pesticide residues on food/fruits, and hydrogen
factors, while human serum is the plasma without fibrinogens.
adsorption on single-crystal flat surfaces of Pt and Si. SHINERS
The components of human plasma include proteins, hormones,
expands the flexibility of SERS for various applications in
ions, glucose, etc. To predict and validate the ability of SERS-
biology, chemistry, medicine, and environmental monitoring.
based immunoassay in real sample detection, the proof-of-
Another popular strategy to fabricate stable SERS nanoprobes concept experiment is usually performed by quantifying the
is using DNA as the template to assemble nanoparticles.455−457 concentration of target molecules in serum or plasma. Li et al.
Complementary hybridization between specially designed DNA developed a multiplex immunoassay for the simultaneous
strands can facilitate the formation of stable and sophisticated detection of three serological cancer biomarkers using the SERS
nanostructures with fine SERS-enhancing abilities. technique.112 The platform was able to realize the detection of
To improve the SERS activity, “hot spots” are frequently CA15-3, CA27-29, and CEA in human serum with LODs down
used for signal amplification in SERS nanoprobes. However, the to 0.99 U/mL, 0.13 U/mL, and 0.05 ng/mL, respectively. The
creation of SERS hot spots often leads to uncontrollable 3D hierarchical plasmonic nanoarchitecture enhanced SERS
aggregation of the nanoparticles. There is a balance between immunosensor developed by Li et al. realized EGFR detection
the stability of SERS nanoprobes and high SERS activity. The in human plasma samples from clinical breast cancer
anisotropic structure with uncontrollable hot spots might lead patients.248 The detection results were comparable with those
to signal fluctuation and dependency on incident light obtained from commercial ELISA kits.
polarization. To overcome these problems, Liu et al. fabricated Besides the above examples, a great number of reports have
SERS nanotags with highly uniform and reproducible signals claimed successful quantification of biomolecules in human
using layer-by-layer assembly and cross-linkage of small silver serum or plasma. However, to the best of our knowledge, there
nanoparticles (Figure 31a).458 The problem of reproducibility are no commercial SERS kits for immunoassays to date.
could be solved to a large extent by the assembly of small metal Possible reasons might arise from the low reproducibility
nanoparticles into a highly controllable multilayered shell. The between different tests. Although the reproducibility of SERS-
SERS enhancement is within the narrow range from 1.1 × 107 based immunoassay in a single test (using the same materials
to 1.4 × 107. Chang et al. developed a Ag shell−Au satellite and platform in the same day) could be improved by
nanostructural composite as a NIR-active SERS probe (Figure optimizing the stability and uniformity of immune substrates
31b).459 The assembly of gold nanospheres onto silver and SERS probes, as discussed in the previous section, the
nanoparticles leads to the formation of uniform 3D hot spots. reproducibility of detection results from multiple tests are not
The hetrometallic shell−satellite nanoparticles produced easy to keep in accordance. This is primarily due to the low
7945 DOI: 10.1021/acs.chemrev.7b00027
Chem. Rev. 2017, 117, 7910−7963
Chemical Reviews Review

Figure 32. (a) An integrated barcode chip for the rapid and multiplex protein detection in human blood. Reprinted with permission from ref 461.
Copyright 2008 Nature Publishing Group. (b) An “mChip” HIV assay done in Rwanda on hundreds of locally collected human blood samples.
Reprinted with permission from ref 462. Copyright 2011 Nature Publishing Group.

reproducibility of SERS immunoprobes: (1) Since the SERS surfaces with high resistance to nonspecific adsorption in
probes are usually fabricated by chemical methods, the size and complex matrixes is another important issue to consider in the
shape of the nanoparticles could be varied due to the future research.
differences in the quality of original materials, the mixing 6.2.2. Whole Blood. Whole blood contains a large quantity
speed, the temperature, the pH value of surrounding environ- of blood cells, including red blood cells and white blood cells.
ment, etc. This could become more complicated with respect to Immunoassays for whole blood without preprocessing greatly
the chemical and biological modification process. As a result, it reduce human work, which would be critical for POC diagnosis
is not easy to maintain the high uniformity of SERS probes. in resource-limited areas. Currently, SERS-based immunoassay
Calibration should be carried out before each test to improve has already been used for detecting CTCs in whole blood. For
the precision of detection. (2) The complex components in the example, Wu et al. employed SERS nanoprobes for the direct
environment could lead to the aggregation of SERS probes, detection of CTCs in whole rabbit blood with a LOD down to
which is almost uncontrollable and consequently leads to 5 cells/mL.25 Wang et al. employed EGF−SERS nanoparticles
significant variations in SERS signals and the nonspecific to rapidly detect CTCs in peripheral blood specimens from
adsorption of immunoprobes. Additionally, in physiological patients.396 These platforms have demonstrated the possibility
conditions, the fraction of target molecules is usually miniscule of using SERS-based immunoassay for cancer diagnosis at the
in relation to the other molecules present. Nonspecific cellular level. On the other hand, it would be more meaningful
adsorption of nontarget molecules becomes one of the most to analyze biomarkers at the molecular level. Up to now, there
pressing concerns, which will raise the detection limit of the are a few platforms which have been used for direct
strategy and thereby decrease the sensitivity of the assay. identification of protein or DNA biomarkers from the whole
Generally, the interfering molecules tend to interact with the blood using optical techniques other than SERS. Fan et al. have
surface materials mainly through electrostatic interactions or developed an integrated barcode chip for rapid (10 min) and
van der Waals forces, which is far weaker than that between the multiplex protein detection in human blood (Figure 32a).461
target molecules and the recognition probes. Thus, one typical The microfluidic chip enables the isolation of blood cells and
way to solve the problem of nonspecific adsorption is to reduce the measurement of plasma protein biomarkers at the same
the reaction time. Driskell et al. introduced the concept of a time on a single chip using fluorescence immunoassays. The
rotating capture substrate instead of a conventional static device holds potential for inexpensive, noninvasive, and
substrate.460 This facile method increased the flux of target informative clinical diagnoses. Chin et al. performed an
antigen and SERS labels to the solid substrate, resulting in a “mChip” assay in Rwanda on hundreds of locally collected
lower nonspecific adsorption. Similarly, an accelerated SERS human blood samples, which enabled a rapid diagnosis of HIV
immunoassay based on a syringe pump was proposed to using unprocessed whole blood (Figure 32b).462 Unlike
actively transport the analyte and extrinsic Raman labels to the benchtop ELISA assays, the mChip can perform these
capture surface, which avoided the nonspecific binding resulted immunoassays with minimal equipment and can be completed
from the passive diffusion.254 Recently, Wang et al. utilized ac- within 20 min using only several microliters of blood sample,
EHD-induced nanoscaled surface shear forces to remove the which is an ideal choice for diagnoses in resource-limited areas.
nonspecific adsorption, which also enhanced the capture We envision that the introduction of SERS to this platform
kinetics simultaneously.279 Therefore, how to construct the might improve the immunoassay performance in terms of the
7946 DOI: 10.1021/acs.chemrev.7b00027
Chem. Rev. 2017, 117, 7910−7963
Chemical Reviews Review

Figure 33. (a) Scheme of the flow-focusing microfluidic device used for controlled Ag-NP aggregation. (b) At the flow-focusing junction, the sample
stream is enveloped by the side streams and diffusion drives lateral mass transport between the laminar flows, here visualized with a fluorescent dye.
(c) Schematic of the reaction. (d) Latent variables calculated for the calibration data after varimax rotation of the principal components. Reprinted
with permission from ref 464. Copyright 2013 American Chemical Society.

sensitivity and multiplexing ability, as long as the reproducibility spectroscopic device for in vivo and intraoperative cancer
of the SERS technique is improved. detection that provides promising routes for real-time tumor
6.2.3. Other. Besides human blood, there are other human detection and image-guided surgery.468 On account of the high
body fluids containing various biomarkers, such as saliva, urine, sensitivity and excellent multiplexing ability, SERS-based
and sputum. SERS-based immunoassays have been used for the immunoassay is promising for POC applications. To further
detection of Hg2+ in human saliva and urine,366 pyocyanin in promote practical and commercial applications, the overall
sputum,463 drugs in human saliva and urine (Figure 33),464 etc. costs should be reduced. First, the fabrication of SERS
In addition to body fluids, SERS has been applied to analyze nanoprobes requires many more antibodies than the
targets in food samples, such as Escherichia coli O157 in apple fluorescence method. The bioconjugation efficiency should be
juice,465 zearalenone in feed samples,359 kanamycin in milk improved to reduce the antibody consumption. Second, the
samples,383 and Salmonella typhimurium and Staphylococcus POC SERS measurement requires a sophisticated portable
aureus in pork samples.441 All of these works make SERS an instrument, which increases the financial burden, especially for
excellent candidate for environmental monitoring and food rural areas. Consequently, advanced techniques for the design
quality and safety evaluation. Similar to the blood tests, the and fabrication of portable Raman spectrometer should be
quantification of other biochemical substances in real samples is developed. Third, POC diagnosis requires a simple readout of
not an easy job due to the interference from other species in SERS spectra, which becomes more sophisticated for SERS
the complex environment. The testing parameters should be decoding in multiplex analysis. Therefore, new software
carefully adjusted to reach the best performance.
programs integrated with data processing, analysis, and readout
6.3. Point-of-Care Testing (POCT) should be developed, so that the end-point users could easily
POCT refers to the test at or near the point-of-care, that is, at obtain the detection results. All the above issues should be
the time and place of patient care. POCT is critical to considered for turning the proof-of-concept research into
healthcare in resource-limited areas. An ideal POCT device commercial POC systems.
should fulfill the following requirements: small size, low sample 6.4. Practical SERS-Encoding Technique
consumption, low costs, easy operation, and intuitive readout
without complicated instrumentation. So far, extensive efforts SERS-encoding technique has been widely used for multiplex
have been made toward POC SERS immunoassays. Worsley et biosensing.70 The narrow Raman bands endow SERS with an
al. developed a lateral flow SERS immunoassay for the excellent coding capacity. In principle, SERS encoding would
multiplex detection of cardiac and inflammation biomarkers.270 enable the simultaneous detection of more than 100 kinds of
The combination of the SERS technique with the lateral flow targets, which could be differentiated from each other by
platform enables an easy and rapid detection of biomarkers, Raman frequencies. However, realistically, simultaneous
which would become one potential candidate for POC detection of only a few (two to five) kinds of biomolecules
diagnosis. Saha et al. performed SERS immunoassay on a has been achieved by SERS encoding to date. There are two
paper-based microfluidic platform, in which both the plasmonic main practical problems to be settled. On the one hand, the
nanoparticles and analytes were in mobile phase.466 The actual kinds of Raman reporters without spectral band overlap
analyte-induced controlled particle aggregation produced are still limited. Commonly used Raman reporters with large
reproducible and much enhanced SERS signals, which resulted scattering cross sections are usually similar in chemical
in a sensitivity beyond picomolar concentration. The presented structures, which inevitably leads to band overlaps. On the
approach is simple, rapid, and cost-effective and requires other hand, nonspecific antibody−antigen interaction, physical
microliter sample volume. In terms of the SERS measurement, adsorption, and cross-reactions among multiple encoded
portable Raman spectrometers have been widely used for in situ nanoprobes reduce the specificity of SERS, making it difficult
SERS analysis.105,467 Mohs et al. developed a hand-held to distinguish different analytes.
7947 DOI: 10.1021/acs.chemrev.7b00027
Chem. Rev. 2017, 117, 7910−7963
Chemical Reviews Review

Figure 34. (a) “Switch-off” biosensing for chymotrypsin-catalyzed reaction by SPR−SERS spectroscopy. Reprinted with permission from ref 472.
Copyright 2013 Royal Society of Chemistry. (b) Mercury ion detection by optical waveguide and surface-enhanced Raman spectroscopy. Reprinted
with permission from ref 473. Copyright 2012 Elsevier B.V.

To minimize band overlaps, it is ideal that the Raman immunoassay has some intrinsic disadvantages. First, the typical
molecules only have one strong Raman band, which can greatly sandwich immunoassay format fails to provide time-dependent
minimize possible band overlaps between different codes. One information on the analytes in one sensing unit. Dynamic
practical solution is to synthesize a group of Raman reporters analysis requires repetitive measurements at different time
with similar but different chemical structures. The characteristic points. Second, SERS usually has a relatively slow response and
Raman bands of these reporters should be close to each other readout speed, making it relatively unsuitable for real-time
and without obvious spectral overlaps to obtain the maximum analysis. Third, it is challenging to obtain reproducible SERS
kinds of different codes.431 Another solution is to combine signals due to the instability of nanoprobes in complex
different encoding techniques together. For example, the microenvironments. Therefore, the combination of SERS with
SERS−fluorescence joint spectral encoding technique proposed other detection techniques is expected to overcome some
by our group greatly increased the number of different codes intrinsic shortcomings of SERS-based immunoassays.
that can be practically obtained.17,135 Similarly, Zhang et al. 6.5.1. SERS−SPR Immunoassay. SERS has been
developed dual-encoded microbeads through a host−guest combined with SPR for dual mode biosensing. SPR could be
nanostructure, in which fluorescent dyes and quantum dots used for real-time monitoring of antibody−antigen interactions,
were used for joint encoding.469 The idea of joint encoding while SERS enables the analysis of the chemical structures of
provides an alternative for high-throughput multiplex detection. the analytes. By combining SERS with SPR, the relatively low
Future work in this field should focus on simplifying the reproducibility of SERS detection could be compensated by
structure and fabrication protocol of the nanoparticles SPR detection. Fu et al. proposed a “switch-off” biosensing
incorporated with different labels. Finally, complex Raman strategy for chymotrypsin detection based on the combined use
spectra could be analyzed with data processing methods like of the SERS and SPR techniques (Figure 34a).472 This
principle component analysis (PCA) and support vector approach could be used for real-time monitoring of the
machine (SVM), so that the merged Raman bands from reaction process, and the sensitivity reached down to 0.4 nM.
different reporters could be further extracted for simultaneous Similarly, Ma et al. developed a SERS−SPR dual-mode sensor
quantitative analysis of multiple targets.399,470,471 by assembling Au−pATP multilayers onto an optical waveguide
Another critical problem that limits the practical application (Figure 34b).473 The integrated chip was used for Hg2+
of the SERS-encoding technique is nonspecific interactions. To detection with a low concentration of 1 nM. The combination
reduce the nonspecific adsorption of SERS nanoprobes, of SERS with SPR provides a novel idea for real-time, specific,
researchers should pay much attention to experimental details, and sensitive detection of various target molecules.
including the orientation of antibodies, the monodispersity of 6.5.2. SERS−Fluorescence Immunoassay. A variety of
nanoparticles, and the blocking of nonspecific adsorption sites.
nanoprobes that simultaneously incorporate SERS and
On the other hand, antibody−antigen cross-reactions are
fluorescence signals into a composite nanoparticle have been
common in immune reactions, which could be reduced by
used for immunoassays. The fluorescence mode could be used
improving the quality of antibodies. This relies heavily on the
for fast screening of biomolecules, while the SERS mode can be
synthetic technology of highly purified antibodies. In general,
used for sensitive quantitative analysis. Previously, we have
all the above issues related to biomolecular interactions would
designed a SERS−fluorescence dual-mode nanoprobe based on
limit the available number of SERS codes that can be used for
multilayer core−shell nanoparticles.61 In the nanoprobe,
high-throughput biosensing. To fully exploit the excellent
encoding capacity of the SERS technique, the above problems Raman-reporter-labeled nanoparticles provide SERS signals,
should be carefully concerned for future researches. while quantum dots provide fluorescence signals. The dual-
mode nanoprobe could be used for rapid and sensitive
6.5. Combining SERS with Other Techniques for bioanalysis and imaging. Recently, Zou et al. employed
Multimodal Immunoassay graphene quantum dots as both the SERS and fluorescence
SERS has shown its advantage in ultrasensitive detection, labels, which simplifies the structure of the immunoprobes.115
structural analysis, and high stability. However, SERS-based Dual-mode nanoprobes partially overcome the slow readout
7948 DOI: 10.1021/acs.chemrev.7b00027
Chem. Rev. 2017, 117, 7910−7963
Chemical Reviews Review

Figure 35. (a) A SERS−electrochemical biosensos for monitoring DNA−drug interactions. Reprinted with permission from ref 474. Copyright 2016
Elsevier B.V. (b) A SERS−electrochemistry dual-mode immunosensor for simultaneous detection of multiple biomarkers. Reprinted with permission
from ref 475. Copyright 2015 Royal Society of Chemistry.

speed of Raman spectroscopy and thus hold a potential for substrates could be developed to enrich the target molecules.
high-throughput detection. With these platforms, SERS-based immunoassay has found
6.5.3. SERS−Electrochemistry Immunoassay. Electro- enormous applications in clinical diagnosis, environmental
chemical immunoassay is a label-free and sensitive detection monitoring, and biochemical analysis. Furthermore, combined
technique that has attracted much attention. The fast response with the optical-encoding technique, SERS shows a great
and ease of integration make it suitable for POC applications. promise for high-throughput assays.
Ilkhani et al. designed a SERS−electrochemical biosensor for Currently, researchers have tried to apply these SERS
chemotherapeutic drug screening, in which the DNA−drug platforms for detecting real samples. Some of the detection
interaction was monitored through SERS−electrochemical results showed a comparable performance to those using ELISA
transduction (Figure 35a).474 The variation of SERS spectra kits, which is the current employed golden standard. However,
indicated the binding between the DNA and drug, while the up to now, none of these SERS-based immunosensing
electrochemical function realized quantitative detection with a platforms have been approved for clinical uses. Looking into
LOD down to 8 μg/mL. Lu et al. developed a SERS− the future, there is a great demand for developing highly
electrochemistry dual-mode immunosensor for simultaneous reproducible SERS-active immunoassay platforms for practical
detection of multiple biomarkers (Figure 35b).475 Simultaneous applications, such as clinical diagnosis. Fortunately, the highly
detection of CEA and cytokeratin-19 (CK-19) showed high uniform nanosubstrates fabricated by the lithography, template,
sensitivity, selectivity, long-term stability, and reliability. and self-assembly technology have provided solutions to
6.5.4. Immunoassay with Hyperbolic Metamaterials. improve the reproducibility of SERS substrates (section
Recently, Sreekanth et al. developed an extremely sensitive 6.1.1). Meanwhile, various SERS-active nanoprobes constructed
biosensing platform based on hyperbolic metamaterials that by the assembly methods based on templates, electrostatic
allows the detection of low molecular weight biomolecules in adsorption, and surface chemistry help to maintain the
highly diluted solutions.476 The platform can support highly monodispersity of the nanoparticles in complex environments
confined bulk plasmon guided modes over a broad wavelength (section 6.1.2). For real sample detection, novel surface
range from visible to near IR. They reported the ability of this chemical engineering should be designed to minimize the
metamaterial platform to detect ultralow molecular weight (244 nonspecific adsorption of biofluids (section 6.2).
Da) biomolecules at picomolar concentrations using an affinity Additionally, it would be beneficial to develop SERS-based
model based on streptavidin−biotin interaction. This novel immunoassay platforms with small size, low sample con-
platform could potentially be combined with SERS for sumption, reduced cost, easy operation, and intuitive readout.
multimodal immunoassay with extreme sensitivity. In this respect, the microfluidic chips show the highest potential
for POC applications, while the paper-based test stripes could
7. CONCLUSION become an excellent choice for household assay devices
SERS offers many unique advantages over traditional immuno- (section 6.3). At present, some commercially available
assay techniques, including ultrasensitivity and excellent immunoassay systems based on microfluidics and paper have
multiplexing ability. The diversity of SERS-active platforms, been developed. For example, the FDA-approved i-STAT hand-
from traditional solid substrates and liquid microbeads to held microfluidic device enables the detection of cardiac
microfluidic chips, paper substrates, optical fibers, and hydro- markers with easy operation, and the paper-based pregnancy
phobic substrates, provides flexible choices for various test strips have been widely used for household early pregnancy
applications. Specifically, the standard sandwich immunoassay diagnosis. Summarily, great efforts are still needed to promote
on solid substrate has laid the foundation for the development SERS-based immunoassay from proof-of-concept research
of other platforms; to simplify the assay protocols, microbeads works to commercially available analytical tools.
are employed to establish immunoassays in liquid phase; for
parallel analysis of large amounts of samples with low volumes, AUTHOR INFORMATION
the microfluidic platform offers one of the best choices; to
enable on-site detection with remote control, immunoassays Corresponding Author
with optical fibers could be used; with the purpose of reducing *E-mail: cyp@seu.edu.cn.
the assay expenses, the paper-based substrate, which requires
ORCID
minimum cost for fabrication, might become an option; and in
order to achieve ultrasensitive analysis, superhydrophobic Yiping Cui: 0000-0002-4648-2506
7949 DOI: 10.1021/acs.chemrev.7b00027
Chem. Rev. 2017, 117, 7910−7963
Chemical Reviews Review

Notes Foundation of Jiangsu Province (No. BK20150623), the


The authors declare no competing financial interest. Science Foundation for The Excellent Youth Scholars of
Southeast University, and the Fundamental Research Funds for
Biographies the Central Universities.
Zhuyuan Wang received her B.S. in electronic science and engineering
in 2001 and her Ph.D. degree in physical electronics in 2005, both ABBREVIATIONS USED
from Southeast University, Nanjing, China. From 2005 to 2006, she 2-NAT = 2-naphthalenethiol
worked as a postdoctoral fellow in the State University of New York at 4-MPy = 4-mercaptopyridine
Buffalo, Buffalo, NY. From 2006 to 2011, she was an associate 4-MT = 4-methoxythiophenol
professor in the Advanced Photonics Center at Southeast University, ac-EHD = alternating current electrohydrodynamic
where she became a full professor in 2011. She has published over 120 AFB1 = aflatoxin B1
papers in key journals. Her research interests cover a number of optical AFP = α-fetoprotein
spectroscopic techniques in biological applications, such as surface- BDT = benzenedithiol
enhanced Raman scattering (SERS), fluorescence, and nonlinear CA19-9 = carbohydrate antigen 19-9
optical spectroscopy. CEA = carcinoembryonic antigen
Shenfei Zong received her B.S. in electronic science and engineering in CTC = circulating tumor cell
2010 and her Ph.D. degree in optical engineering in 2014, both from CV = crystal violet
Southeast University, Nanjing, China. Since then, she has been a DTNB = 5,5′-dithiobis(2-nitrobenzoic acid)
lecturer at the Advanced Photonics Center of Southeast University. EGFR = epidermal growth factor receptor
She is now focusing on cell imaging and biological analyte detection EIA = enzyme immunoassay
using optical spectroscopic techniques including SERS and fluo- ELISA = enzyme-linked immunosorbent assay
rescence. EpCAM = epithelial cell adhesion molecule
Lei Wu received his B.S. in electronic science and engineering from FA = folic acid
Southeast University, Nanjing, China, in 2012. He is currently a Ph.D. FISH = fluorescent in situ hybridization
candidate at the Advanced Photonics Center under the supervision of GERS = graphene-enhanced Raman scattering
Profs. Yiping Cui and Zhuyuan Wang. His research interests include HAV = hepatitis A virus
the development and application of SERS−microfluidic immunosen- HBV = hepatitis B virus
sors for the detection of disease biomarkers. HCPCF = hollow core photonic crystal fiber
HER2 = human epidermal growth factor receptor 2
Dan Zhu received her B.S. in electronic science and engineering from HIV = human immunodeficiency virus
Southeast University, Nanjing China, in 2011. She is currently a Ph.D. ICT = immuno-chromatographic test
candidate at the Advanced Photonics Center under the supervision of LOC = lab-on-a-chip
Profs. Yiping Cui and Zhuyuan Wang. Her research focuses on the LOD = limit of detection
interaction between liposomal nanohybrids and tumor cells. MBA or 4MBA = 4-mercaptobenzoic acid
Yiping Cui received his B.S., M.S., and Ph.D. degrees from the MIP = molecularly imprinted polymer
Department of Electronic Engineering, Southeast University, Nanjing, miRNA = microRNA
China in 1982, 1984, and 1994, respectively. Since 1993, he has been a MMC = 2,7-mercapto-4-methylcoumarin
professor in the Department of Electronics (now the School of MMTAA = 2-mercapto-4-methyl-5-thiazoleacetic acid
Electronic Science and Engineering), Southeast University, Nanjing, MUC1 = mucin-1
China. From 1995 to 1996, he was a visiting professor at the State NPs = nanoparticles
University of New York at Buffalo. He established the Advanced NRs = nanorods
Photonics Center of Southeast University and has been engaged in NSs = nanostars
research on polymeric nonlinear optics, nanophotonic materials, and OMQ NPs = organic−metal−quantum dot hybrid nano-
biophotonics. He has published over 470 papers in key journals and particles
coauthored two book chapters in monographs“Optics of Intense pATP = p-aminothiophenol
Light and Its Applications” and “Physics of Nonlinear Optics” (English PCR = polymerase chain reaction
version)and a graduate student textbook, Laser Physics. For his POC = point-of-care
pioneering contributions to the understanding of nonlinear and PSA = prostate-specific antigen
luminescent properties of polymeric, organic, and nanophotonic QDs = quantum dots
materials, he was elected as a fellow of the Optical Society of America.
He is vice president of the 27th CIE (Commission International de REFERENCES
L’Eclairage, International Commission on Illumination). He is also a
(1) Templin, M. F.; Stoll, D.; Schrenk, M.; Traub, P. C.; Vohringer,
Cheung Kong Professor and the winner of China National Funds for
C. F.; Joos, T. O. Protein Microarray Technology. Trends Biotechnol.
Distinguished Young Scientist in China. 2002, 20, 160−166.
(2) Giokas, D. L.; Vlessidis, A. G.; Tsogas, G. Z.; Evmiridis, N. P.
ACKNOWLEDGMENTS Nanoparticle-Assisted Chemiluminescence and Its Applications in
Analytical Chemistry. TrAC, Trends Anal. Chem. 2010, 29, 1113−1126.
This work was supported by the Natural Science Foundation of (3) MacBeath, G. Protein Microarrays and Proteomics. Nat. Genet.
China (NSFC) (No. 61535003), the National Key Basic 2002, 32, 526−532.
Research Program of China (No. 2015CB352002), the (4) Campion, A.; Kambhampati, P. Surface-Enhanced Raman
Excellent Youth Foundation of Jiangsu Province scattering. Chem. Soc. Rev. 1998, 27, 241−250.
(BK20140023), the Natural Science Foundation of China (5) Haisch, C. Raman-Based Microarray Readout: A Review. Anal.
(NSFC) (No. 61675042, 61505027), the Natural Science Bioanal. Chem. 2016, 408, 4535−4545.

7950 DOI: 10.1021/acs.chemrev.7b00027


Chem. Rev. 2017, 117, 7910−7963
Chemical Reviews Review

(6) Granger, J. H.; Schlotter, N. E.; Crawford, A. C.; Porter, M. D. (29) Akter, R.; Rhee, C. K.; Rahman, M. A. A Highly Sensitive
Prospects for Point-of-Care Pathogen Diagnostics Using Surface- Quartz Crystal Microbalance Immunosensor Based on Magnetic Bead-
Enhanced Raman Scattering (SERS). Chem. Soc. Rev. 2016, 45, 3865− Supported Bienzymes Catalyzed Mass Enhancement Strategy. Biosens.
3882. Bioelectron. 2015, 66, 539−546.
(7) Wang, Y.; Yan, B.; Chen, L. SERS Tags: Novel Optical (30) Luo, Y.; Liu, T.; Zhu, J.; Kong, L.; Wang, W.; Tan, L. Label-Free
Nanoprobes for Bioanalysis. Chem. Rev. 2013, 113, 1391−1428. and Sensitive Detection of Thrombomodulin, a Marker of Endothelial
(8) Porter, M. D.; Lipert, R. J.; Siperko, L. M.; Wang, G.; Narayanan, Cell Injury, Using Quartz Crystal Microbalance. Anal. Chem. 2015, 87,
R. SERS as a Bioassay Platform: Fundamentals, Design, and 11277−11284.
Applications. Chem. Soc. Rev. 2008, 37, 1001−1011. (31) Lee, J. S.; Joung, H.-A.; Kim, M.-G.; Park, C. B. Graphene-Based
(9) Laing, S.; Gracie, K.; Faulds, K. Multiplex In vitro Detection Chemiluminescence Resonance Energy Transfer for Homogeneous
Using SERS. Chem. Soc. Rev. 2016, 45, 1901−1918. Immunoassay. ACS Nano 2012, 6, 2978−2983.
(10) Moskovits, M. Surface-Enhanced Spectroscopy. Rev. Mod. Phys. (32) Zong, C.; Wu, J.; Xu, J.; Ju, H.; Yan, F. Multilayer Hemin/G-
1985, 57, 783−826. quadruplex Wrapped Gold Nanoparticles as Tag for Ultrasensitive
(11) Nie, S. M.; Emory, S. R. Probing Single Molecules and Single Multiplex Immunoassay by Chemiluminescence Imaging. Biosens.
Nanoparticles by Surface-Enhanced Raman Scattering. Science 1997, Bioelectron. 2013, 43, 372−378.
275, 1102−1106. (33) Ma, L.; Sun, Y.; Kang, X.; Wan, Y. Development of Nanobody-
(12) Fabris, L. SERS Tags: The Next Promising Tool for Based Flow Injection Chemiluminescence Immunoassay for Sensitive
Personalized Cancer Detection? Chemnanomat 2016, 2, 249−258. Detection of Human Prealbumin. Biosens. Bioelectron. 2014, 61, 165−
(13) Samanta, A.; Jana, S.; Das, R. K.; Chang, Y. T. Biocompatible 171.
Surface-Enhanced Raman Scattering Nanotags for In vivo Cancer (34) Geißler, D.; Stufler, S.; Löhmannsröben, H.-G.; Hildebrandt, N.
Detection. Nanomedicine 2014, 9, 523−535. Six-Color Time-Resolved Forster Resonance Energy Transfer for
(14) Vo-Dinh, T.; Fales, A. M.; Griffin, G. D.; Khoury, C. G.; Liu, Y.; Ultrasensitive Multiplexed Biosensing. J. Am. Chem. Soc. 2013, 135,
Ngo, H.; Norton, S. J.; Register, J. K.; Wang, H.-N.; Yuan, H. 1102−1109.
Plasmonic Nanoprobes: From Chemical Sensing to Medical (35) Wegner, K. D.; Jin, Z.; Lindén, S.; Jennings, T. L.; Hildebrandt,
Diagnostics and Therapy. Nanoscale 2013, 5, 10127−10140. N. Quantum-Dot-Based Forster Resonance Energy Transfer Immuno-
(15) Wang, Y. W.; Doerksen, J. D.; Kang, S.; Walsh, D.; Yang, Q.; assay for Sensitive Clinical Diagnostics of Low-Volume Serum
Hong, D.; Liu, J. T. C. Multiplexed Molecular Imaging of Fresh Tissue Samples. ACS Nano 2013, 7, 7411−7419.
Surfaces Enabled by Convection-Enhanced Topical Staining with (36) Wegner, K. D.; Lindén, S.; Jin, Z.; Jennings, T. L.; Khoulati, R.
SERS-Coded Nanoparticles. Small 2016, 12, 5612−5621. e.; van Bergen en Henegouwen, P. M.; Hildebrandt, N. Nanobodies
(16) Liu, B.; Zhao, X. W.; Jiang, W.; Fu, D. G.; Gu, Z. Z. Multiplex and Nanocrystals: Highly Sensitive Quantum Dot-Based Homoge-
Bioassays Encoded by Photonic Crystal Beads and SERS Nanotags. neous FRET Immunoassay for Serum-Based EGFR Detection. Small
Nanoscale 2016, 8, 17465−17471. 2014, 10, 734−740.
(17) Wang, Z. Y.; Zong, S. F.; Li, W.; Wang, C. L.; Xu, S. H.; Chen, (37) Bui, M.-P. N.; Ahmed, S.; Abbas, A. Single-Digit Pathogen and
H.; Cui, Y. P. SERS-Fluorescence Joint Spectral Encoding Using Attomolar Detection with the Naked Eye Using Liposome-Amplified
Organic-Metal-QD Hybrid Nanoparticles with A Huge Encoding Plasmonic Immunoassay. Nano Lett. 2015, 15, 6239−6246.
Capacity for High-Throughput Biodetection: Putting Theory into (38) Sanjay, S. T.; Dou, M.; Sun, J.; Li, X. A Paper/Polymer Hybrid
Practice. J. Am. Chem. Soc. 2012, 134, 2993−3000. Microfluidic Microplate for Rapid Quantitative Detection of Multiple
(18) Gosling, J. P. A Decade of Development in Immunoassay Disease Biomarkers. Sci. Rep. 2016, 6, 30474.
Methodology. Clin. Chem. 1990, 36, 1408−1427. (39) Tang, B. C.; Wang, J. J.; Hutchison, J. A.; Ma, L.; Zhang, N.;
(19) Borrebaeck, C. A. K. Antibodies in Diagnostics - From Guo, H.; Hu, Z. B.; Li, M.; Zhao, Y. L. Ultrasensitive, Multiplex Raman
Immunoassays to Protein Chips. Immunol. Today 2000, 21, 379−382. Frequency Shift Immunoassay of Liver Cancer Biomarkers in
(20) Yalow, R. S.; Berson, S. A. Assay of Plasma Insulin in Human Physiological Media. ACS Nano 2016, 10, 871−879.
Subects by Immunological Methods. Nature 1959, 184, 1648−1649. (40) Chung, E.; Lee, J.; Yu, J.; Lee, S.; Kang, J. H.; Chung, I. Y.;
(21) Yalow, R. S.; Berson, S. A. Immunoassay of Endogenous Plasma Choo, J. Use of Surface-Enhanced Raman Scattering to Quantify
Insulin in Man. J. Clin. Invest. 1960, 39, 1157−1175. EGFR Markers Uninhibited by Cetuximab Antibodies. Biosens.
(22) Engvall, E.; Perlmann, P. Enzyme-Linked Immunosorbent Assay Bioelectron. 2014, 60, 358−365.
(ELISA) Quantitative Assay of Immunoglobulin-G. Immunochemistry (41) Wang, W.; Wang, W.; Liu, L.; Xu, L.; Kuang, H.; Zhu, J.; Xu, C.
1971, 8, 871−874. Nanoshell-Enhanced Raman Spectroscopy on Microplate for Staph-
(23) Van Weemen, B. K.; Schuurs, A. H. W. Immunoassay Using ylococcal Enterotoxin B Sensing. ACS Appl. Mater. Interfaces 2016, 8,
Antigen-Enzyme Conjugates. FEBS Lett. 1971, 15, 232−236. 15591−15597.
(24) Chikkaveeraiah, B. V.; Bhirde, A. A.; Morgan, N. Y.; Eden, H. S.; (42) He, Y.; Wang, Y.; Yang, X.; Xie, S.; Yuan, R.; Chai, Y. Metal
Chen, X. Y. Electrochemical Immunosensors for Detection of Cancer Organic Frameworks Combining CoFe2O4 Magnetic Nanoparticles as
Protein Biomarkers. ACS Nano 2012, 6, 6546−6561. Highly Efficient SERS Sensing Platform for Ultrasensitive Detection of
(25) Wu, X.; Luo, L.; Yang, S.; Ma, X.; Li, Y.; Dong, C.; Tian, Y.; N-Terminal Pro-Brain Natriuretic Peptide. ACS Appl. Mater. Interfaces
Zhang, L. e.; Shen, Z.; Wu, A. Improved SERS Nanoparticles for 2016, 8, 7683−7690.
Direct Detection of Circulating Tumor Cells in the Blood. ACS Appl. (43) Vasilescu, A.; Purcarea, C.; Popa, E.; Zamfir, M.; Mihai, I.;
Mater. Interfaces 2015, 7, 9965−9971. Litescu, S.; David, S.; Gaspar, S.; Gheorghiu, M.; Marty, J.-L. Versatile
(26) Yang, F.; Zuo, X.; Li, Z.; Deng, W.; Shi, J.; Zhang, G.; Huang, SPR Aptasensor for Detection of Lysozyme Dimer in Oligomeric and
Q.; Song, S.; Fan, C. A Bubble-Mediated Intelligent Microscale Aggregated Mixtures. Biosens. Bioelectron. 2016, 83, 353−360.
Electrochemical Device for Single-Step Quantitative Bioassays. Adv. (44) Soler, M.; Estevez, M.-C.; de Lourdes Moreno, M.; Cebolla, A.;
Mater. 2014, 26, 4671−4676. Lechuga, L. M. Label-Free SPR Detection of Gluten Peptides in Urine
(27) Johari-Ahar, M.; Rashidi, M.; Barar, J.; Aghaie, M.; for Non-Invasive Celiac Disease Follow-up. Biosens. Bioelectron. 2016,
Mohammadnejad, D.; Ramazani, A.; Karami, P.; Coukos, G.; Omidi, 79, 158−164.
Y. An Ultra-Sensitive Impedimetric Immunosensor for Detection of (45) Pal, M. K.; Rashid, M.; Bisht, M. Multiplexed Magnetic
the Serum Oncomarker CA-125 in Ovarian Cancer Patients. Nanoscale Nanoparticle-Antibody Conjugates (MNPs-ABS) Based Prognostic
2015, 7, 3768−3779. Detection of Ovarian Cancer Biomarkers, CA-125, β-2M and ApoA1
(28) Zhai, Q.; Zhang, X.; Li, J.; Wang, E. Molybdenum Carbide Using Fluorescence Spectroscopy with Comparison of Surface
Nanotubes: A Novel Multifunctional Material for Label-Free Electro- Plasmon Resonance (SPR) Analysis. Biosens. Bioelectron. 2015, 73,
chemical Immunosensing. Nanoscale 2016, 8, 15303−15308. 146−152.

7951 DOI: 10.1021/acs.chemrev.7b00027


Chem. Rev. 2017, 117, 7910−7963
Chemical Reviews Review

(46) Chianella, I.; Guerreiro, A.; Moczko, E.; Caygill, J. S.; Piletska, E. (66) Wu, L.; Wang, Z. Y.; Fan, K. Q.; Zong, S. F.; Cui, Y. P. A SERS-
V.; Perez De Vargas Sansalvador, I. M.; Whitcombe, M. J.; Piletsky, S. Assisted 3D Barcode Chip for High-Throughput Biosensing. Small
A. Direct Replacement of Antibodies with Molecularly Imprinted 2015, 11, 2798−2806.
Polymer Nanoparticles in ELISA-Development of a Novel Assay for (67) Kneipp, J.; Kneipp, H.; Kneipp, K. SERS - a Single-Molecule
Vancomycin. Anal. Chem. 2013, 85, 8462−8468. and Nanoscale Tool for Bioanalytics. Chem. Soc. Rev. 2008, 37, 1052−
(47) Glavan, A. C.; Christodouleas, D. C.; Mosadegh, B.; Yu, H. D.; 1060.
Smith, B. S.; Lessing, J.; Fernandez-Abedul, M. T.; Whitesides, G. M. (68) Qian, X. M.; Nie, S. M. Single-Molecule and Single-Nanoparticle
Folding Analytical Devices for Electrochemical ELISA in Hydrophobic SERS: From Fundamental Mechanisms to Biomedical Applications.
R-H Paper. Anal. Chem. 2014, 86, 11999−12007. Chem. Soc. Rev. 2008, 37, 912−920.
(48) Hu, L.; Xu, G. Applications and Trends in Electro- (69) Le Ru, E. C.; Etchegoin, P. G. Single-Molecule Surface-
chemiluminescence. Chem. Soc. Rev. 2010, 39, 3275−3304. Enhanced Raman Spectroscopy. Annu. Rev. Phys. Chem. 2012, 63, 65−
(49) Esteve-Turrillas, F. A.; Abad-Fuentes, A. Applications of 87.
Quantum Dots as Probes in Immunosensing of Small-Sized Analytes. (70) Rodriguez-Lorenzo, L.; Fabris, L.; Alvarez-Puebla, R. A.
Biosens. Bioelectron. 2013, 41, 12−29. Multiplex Optical Sensing with Surface-Enhanced Raman Scattering:
(50) Taitt, C. R.; Anderson, G. P.; Ligler, F. S. Evanescent Wave A Critical Review. Anal. Chim. Acta 2012, 745, 10−23.
Fluorescence Biosensors: Advances of the Last Decade. Biosens. (71) Cialla, D.; Pollok, S.; Steinbrucker, C.; Weber, K.; Popp, J.
Bioelectron. 2016, 76, 103−112. SERS-Based Detection of Biomolecules. Nanophotonics 2014, 3, 383−
(51) Olaru, A.; Bala, C.; Jaffrezic-Renault, N.; Aboul-Enein, H. Y. 411.
Surface Plasmon Resonance (SPR) Biosensors in Pharmaceutical (72) Lane, L. A.; Qian, X. M.; Nie, S. M. SERS Nanoparticles in
Analysis. Crit. Rev. Anal. Chem. 2015, 45, 97−105. Medicine: From Label-Free Detection to Spectroscopic Tagging.
(52) Kurosawa, S.; Park, J.-W.; Aizawa, H.; Wakida, S.-I.; Tao, H.; Chem. Rev. 2015, 115, 10489−10529.
Ishihara, K. Quartz Crystal Microbalance Immunosensors for Environ- (73) Wu, L.; Wang, Z. Y.; Zong, S. F.; Huang, Z.; Zhang, P. Y.; Cui,
mental Monitoring. Biosens. Bioelectron. 2006, 22, 473−481. Y. P. A SERS-Based Immunoassay with Highly Increased Sensitivity
(53) Raman, C. V.; Krishnan, K. S. A New Type of Secondary Using Gold/Silver Core-Shell Nanorods. Biosens. Bioelectron. 2012, 38,
Radiation. Nature 1928, 121, 501−502. 94−99.
(54) Landsberg, G.; Mandelstam, L. Eine neue Erscheinung bei der (74) Hong, W.; Seo, H. K.; Jung, Y. M. SERS Immunoassay Using
Lichtzerstreuung in Krystallen. Naturwissenschaften 1928, 16, 557− Microcontact Printing for Application of Sensitive Biosensors. Bull.
558. Korean Chem. Soc. 2011, 32, 4281−4285.
(55) Fleischmann, M.; Hendra, P. J.; McQuillan, A. J. Raman-Spectra (75) Vianna, P. G.; Grasseschi, D.; Costa, G. K. B.; Carvalho, I. C. S.;
of Pyridine Absorbed at a Silver Electrone. Chem. Phys. Lett. 1974, 26, Domingues, S. H.; Fontana, J.; de Matos, C. J. S. Graphene Oxide/
163−166. Gold Nanorod Nanocomposite for Stable Surface-Enhanced Raman
(56) Talian, I.; Mogensen, K. B.; Orinak, A.; Kaniansky, D.; Hubner, Spectroscopy. ACS Photonics 2016, 3, 1027−1035.
(76) Murphy, S. V.; Atala, A. 3D Bioprinting of Tissues and Organs.
J. Surface-Enhanced Raman Spectroscopy on Novel Black Silicon-
Nat. Biotechnol. 2014, 32, 773−785.
Based Nanostructured Surfaces. J. Raman Spectrosc. 2009, 40, 982−
(77) Yang, J.; Wang, Z. Y.; Zong, S. F.; Song, C. Y.; Zhang, R. H.;
986.
Cui, Y. P. Distinguishing Breast Cancer Cells Using Surface-Enhanced
(57) Yuan, H. K.; Fales, A. M.; Khoury, C. G.; Liu, J.; Vo-Dinh, T.
Raman Scattering. Anal. Bioanal. Chem. 2012, 402, 1093−1100.
Spectral Characterization and Intracellular Detection of Surface-
(78) Zong, S.; Wang, Z.; Chen, H.; Cui, Y. Ultrasensitive Telomerase
Enhanced Raman Scattering (SERS)-Encoded Plasmonic Gold
Activity Detection by Telomeric Elongation Controlled Surface
Nanostars. J. Raman Spectrosc. 2013, 44, 234−239. Enhanced Raman Scattering. Small 2013, 9, 4215−4220.
(58) Jana, D.; Mandal, A.; De, G. High Raman Enhancing Shape- (79) Wu, L.; Wang, Z. Y.; Zong, S. F.; Cui, Y. P. Rapid and
Tunable Ag Nanoplates in Alumina: A Reliable and Efficient SERS Reproducible Analysis of Thiocyanate in Real Human Serum and
Technique. ACS Appl. Mater. Interfaces 2012, 4, 3330−3334. Saliva Using a Droplet SERS-Microfluidic Chip. Biosens. Bioelectron.
(59) Ni, J.; Lipert, R. J.; Dawson, G. B.; Porter, M. D. Immunoassay 2014, 62, 13−18.
Readout Method Using Extrinsic Raman Labels Adsorbed on (80) Tao, A.; Kim, F.; Hess, C.; Goldberger, J.; He, R. R.; Sun, Y. G.;
Immunogold Colloids. Anal. Chem. 1999, 71, 4903−4908. Xia, Y. N.; Yang, P. D. Langmuir-Blodgett Silver Nanowire Monolayers
(60) Li, J.-M.; Wei, C.; Ma, W.-F.; An, Q.; Guo, J.; Hu, J.; Wang, C.- for Molecular Sensing Using Surface-Enhanced Raman Spectroscopy.
C. Multiplexed SERS Detection of DNA Targets in a Sandwich- Nano Lett. 2003, 3, 1229−1233.
Hybridization Assay Using SERS-Encoded Core-Shell Nanospheres. J. (81) Pei, Y. W.; Wang, Z. Y.; Zong, S. F.; Cui, Y. P. Highly Sensitive
Mater. Chem. 2012, 22, 12100−12106. SERS-Based Immunoassay with Simultaneous Utilization of Self-
(61) Zong, S.; Wang, Z.; Zhang, R.; Wang, C.; Xu, S.; Cui, Y. A Assembled Substrates of Gold Nanostars and Aggregates of Gold
Multiplex and Straightforward Aqueous Phase Immunoassay Protocol Nanostars. J. Mater. Chem. B 2013, 1, 3992−3998.
Through the Combination of SERS-Fluorescence Dual Mode (82) Liu, M.; Wang, Z. Y.; Zong, S. F.; Zhang, R. H.; Zhu, D.; Xu, S.
Nanoprobes and Magnetic Nanobeads. Biosens. Bioelectron. 2013, 41, H.; Wang, C. L.; Cui, Y. P. SERS-Based DNA Detection in Aqueous
745−751. Solutions Using Oligonucleotide-Modified Ag Nanoprisms and Gold
(62) Zheng, F.; Cheng, Y.; Wang, J.; Lu, J.; Zhang, B.; Zhao, Y.; Gu, Nanoparticles. Anal. Bioanal. Chem. 2013, 405, 6131−6136.
Z. Aptamer-Functionalized Barcode Particles for the Capture and (83) Song, C. Y.; Wang, Z. Y.; Zhang, R. H.; Yang, J.; Tan, X. B.; Cui,
Detection of Multiple Types of Circulating Tumor Cells. Adv. Mater. Y. P. Highly Sensitive Immunoassay Based on Raman Reporter-
2014, 26, 7333−7338. Labeled Immuno-Au Aggregates and SERS-Active Immune Substrate.
(63) Lee, M.; Lee, K.; Kim, K. H.; Oh, K. W.; Choo, J. SERS-Based Biosens. Bioelectron. 2009, 25, 826−831.
Immunoassay Using a Gold Array-Embedded Gradient Microfluidic (84) Wang, Z.; Zong, S.; Chen, H.; Wu, H.; Cui, Y. Silica Coated
Chip. Lab Chip 2012, 12, 3720−3727. Gold Nanoaggregates Prepared by Reverse Microemulsion Method:
(64) Abbas, A.; Brimer, A.; Slocik, J. M.; Tian, L. M.; Naik, R. R.; Dual Mode Probes for Multiplex Immunoassay Using SERS and
Singamaneni, S. Multifunctional Analytical Platform on a Paper Strip: Fluorescence. Talanta 2011, 86, 170−177.
Separation, Preconcentration, and Subattomolar Detection. Anal. (85) Lim, D. K.; Jeon, K. S.; Kim, H. M.; Nam, J. M.; Suh, Y. D.
Chem. 2013, 85, 3977−3983. Nanogap-Engineerable Raman-Active Nanodumbbells for Single-
(65) Zhu, D.; Wang, Z. Y.; Zong, S. F.; Chen, H.; Chen, P.; Cui, Y. P. Molecule Detection. Nat. Mater. 2010, 9, 60−67.
Wavenumber-Intensity Joint SERS Encoding Using Silver Nano- (86) Dadosh, T.; Sperling, J.; Bryant, G. W.; Breslow, R.; Shegai, T.;
particles for Tumor Cell Targeting. RSC Adv. 2014, 4, 60936−60942. Dyshel, M.; Haran, G.; Bar-Joseph, I. Plasmonic Control of the Shape

7952 DOI: 10.1021/acs.chemrev.7b00027


Chem. Rev. 2017, 117, 7910−7963
Chemical Reviews Review

of the Raman Spectrum of a Single Molecule in a Silver Nanoparticle (104) Wu, X.; Wang, Z. Y.; Zhu, D.; Zong, S. F.; Yang, L. P.; Zhong,
Dimer. ACS Nano 2009, 3, 1988−1994. Y.; Cui, Y. P. pH and Thermo Dual-Stimuli-Responsive Drug Carrier
(87) Osberg, K. D.; Rycenga, M.; Harris, N.; Schmucker, A. L.; Based on Mesoporous Silica Nanoparticles Encapsulated in a
Langille, M. R.; Schatz, G. C.; Mirkin, C. A. Dispersible Gold Nanorod Copolymer-Lipid Bilayer. ACS Appl. Mater. Interfaces 2013, 5,
Dimers with Sub-5 nm Gaps as Local Amplifiers for Surface-Enhanced 10895−10903.
Raman Scattering. Nano Lett. 2012, 12, 3828−3832. (105) Li, J. F.; Huang, Y. F.; Ding, Y.; Yang, Z. L.; Li, S. B.; Zhou, X.
(88) Alber, I.; Sigle, W.; Muller, S.; Neumann, R.; Picht, O.; Rauber, S.; Fan, F. R.; Zhang, W.; Zhou, Z. Y.; Wu, D. Y.; Tian, Z. Q.; et al.
M.; van Aken, P. A.; Toimil-Molares, M. E. Visualization of Multipolar Shell-Isolated Nanoparticle-Enhanced Raman Spectroscopy. Nature
Longitudinal and Transversal Surface Plasmon Modes in Nanowire 2010, 464, 392−395.
Dimers. ACS Nano 2011, 5, 9845−9853. (106) Jiang, T.; Wang, X.; Zhou, J.; Chen, D.; Zhao, Z. Hydrothermal
(89) Gopalakrishnan, A.; Chirumamilla, M.; De Angelis, F.; Toma, Synthesis of Ag@MSiO2@Ag Three Core-Shell Nanoparticles and
A.; Zaccaria, R. P.; Krahne, R. Bimetallic 3D Nanostar Dimers in Ring Their Sensitive and Stable SERS Properties. Nanoscale 2016, 8, 4908−
Cavities: Recyclable and Robust Surface-Enhanced Raman Scattering 4914.
Substrates for Signal Detection from Few Molecules. ACS Nano 2014, (107) Toh, S. Y.; Citartan, M.; Gopinath, S. C. B.; Tang, T. H.
8, 7986−7994.
Aptamers as a Replacement for Antibodies in Enzyme-Linked
(90) Stranahan, S. M.; Titus, E. J.; Willets, K. A. Discriminating
Immunosorbent Assay. Biosens. Bioelectron. 2015, 64, 392−403.
Nanoparticle Dimers from Higher Order Aggregates through Wave-
(108) Fang, X.; Tan, W. Aptamers Generated from Cell-SELEX for
length-Dependent SERS Orientational Imaging. ACS Nano 2012, 6,
Molecular Medicine: A Chemical Biology Approach. Acc. Chem. Res.
1806−1813.
(91) Tripathy, S.; Marty, R.; Lin, V. K.; Teo, S. L.; Ye, E. Y.; Arbouet, 2010, 43, 48−57.
A.; Saviot, L.; Girard, C.; Han, M. Y.; Mlayah, A. Acousto-Plasmonic (109) Tan, W.; Donovan, M. J.; Jiang, J. Aptamers from Cell-Based
and Surface-Enhanced Raman Scattering Properties of Coupled Gold Selection for Bioanalytical Applications. Chem. Rev. 2013, 113, 2842−
Nanospheres/Nanodisk Trimers. Nano Lett. 2011, 11, 431−437. 2862.
(92) Zhao, B.; Shen, J.; Chen, S.; Wang, D.; Li, F.; Mathur, S.; Song, (110) Zhang, X. F.; Du, X. Z. Carbon Nanodot-Decorated Ag@SiO2
S.; Fan, C. Gold Nanostructures Encoded by Non-Fluorescent Small Nanoparticles for Fluorescence and Surface-Enhanced Raman
Molecules in PolyA-Mediated Nanogaps as Universal SERS Nanotags Scattering Immunoassays. ACS Appl. Mater. Interfaces 2016, 8,
for Recognizing Various Bioactive Molecules. Chem. Sci. 2014, 5, 1033−1040.
4460−4466. (111) Liang, J. J.; Liu, H. W.; Huang, C. H.; Yao, C. Z.; Fu, Q. Q.; Li,
(93) Song, J.; Duan, B.; Wang, C.; Zhou, J.; Pu, L.; Fang, Z.; Wang, X. Q.; Cao, D. L.; Luo, Z.; Tang, Y. Aggregated Silver Nanoparticles
P.; Lim, T. T.; Duan, H. SERS-Encoded Nanogapped Plasmonic Based Surface-Enhanced Raman Scattering Enzyme-Linked Immuno-
Nanoparticles: Growth of Metallic Nanoshell by Templating Redox- sorbent Assay for Ultrasensitive Detection of Protein Biomarkers and
Active Polymer Brushes. J. Am. Chem. Soc. 2014, 136, 6838−6841. Small Molecules. Anal. Chem. 2015, 87, 5790−5796.
(94) Lim, D.-K.; Jeon, K.-S.; Hwang, J.-H.; Kim, H.; Kwon, S.; Suh, Y. (112) Li, M.; Kang, J. W.; Sukumar, S.; Dasari, R. R.; Barman, I.
D.; Nam, J.-M. Highly Uniform and Reproducible Surface-Enhanced Multiplexed Detection of Serological Cancer Markers with Plasmon-
Raman Scattering from DNA-Tailorable Nanoparticles with 1-nm Enhanced Raman Spectro-Immunoassay. Chem. Sci. 2015, 6, 3906−
Interior Gap. Nat. Nanotechnol. 2011, 6, 452−460. 3914.
(95) Zong, S. F.; Wang, L.; Chen, C.; Lu, J.; Zhu, D.; Zhang, Y. Z.; (113) Ye, J.; Chen, Y.; Liu, Z. A Boronate Affinity Sandwich Assay:
Wang, Z. Y.; Cui, Y. P. Facile Detection of Tumor-Derived Exosomes An Appealing Alternative to Immunoassays for the Determination of
Using Magnetic Nanobeads and SERS Nanoprobes. Anal. Methods Glycoproteins. Angew. Chem., Int. Ed. 2014, 53, 10386−10389.
2016, 8, 5001−5008. (114) Zheng, P.; Li, M.; Jurevic, R.; Cushing, S. K.; Liu, Y. X.; Wu, N.
(96) Zong, S. F.; Wang, Z. Y.; Yang, J.; Wang, C. L.; Xu, S. H.; Cui, Y. Q. A Gold Nanohole Array Based Surface-Enhanced Raman Scattering
P. A SERS and Fluorescence Dual Mode Cancer Cell Targeting Probe Biosensor for Detection of Silver(I) and Mercury(II) in Human Saliva.
Based on Silica Coated Au@Ag Core-Shell Nanorods. Talanta 2012, Nanoscale 2015, 7, 11005−11012.
97, 368−375. (115) Zou, F. M.; Zhou, H. J.; Tan, T. V.; Kim, J.; Koh, K.; Lee, J.
(97) Zhang, Y. Z.; Wang, Z. Y.; Wu, L.; Pei, Y. W.; Chen, P.; Cui, Y. Dual-Mode SERS-Fluorescence Immunoassay Using Graphene
P. Rapid Simultaneous Detection of Multi-Pesticide Residues on Apple Quantum Dot Labeling on One-Dimensional Aligned Magneto-
Using SERS Technique. Analyst 2014, 139, 5148−5154. plasmonic Nanoparticles. ACS Appl. Mater. Interfaces 2015, 7,
(98) Zong, S. F.; Wang, Z. Y.; Chen, H.; Yang, J.; Cui, Y. P. Surface 12168−12175.
Enhanced Raman Scattering Traceable and Glutathione Responsive (116) Liu, J.; Yin, D.; Wang, S.; Chen, H.-Y.; Liu, Z. Probing Low-
Nanocarrier for the Intracellular Drug Delivery. Anal. Chem. 2013, 85,
Copy-Number Proteins in a Single Living Cell. Angew. Chem., Int. Ed.
2223−2230.
2016, 55, 13215−13218.
(99) Huang, S.; Ling, X.; Liang, L.; Song, Y.; Fang, W.; Zhang, J.;
(117) Yang, J.; Palla, M.; Bosco, F. G.; Rindzevicius, T.; Alstrom, T.
Kong, J.; Meunier, V.; Dresselhaus, M. S. Molecular Selectivity of
S.; Schmidt, M. S.; Boisen, A.; Ju, J. Y.; Lin, Q. Surface-Enhanced
Graphene-Enhanced Raman Scattering. Nano Lett. 2015, 15, 2892−
2901. Raman Spectroscopy Based Quantitative Bioassay on Aptamer-
(100) Zhang, N.; Tong, L.; Zhang, J. Graphene-Based Enhanced Functionalized Nanopillars Using Large-Area Raman Mapping. ACS
Raman Scattering toward Analytical Applications. Chem. Mater. 2016, Nano 2013, 7, 5350−5359.
28, 6426−6435. (118) Schmit, V. L.; Martoglio, R.; Carron, K. T. Lab-on-a-Bubble
(101) Wang, Y. Q.; Chen, L. X.; Liu, P. Biocompatible Triplex Ag@ Surface Enhanced Raman Indirect Immunoassay for Cholera. Anal.
SiO2@mTiO2 Core-Shell Nanoparticles for Simultaneous Fluores- Chem. 2012, 84, 4233−4236.
cence-SERS Bimodal Imaging and Drug Delivery. Chem. - Eur. J. 2012, (119) Wei, L. J.; Jin, B.; Dai, S. Polymer Microbead-Based Surface
18, 5935−5943. Enhanced Raman Scattering Immunoassays. J. Phys. Chem. C 2012,
(102) Knauer, M.; Ivleva, N. P.; Niessner, R.; Haisch, C. A Flow- 116, 17174−17181.
Through Microarray Cell for the Online SERS Detection of Antibody- (120) Tai, Y.; Wang, L.; Yan, G.; Gao, J.-m.; Yu, H.; Zhang, L. Recent
Captured E. coli Bacteria. Anal. Bioanal. Chem. 2012, 402, 2663−2667. Research Progress on the Preparation and Application of Magnetic
(103) Kim, K.; Lee, Y. M.; Lee, H. B.; Shin, K. S. Silver Salts of Nanospheres. Polym. Int. 2011, 60, 976−994.
Aromatic Thiols Applicable as Core Materials of Molecular Sensors (121) Iranifam, M. Analytical Applications of Chemiluminescence-
Operating via SERS and Fluorescence. Biosens. Bioelectron. 2009, 24, Detection Systems Assisted by Magnetic Microparticles and Nano-
3615−3621. particles. TrAC, Trends Anal. Chem. 2013, 51, 51−70.

7953 DOI: 10.1021/acs.chemrev.7b00027


Chem. Rev. 2017, 117, 7910−7963
Chemical Reviews Review

(122) Dunbar, S. A. Applications of Luminex (R) xMAP (TM) Spectroscopy for the Detection of Staphylococcus aureus Based on
technology for Rapid, High-Throughput Multiplexed Nucleic Acid Aptamer Recognition. ACS Appl. Mater. Interfaces 2015, 7, 20919−
Detection. Clin. Chim. Acta 2006, 363, 71−82. 20929.
(123) Wilson, R.; Cossins, A. R.; Spiller, D. G. Encoded Microcarriers (140) Xiong, K.; Wei, W.; Jin, Y.; Wang, S.; Zhao, D.; Wang, S.; Gao,
for High-Throughput Multiplexed Detection. Angew. Chem., Int. Ed. X.; Qiao, C.; Yue, H.; Ma, G.; et al. Biomimetic Immuno-
2006, 45, 6104−6117. Magnetosomes for High-Performance Enrichment of Circulating
(124) Yan, W. J.; Yang, L. P.; Zhuang, H.; Wu, H. Z.; Zhang, J. H. Tumor Cells. Adv. Mater. 2016, 28, 7929−7935.
Engineered ″hot″ Core-Shell Nanostructures for Patterned Detection (141) Craighead, H. Future Lab-on-a-Chip Technologies for
of Chloramphenicol. Biosens. Bioelectron. 2016, 78, 67−72. Interrogating Individual Molecules. Nature 2006, 442, 387−393.
(125) Li, J.; Dong, S. J.; Tong, J. J.; Zhu, P. Z.; Diao, G. W.; Yang, Z. (142) Huang, J. A.; Zhang, Y. L.; Ding, H.; Sun, H. B. SERS-Enabled
J. 3D Ordered Silver Nanoshells Silica Photonic Crystal Beads for Lab-on-a-Chip Systems. Adv. Opt. Mater. 2015, 3, 618−633.
Multiplex Encoded SERS Bioassay. Chem. Commun. 2016, 52, 284− (143) Bange, A.; Halsall, H. B.; Heineman, W. R. Microfluidic
287. Immunosensor Systems. Biosens. Bioelectron. 2005, 20, 2488−2503.
(126) Lee, J. H.; Xu, P. F.; Domaille, D. W.; Choi, C.; Jin, S.; Cha, J. (144) Menegatti, E.; Berardi, D.; Messina, M.; Ferrante, I.; Giachino,
N. M13 Bacteriophage as Materials for Amplified Surface Enhanced O.; Spagnolo, B.; Restagno, G.; Cognolato, L.; Roccatello, D. Lab-on-
Raman Scattering Protein Sensing. Adv. Funct. Mater. 2014, 24, 2079− a-Chip: Emerging Analytical Platforms for Immune-Mediated
2084. Diseases. Autoimmun. Rev. 2013, 12, 814−820.
(127) Feng, J. J.; Wu, X. L.; Ma, W.; Kuang, H.; Xu, L. G.; Xu, C. L. A (145) Gao, R.; Ko, J.; Cha, K.; Jeon, J. H.; Rhie, G.-e.; Choi, J.;
SERS Active Bimetallic Core-Satellite Nanostructure for the Ultra- deMello, A. J.; Choo, J. Fast and Sensitive Detection of An Anthrax
sensitive Detection of Mucin-1. Chem. Commun. 2015, 51, 14761− Biomarker Using SERS-Based Solenoid Microfluidic Sensor. Biosens.
14763. Bioelectron. 2015, 72, 230−236.
(128) Ma, M.; Yin, H. H.; Xu, L. G.; Wu, X. L.; Kuang, H.; Wang, L. (146) Li, J.; Skeete, Z.; Shan, S. Y.; Yan, S.; Kurzatkowska, K.; Zhao,
B.; Xu, C. L. Ultrasensitive Aptamer-Based SERS Detection of PSAs by W.; Ngo, Q. M.; Holubovska, P.; Luo, J.; Hepel, M.; et al. Surface
Heterogeneous Satellite Nanoassemblies. Chem. Commun. 2014, 50, Enhanced Raman Scattering Detection of Cancer Biomarkers with
9737−9740. Bifunctional Nanocomposite Probes. Anal. Chem. 2015, 87, 10698−
(129) Li, A. K.; Tang, L. J.; Song, D.; Song, S. S.; Ma, W.; Xu, L. G.; 10702.
Kuang, H.; Wu, X. L.; Liu, L. Q.; Chen, X.; et al. A SERS-Active Sensor (147) Kaminska, A.; Witkowska, E.; Winkler, K.; Dziecielewski, I.;
Based on Heterogeneous Gold Nanostar Core-Silver Nanoparticle Weyher, J. L.; Waluk, J. Detection of Hepatitis B Virus Antigen From
Satellite Assemblies for Ultrasensitive Detection of AflatoxinB1. Human Blood: SERS Immunoassay in a Microfluidic System. Biosens.
Nanoscale 2016, 8, 1873−1878. Bioelectron. 2015, 66, 461−467.
(130) Wu, X. L.; Chen, X.; Gao, F. L.; Ma, W.; Xu, L. G.; Kuang, H.; (148) Mao, H.; Wu, W.; She, D.; Sun, G.; Lv, P.; Xu, J. Microfluidic
Li, A. K.; Xu, C. L. SERS Encoded Nanoparticle Heterodimers for the Surface-Enhanced Raman Scattering Sensors Based on Nanopillar
Ultrasensitive Detection of Folic Acid. Biosens. Bioelectron. 2016, 75, Forests Realized by an Oxygen-Plasma-Stripping-of-Photoresist
55−58. Technique. Small 2014, 10, 127−134.
(131) Xu, L.; Yan, W.; Ma, W.; Kuang, H.; Wu, X.; Liu, L.; Zhao, Y.; (149) Zhao, Y. Q.; Zhang, Y. L.; Huang, J. A.; Zhang, Z. Y.; Chen, X.
Wang, L.; Xu, C. SERS Encoded Silver Pyramids for Attomolar F.; Zhang, W. J. Plasmonic Nanopillar Array Embedded Microfluidic
Detection of Multiplexed Disease Biomarkers. Adv. Mater. 2015, 27, Chips: An In situ SERS Monitoring Platform. J. Mater. Chem. A 2015,
1706−1711. 3, 6408−6413.
(132) MacLaughlin, C. M.; Mullaithilaga, N.; Yang, G. S.; Ip, S. Y.; (150) Yick, S.; Han, Z. J.; Ostrikov, K. Atmospheric Microplasma-
Wang, C.; Walker, G. C. Surface-Enhanced Raman Scattering Dye- Functionalized 3D Microfluidic Strips within Dense Carbon Nanotube
Labeled Au Nanoparticles for Triplexed Detection of Leukemia and Arrays Confine Au Nanodots for SERS Sensing. Chem. Commun. 2013,
Lymphoma Cells and SERS Flow Cytometry. Langmuir 2013, 29, 49, 2861−2863.
1908−1919. (151) De Angelis, F.; Malerba, M.; Patrini, M.; Miele, E.; Das, G.;
(133) Li, C. Y.; Ma, C.; Wang, F.; Xi, Z. J.; Wang, Z. F.; Deng, Y.; He, Toma, A.; Zaccaria, R. P.; Di Fabrizio, E. 3D Hollow Nanostructures
N. Y. Preparation and Biomedical Applications of Core-Shell Silica/ as Building Blocks for Multifunctional Plasmonics. Nano Lett. 2013,
Magnetic Nanoparticle Composites. J. Nanosci. Nanotechnol. 2012, 12, 13, 3553−3558.
2964−2972. (152) La Rocca, R.; Messina, G. C.; Dipalo, M.; Shalabaeva, V.; De
(134) Amram, D.; Rabkin, E. Core(Fe)-Shell(Au) Nanoparticles Angelis, F. Out-of-Plane Plasmonic Antennas for Raman Analysis in
Obtained from Thin Fe/Au Bilayers Employing Surface Segregation. Living Cells. Small 2015, 11, 4632−4637.
ACS Nano 2014, 8, 10687−10693. (153) Messina, G. C.; Malerba, M.; Zilio, P.; Miele, E.; Dipalo, M.;
(135) Wang, Z. Y.; Zong, S. F.; Chen, H.; Wang, C. L.; Xu, S. H.; Ferrara, L.; De Angelis, F. Hollow Plasmonic Antennas for Broadband
Cui, Y. P. SERS-Fluorescence Joint Spectral Encoded Magnetic SERS Spectroscopy. Beilstein J. Nanotechnol. 2015, 6, 492−498.
Nanoprobes for Multiplex Cancer Cell Separation. Adv. Healthcare (154) Song, H.; Chen, D. L.; Ismagilov, R. F. Reactions in Droplets
Mater. 2014, 3, 1889−1897. in Microflulidic Channels. Angew. Chem., Int. Ed. 2006, 45, 7336−
(136) Wang, R.; Chon, H.; Lee, S.; Cheng, Z. Y.; Hong, S. H.; Yoon, 7356.
Y. H.; Choo, J. Highly Sensitive Detection of Hormone Estradiol E2 (155) Muhlig, A.; Bocklitz, T.; Labugger, I.; Dees, S.; Henk, S.;
Using Surface-Enhanced Raman Scattering Based Immunoassays for Richter, E.; Andres, S.; Merker, M.; Stockel, S.; Weber, K.; et al. LOC-
the Clinical Diagnosis of Precocious Puberty. ACS Appl. Mater. SERS: A Promising Closed System for the Identification of
Interfaces 2016, 8, 10665−10672. Mycobacteria. Anal. Chem. 2016, 88, 7998−8004.
(137) Baniukevic, J.; Boyaci, I. H.; Bozkurt, A. G.; Tamer, U.; (156) Gao, R.; Cheng, Z.; deMello, A. J.; Choo, J. Wash-Free
Ramanavicius, A.; Ramanaviciene, A. Magnetic Gold Nanoparticles in Magnetic Immunoassay of the PSA Cancer Marker Using SERS and
SERS-Based Sandwich Immunoassay for Antigen Detection by Well Droplet Microfluidics. Lab Chip 2016, 16, 1022−1029.
Oriented Antibodies. Biosens. Bioelectron. 2013, 43, 281−288. (157) Hatab, N. A.; Rouleau, C. M.; Retterer, S. T.; Eres, G.;
(138) Jun, B. H.; Noh, M. S.; Kim, J.; Kim, G.; Kang, H.; Kim, M. S.; Hatzinger, P. B.; Gu, B. An Integrated Portable Raman Sensor with
Seo, Y. T.; Baek, J.; Kim, J. H.; Park, J.; et al. Multifunctional Silver- Nanofabricated Gold Bowtie Array Substrates for Energetics
Embedded Magnetic Nanoparticles as SERS Nanoprobes and Their Detection. Analyst 2011, 136, 1697−1702.
Applications. Small 2010, 6, 119−125. (158) Zavaleta, C. L.; Garai, E.; Liu, J. T. C.; Sensarn, S.; Mandella,
(139) Wang, J. F.; Wu, X. Z.; Wang, C. W.; Shao, N. S.; Dong, P. T.; M. J.; Van de Sompel, D.; Friedland, S.; Van Dam, J.; Contag, C. H.;
Xiao, R.; Wang, S. Q. Magnetically Assisted Surface-Enhanced Raman Gambhir, S. S. A Raman-Based Endoscopic Strategy for Multiplexed

7954 DOI: 10.1021/acs.chemrev.7b00027


Chem. Rev. 2017, 117, 7910−7963
Chemical Reviews Review

Molecular Imaging. Proc. Natl. Acad. Sci. U. S. A. 2013, 110, E2288− (177) Simon, C. G.; Lin-Gibson, S. Combinatorial and High-
E2297. Throughput Screening of Biomaterials. Adv. Mater. 2011, 23, 369−
(159) Yang, X.; Tanaka, Z.; Newhouse, R.; Xu, Q.; Chen, B.; Chen, 387.
S.; Zhang, J. Z.; Gu, C. Portable Fiber Sensors Based on Surface- (178) Georgiou, G.; Ippolito, G. C.; Beausang, J.; Busse, C. E.;
Enhanced Raman scattering. Rev. Sci. Instrum. 2010, 81, 123103. Wardemann, H.; Quake, S. R. The Promise and Challenge of High-
(160) Dinish, U. S.; Balasundaram, G.; Chang, Y. T.; Olivo, M. Throughput Sequencing of the Antibody Repertoire. Nat. Biotechnol.
Sensitive Multiplex Detection of Serological Liver Cancer Biomarkers 2014, 32, 158−168.
Using SERS-Active Photonic Crystal Fiber Probe. J. Biophotonics 2014, (179) Giri, S.; Sykes, E. A.; Jennings, T. L.; Chan, W. C. W. Rapid
7, 956−965. Screening of Genetic Biomarkers of Infectious Agents Using Quantum
(161) Scaffidi, J. P.; Gregas, M. K.; Seewaldt, V.; Vo-Dinh, T. SERS- Dot Barcodes. ACS Nano 2011, 5, 1580−1587.
Based Plasmonic Nanobiosensing in Single Living Cells. Anal. Bioanal. (180) Zhang, F.; Shi, Q. H.; Zhang, Y. C.; Shi, Y. F.; Ding, K. L.;
Chem. 2009, 393, 1135−1141. Zhao, D. Y.; Stucky, G. D. Fluorescence Upconversion Microbarcodes
(162) Wang, Y.; Kang, S.; Khan, A.; Ruttner, G.; Leigh, S. Y.; Murray, for Multiplexed Biological Detection: Nucleic Acid Encoding. Adv.
M.; Abeytunge, S.; Peterson, G.; Rajadhyaksha, M.; Dintzis, S.; et al. Mater. 2011, 23, 3775−3779.
Quantitative Molecular Phenotyping with Topically Applied SERS (181) Zhao, Y. J.; Xie, Z. Y.; Gu, H. C.; Jin, L.; Zhao, X. W.; Wang, B.
Nanoparticles for Intraoperative Guidance of Breast Cancer P.; Gu, Z. Z. Multifunctional Photonic Crystal Barcodes From
Lumpectomy. Sci. Rep. 2016, 6, 21242. Microfluidics. NPG Asia Mater. 2012, 4, e25.
(163) Hu, J.; Wang, S. Q.; Wang, L.; Li, F.; Pingguan-Murphy, B.; Lu, (182) Birtwell, S. W.; Broder, G. R.; Roach, P. L.; Morgan, H.
T. J.; Xu, F. Advances in Paper-Based Point-of-Care Diagnostics. Multiplexed Suspension Array Platform for High-Throughput Protein
Biosens. Bioelectron. 2014, 54, 585−597. Assays. Biomed. Microdevices 2012, 14, 651−657.
(164) Nery, E. W.; Kubota, L. T. Sensing Approaches on Paper- (183) Lee, J.; Bisso, P. W.; Srinivas, R. L.; Kim, J. J.; Swiston, A. J.;
Based Devices: a Review. Anal. Bioanal. Chem. 2013, 405, 7573−7595. Doyle, P. S. Universal Process-Inert Encoding Architecture for
(165) Hoppmann, E. P.; Yu, W. W.; White, I. M. Highly Sensitive Polymer Microparticles. Nat. Mater. 2014, 13, 524−529.
and Flexible Inkjet Printed SERS Sensors on Paper. Methods 2013, 63, (184) Moran, E. J.; Sarshar, S.; Cargill, J. F.; Shahbaz, M. M.; Lio, A.;
219−224. Mjalli, A. M. M.; Armstrong, R. W. Radio-Frequency Tag Encoded
(166) Liu, J.; Zhou, J.; Tang, B.; Zeng, T.; Li, Y. L.; Li, J. L.; Ye, Y.; Combinatorial Library Method for the Discovery of Tripeptide-
Wang, X. G. Surface Enhanced Raman Scattering (SERS) Fabrics for Substituted Cinnamic Acid Inhibitors of the Protein-Tyrosine-
Trace Analysis. Appl. Surf. Sci. 2016, 386, 296−302. Phosphatase PTP 1B. J. Am. Chem. Soc. 1995, 117, 10787−10788.
(167) Große, S.; Wilke, P.; Börner, H. G. Easy Access to Functional (185) Nicolaou, K. C.; Xiao, X. Y.; Parandoosh, Z.; Senyei, A.; Nova,
Patterns on Cellulose Paper by Combining Laser Printing and M. P. Radiofrequency Encoded Combinatorial Chemistry. Angew.
Material-Specific Peptide Adsorption. Angew. Chem., Int. Ed. 2016, 55, Chem., Int. Ed. Engl. 1995, 34, 2289−2291.
(186) Benecky, M. J.; Post, D. R.; Schmitt, S. M.; Kochar, M. S.
11266−11270.
(168) De Angelis, F.; Gentile, F.; Mecarini, F.; Das, G.; Moretti, M.; Detection of Hepatitis B Surface Antigen in Whole Blood by Coupled
Candeloro, P.; Coluccio, M. L.; Cojoc, G.; Accardo, A.; Liberale, C.; Particle Light Scattering (Copalis(TM)). Clin. Chem. 1997, 43, 1764−
1770.
et al. Breaking the Diffusion Limit with Super-Hydrophobic Delivery
(187) Vaino, A. R.; Janda, K. D. Euclidean Shape-Encoded
of Molecules to Plasmonic Nanofocusing SERS Structures. Nat.
Combinatorial Chemical Libraries. Proc. Natl. Acad. Sci. U. S. A.
Photonics 2011, 5, 682−688.
2000, 97, 7692−7696.
(169) Miele, E.; Malerba, M.; Dipalo, M.; Rondanina, E.; Toma, A.;
(188) Liang, Y.; Abdelrahman, A. I.; Baranov, V.; Winnik, M. A. The
Angelis, F. D. Controlling Wetting and Self-Assembly Dynamics by
Synthesis and Characterization of Lanthanide-Encoded Poly(styrene-
Tailored Hydrophobic and Oleophobic Surfaces. Adv. Mater. 2014, 26, co-methacrylic acid) Microspheres. Polymer 2011, 52, 5040−5052.
4179−4183. (189) Akhras, M. S.; Pettersson, E.; Diamond, L.; Unemo, M.;
(170) Li, Z. J.; Zhu, K. X.; Zhao, Q.; Meng, A. The Enhanced SERS Okamoto, J.; Davis, R. W.; Pourmand, N. The Sequencing Bead Array
Effect of Ag/ZnO Nanoparticles Through Surface Hydrophobic (SBA), A Next-Generation Digital Suspension Array. PLoS One 2013,
Modification. Appl. Surf. Sci. 2016, 377, 23−29. 8, e76696.
(171) Leiterer, C.; Zopf, D.; Seise, B.; Jahn, F.; Weber, K.; Popp, J.; (190) Hayward, T. J.; Hong, B.; Vyas, K. N.; Palfreyman, J. J.;
Cialla-May, D.; Fritzsche, W. Fast Self-assembly of Silver Nanoparticle Cooper, J. F. K.; Jiang, Z.; Jeong, J. R.; Llandro, J.; Mitrelias, T.; Bland,
Monolayer in Hydrophobic Environment and Its Application as SERS J. A. C.; et al. Magnetic Micro-Barcodes for Molecular Tagging
Substrate. J. Nanopart. Res. 2014, 16, 2467. Applications. J. Phys. D: Appl. Phys. 2010, 43, 175001.
(172) Hou, X. M.; Zhang, X. L.; Chen, S. T.; Fang, Y.; Yan, J. L.; Li, (191) Liong, M.; Hoang, A. N.; Chung, J.; Gural, N.; Ford, C. B.;
N.; Qi, P. X. Facile Synthesis of SERS Active Ag Nanoparticles in the Min, C.; Shah, R. R.; Ahmad, R.; Fernandez-Suarez, M.; Fortune, S.
Presence of Tri-n-octylphosphine Sulfide. Appl. Surf. Sci. 2011, 257, M.; et al. Magnetic Barcode Assay for Genetic Detection of Pathogens.
4935−4940. Nat. Commun. 2013, 4, 1752.
(173) Wallace, R. A.; Charlton, J. J.; Kirchner, T. B.; Lavrik, N. V.; (192) Hill, H. D.; Mirkin, C. A. The Bio-Barcode Assay for the
Datskos, P. G.; Sepaniak, M. J. Superhydrophobic Analyte Detection of Protein and Nucleic Acid Targets Using DTT-Induced
Concentration Utilizing Colloid-Pillar Array SERS Substrates. Anal. Ligand Exchange. Nat. Protoc. 2006, 1, 324−336.
Chem. 2014, 86, 11819−11825. (193) Pregibon, D. C.; Toner, M.; Doyle, P. S. Multifunctional
(174) Yang, S. K.; Dai, X. M.; Stogin, B. B.; Wong, T. S. Encoded Particles for High-Throughput Biomolecule Analysis. Science
Ultrasensitive Surface-Enhanced Raman Scattering Detection in 2007, 315, 1393−1396.
Common Fluids. Proc. Natl. Acad. Sci. U. S. A. 2016, 113, 268−273. (194) Leng, Y. K.; Sun, K.; Chen, X. Y.; Li, W. W. Suspension Arrays
(175) Tabakman, S. M.; Lau, L.; Robinson, J. T.; Price, J.; Sherlock, Based on Nanoparticle-Encoded Microspheres for High-Throughput
S. P.; Wang, H. L.; Zhang, B.; Chen, Z.; Tangsombatvisit, S.; Jarrell, J. Multiplexed Detection. Chem. Soc. Rev. 2015, 44, 5552−5595.
A.; et al. Plasmonic Substrates for Multiplexed Protein Microarrays (195) Jun, B. H.; Kang, H.; Lee, Y. S.; Jeong, D. H. Fluorescence-
with Femtomolar Sensitivity and Broad Dynamic Range. Nat. Based Multiplex Protein Detection Using Optically Encoded Microbe-
Commun. 2011, 2, 466−474. ads. Molecules 2012, 17, 2474−2490.
(176) Zhang, B.; Kumar, R. B.; Dai, H. J.; Feldman, B. J. A Plasmonic (196) Gellner, M.; Kompe, K.; Schlucker, S. Multiplexing with SERS
Chip for Biomarker Discovery and Diagnosis of Type 1 Diabetes. Nat. Labels Using Mixed SAMs of Raman Reporter Molecules. Anal.
Med. 2014, 20, 948−953. Bioanal. Chem. 2009, 394, 1839−1844.

7955 DOI: 10.1021/acs.chemrev.7b00027


Chem. Rev. 2017, 117, 7910−7963
Chemical Reviews Review

(197) Liu, Y.; Wu, P. Y. Meditating Metal Coenhanced Fluorescence (214) Burnette, W. N. Western Blotting-Electrophoretic Transfer of
and SERS Around Gold Nanoaggregates in Nanosphere as Bifunc- Proteins from Sodium Dodecyl Sulfate-Polyacrylamide Gels to
tional Biosensor for Multiple DNA Targets. ACS Appl. Mater. Interfaces Unmodified Nitrocellulose and Radiographic Detection with Antibody
2013, 5, 5832−5844. and Radioiodinated Protein A. Anal. Biochem. 1981, 112, 195−203.
(198) Zhou, X. Y.; Li, R. Y.; Wu, X. F.; Li, Z. J. A Sensitive, (215) Nicoletti, I.; Migliorati, G.; Pagliacci, M. C.; Grignani, F.;
Switchable and Biocompatible Surface Enhanced Raman Scattering- Riccardi, C. A Rapid and Simple Method for Measuring Thymocyte
Fluorescence Dual Mode Probe Using Bipyramid Gold Nanocrystal- Apotosis by Propidium Iodide Staining and Flow-Cytometry. J.
Gold Nanoclusters for High-Throughput Biodetection. Anal. Methods Immunol. Methods 1991, 139, 271−279.
2014, 6, 2862−2869. (216) Aebersold, R.; Mann, M. Mass Spectrometry-Based Proteo-
(199) Kang, H.; Jeong, S.; Koh, Y.; Cha, M. G.; Yang, J. K.; Kyeong, mics. Nature 2003, 422, 198−207.
S.; Kim, J.; Kwak, S. Y.; Chang, H. J.; Lee, H.; et al. Direct (217) Southern, E. M. Detection of Specitic Sequences Among DNA
Identification of On-Bead Peptides Using Surface-Enhanced Raman Fragments Separated by Gel-Electrophoresis. J. Mol. Biol. 1975, 98,
Spectroscopic Barcoding System for High-Throughput Bioanalysis. Sci. 503−508.
Rep. 2015, 5, 10144. (218) Heid, C. A.; Stevens, J.; Livak, K. J.; Williams, P. M. Real Time
(200) Kim, J. H.; Kang, H.; Kim, S.; Jun, B. H.; Kang, T.; Chae, J.; Quantitative PCR. Genome Res. 1996, 6, 986−994.
Jeong, S.; Kim, J.; Jeong, D. H.; Lee, Y. S. Encoding Peptide Pequences (219) Doering, W. E.; Piotti, M. E.; Natan, M. J.; Freeman, R. G.
with Surface-Enhanced Raman Spectroscopic Nanoparticles. Chem. SERS as a Foundation for Nanoscale, Optically Detected Biological
Commun. 2011, 47, 2306−2308. Labels. Adv. Mater. 2007, 19, 3100−3108.
(201) Li, R. M.; Zhang, Y. T.; Tan, J.; Wan, J. X.; Guo, J.; Wang, C. (220) Vendrell, M.; Maiti, K. K.; Dhaliwal, K.; Chang, Y.-T. Surface-
C. Dual-Mode Encoded Magnetic Composite Microsphere Based on Enhanced Raman Scattering in Cancer Detection and Imaging. Trends
Fluorescence Reporters and Raman Probes as Covert Tag for Biotechnol. 2013, 31, 249−257.
Anticounterfeiting Applications. ACS Appl. Mater. Interfaces 2016, 8, (221) Hurley, I. P.; Coleman, R. C.; Ireland, H. E.; Williams, J. H. H.
9384−9394. Measurement of Bovine IgG by Indirect Competitive ELISA as a
(202) Yamaguchi, A.; Fukuoka, T.; Takahashi, R.; Hara, R.; Utsumi, Means of Detecting Milk Adulteration. J. Dairy Sci. 2004, 87, 543−
Y. Dielectrophoresis-Enabled Surface Enhanced Raman Scattering on 549.
Gold-Decorated Polystyrene Microparticle in Micro-Optofluidic (222) Hurley, I. P.; Coleman, R. C.; Ireland, H. E.; Williams, J. H. H.
Devices for High-Sensitive Detection. Sens. Actuators, B 2016, 230, Use of Sandwich IgG ELISA for the Detection and Quantification of
94−100. Adulteration of Milk and Soft Cheese. Int. Dairy J. 2006, 16, 805−812.
(203) Su, X.; Zhang, J.; Sun, L.; Koo, T. W.; Chan, S.; Sundararajan, (223) Berg, B.; Cortazar, B.; Tseng, D.; Ozkan, H.; Feng, S.; Wei, Q.;
N.; Yamakawa, M.; Berlin, A. A. Composite Organic-Inorganic Chan, R. Y.-L.; Burbano, J.; Farooqui, Q.; Lewinski, M.; et al.
Nanoparticles (COINs) with Chemically Encoded Optical Signatures. Cellphone-Based Hand-Held Microplate Reader for Point-of-Care
Nano Lett. 2005, 5, 49−54. Testing of Enzyme-Linked Immunosorbent Assays. ACS Nano 2015,
(204) Lai, Y. M.; Sun, S. Q.; He, T.; Schlucker, S.; Wang, Y. L. 9, 7857−7866.
Raman-Encoded Microbeads for Spectral Multiplexing with SERS (224) Algaar, F.; Eltzov, E.; Vdovenko, M. M.; Sakharov, I. Y.; Fajs,
Detection. RSC Adv. 2015, 5, 13762−13767.
L.; Weidmann, M.; Mirazimi, A.; Marks, R. S. Fiber-Optic
(205) Han, X. X.; Chen, L.; Ji, W.; Xie, Y. F.; Zhao, B.; Ozaki, Y.
Immunosensor for Detection of Crimean-Congo Hemorrhagic Fever
Label-Free Indirect Immunoassay Using an Avidin-Induced Surface-
IgG Antibodies in Patients. Anal. Chem. 2015, 87, 8394−8398.
Enhanced Raman Scattering Substrate. Small 2011, 7, 316−320.
(225) Li, H.; Zhao, M.; Liu, W.; Chu, W.; Guo, Y. Polydimethylsilox-
(206) Israelsen, N. D.; Wooley, D.; Hanson, C.; Vargis, E. Rational
ane Microfluidic Chemiluminescence Immunodevice with the Signal
Design of Raman-Labeled Nanoparticles for a Ddual-Modality, Light
Amplification Strategy for Sensitive Detection of Human Immuno-
Scattering Immunoassay on a Polystyrene Substrate. J. Biol. Eng. 2016,
10, 2. globin G. Talanta 2016, 147, 430−436.
(207) Kang, T.; Yoo, S. M.; Yoon, I.; Lee, S. Y.; Kim, B. Patterned (226) Li, H. Q.; Wang, C. G.; Ma, Z. F.; Su, Z. M. Colorimetric
Multiplex Pathogen DNA Detection by Au Particle-on-Wire SERS Detection of Immunoglobulin G by Use of Functionalized Gold
Sensor. Nano Lett. 2010, 10, 1189−1193. Nanoparticles on Polyethylenimine Film. Anal. Bioanal. Chem. 2006,
(208) Zheng, J.; Hu, Y. P.; Bai, J. H.; Ma, C.; Li, J. S.; Li, Y. H.; Shi, 384, 1518−1524.
M. L.; Tan, W. H.; Yang, R. H. Universal Surface-Enhanced Raman (227) Shi, H.; Yuan, L.; Wu, Y.; Liu, S. Colorimetric Immunosensing
Scattering Amplification Detector for Ultrasensitive Detection of via Protein Functionalized Gold Nanoparticle Probe Combined with
Multiple Target Analytes. Anal. Chem. 2014, 86, 2205−2212. Atom Transfer Radical Polymerization. Biosens. Bioelectron. 2011, 26,
(209) Valdez, J.; Bawage, S.; Gomez, I.; Singh, S. R. Facile and Rapid 3788−3793.
Detection of Respiratory Syncytial Virus Using Metallic Nanoparticles. (228) Linder, V.; Verpoorte, E.; Thormann, W.; de Rooij, N. F.;
J. Nanobiotechnol. 2016, 14, 13. Sigrist, M. Surface Biopassivation of Replicated Poly(dimethylsiloxane)
(210) Wu, X.; Xia, Y.; Huang, Y.; Li, J.; Ruan, H.; Chen, T.; Luo, L.; Microfluidic Channels and Application to Heterogeneous Immuno-
Shen, Z.; Wu, A. Improved SERS-Active Nanoparticles with Various reaction with On-Chip Fluorescence Detection. Anal. Chem. 2001, 73,
Shapes for CTC Detection without Enrichment Process with 4181−4189.
Supersensitivity and High Specificity. ACS Appl. Mater. Interfaces (229) Wang, M.; Hou, W.; Mi, C. C.; Wang, W. X.; Xu, Z. R.; Teng,
2016, 8, 19928−19938. H. H.; Mao, C. B.; Xu, S. K. Immunoassay of Goat Antihuman
(211) Li, J.; Zhu, Z.; Zhu, B.; Ma, Y.; Lin, B.; Liu, R.; Song, Y.; Lin, Immunoglobulin G Antibody Based on Luminescence Resonance
H.; Tu, S.; Yang, C. Surface-Enhanced Raman Scattering Active Energy Transfer between Near-Infrared Responsive NaYF4:Yb, Er
Plasmonic Nanoparticles with Ultrasmall Interior Nanogap for Upconversion Fluorescent Nanoparticles and Gold Nanoparticles.
Multiplex Quantitative Detection and Cancer Cell Imaging. Anal. Anal. Chem. 2009, 81, 8783−8789.
Chem. 2016, 88, 7828−7836. (230) Lyon, L. A.; Musick, M. D.; Smith, P. C.; Reiss, B. D.; Pena, D.
(212) Feng, J. J.; Xu, L. G.; Cui, G.; Wu, X. L.; Ma, W.; Kuang, H.; J.; Natan, M. J. Surface Plasmon Resonance of Colloidal Au-Modified
Xu, C. L. Building SERS-Active Heteroassemblies for Ultrasensitive Gold Films. Sens. Actuators, B 1999, 54, 118−124.
Bisphenol A Detection. Biosens. Bioelectron. 2016, 81, 138−142. (231) Cao, C.; Sim, S. J. Signal Enhancement of Surface Plasmon
(213) Friguet, B.; Chaffotte, A. F.; Djavadiohaniance, L.; Goldberg, Resonance Immunoassay Using Enzyme Precipitation-Functionalized
M. E. Measurements of the True Affinity Constant In Solution of Gold Nanoparticles: A Femto Molar Level Measurement of Anti-
Antigen-Antibody Complexes by Enzyme-Linked Immunosorbent Glutamic Acid Decarboxylase Antibody. Biosens. Bioelectron. 2007, 22,
Assay. J. Immunol. Methods 1985, 77, 305−319. 1874−1880.

7956 DOI: 10.1021/acs.chemrev.7b00027


Chem. Rev. 2017, 117, 7910−7963
Chemical Reviews Review

(232) Chon, H.; Lim, C.; Ha, S.-M.; Ahn, Y.; Lee, E. K.; Chang, S. I.; to Improve SERS Sensitivity for Protein Detection. Anal. Bioanal.
Seong, G. H.; Choo, J. On-Chip Immunoassay Using Surface- Chem. 2014, 406, 1885−1894.
Enhanced Raman Scattering of Hollow Gold Nanospheres. Anal. (251) Lopez, A.; Lovato, F.; Oh, S. H.; Lai, Y. H.; Filbrun, S.;
Chem. 2010, 82, 5290−5295. Driskell, E. A.; Driskell, J. D. SERS Immunoassay Based on the
(233) Ishikawa, E.; Hashida, S.; Kohno, T.; Hirota, K.; Hashinaka, K.; Capture and Concentration of Antigen-Assembled Gold Nano-
Ishikawa, S. Principle and Applicaions of Ultrasenstice Enzyme- particles. Talanta 2016, 146, 388−393.
Immunoassay (Immune-Complex Transfer Enzyme-Immunoassay) for (252) Yang, J.; Zhen, L.; Ren, F.; Campbell, J.; Rorrer, G. L.; Wang,
Antibodies in Body-Fluids. J. Clin. Lab. Anal. 1993, 7, 376−393. A. X. Ultra-Sensitive Immunoassay Biosensors Using Hybrid
(234) Gapper, L. W.; Copestake, D. E. J.; Otter, D. E.; Indyk, H. E. Plasmonic-Biosilica Nanostructured Materials. J. Biophotonics 2015,
Analysis of Bovine Immunoglobulin G in Milk, Colostrum and Dietary 8, 659−667.
Supplements: a Review. Anal. Bioanal. Chem. 2007, 389, 93−109. (253) Chen, Y. Y.; Cheng, H. W.; Tram, K.; Zhang, S. F.; Zhao, Y.
(235) Zhu, X. Q.; Li, J.; He, H. P.; Huang, M.; Zhang, X. H.; Wang, H.; Han, L. Y.; Chen, Z. P.; Huan, S. Y. A Paper-Based Surface-
S. F. Application of Nanomaterials in the Bioanalytical Detection of Enhanced Resonance Raman Spectroscopic (SERRS) Immunoassay
Disease-Related Genes. Biosens. Bioelectron. 2015, 74, 113−133. Using Magnetic Separation and Enzyme-Catalyzed Reaction. Analyst
(236) Homola, J. Surface Plasmon Resonance Sensors for Detection 2013, 138, 2624−2631.
of Chemical and Biological Species. Chem. Rev. 2008, 108, 462−493. (254) Penn, M. A.; Drake, D. M.; Driskell, J. D. Accelerated Surface-
(237) Crawford, A. C.; Skuratovsky, A.; Porter, M. D. Sampling Enhanced Raman Spectroscopy (SERS)-Based Immunoassay on a
Error: Impact on the Quantitative Analysis of Nanoparticle-Based Gold-Plated Membrane. Anal. Chem. 2013, 85, 8609−8617.
Surface-Enhanced Raman Scattering Immunoassays. Anal. Chem. (255) Tian, S.; Zhou, Q.; Gu, Z. M.; Gu, X. F.; Zheng, J. W.
2016, 88, 6515−6522. Fabrication of a Bowl-Shaped Silver Cavity Substrate for SERS-Based
(238) Kho, K. W.; Dinish, U. S.; Kumar, A.; Olivo, M. Frequency Immunoassay. Analyst 2013, 138, 2604−2612.
Shifts in SERS for Biosensing. ACS Nano 2012, 6, 4892−4902. (256) Ahmed, F. E. Sample Preparation and Fractionation for
(239) Balzerova, A.; Fargasova, A.; Markova, Z.; Ranc, V.; Zboril, R. Proteome Analysis and Cancer Biomarker Discovery by Mass
Magnetically-Assisted Surface Enhanced Raman Spectroscopy (MA- Spectrometry. J. Sep. Sci. 2009, 32, 771−798.
SERS) for Label-Free Determination of Human Immunoglobulin G (257) Haga, H.; Patel, T. Molecular Diagnosis of Intrahepatic
(IgG) in Blood Using Fe3O4@Ag Nanocomposite. Anal. Chem. 2014, Cholangiocarcinoma. J. Hepato-Biliary-Pancreat. Sci. 2015, 22, 114−
86, 11107−11114. 123.
(240) Lin, C. C.; Yang, Y. M.; Chen, Y. F.; Yang, T.-S.; Chang, H. C. (258) Gavelli, G.; Giampalma, E. Sensitivity and Specificity of Chest
A New Protein A Assay Based on Raman Reporter Labeled
X-Ray Screening for Lung Cancer - Review Article. Cancer 2000, 89,
Immunogold Nanoparticles. Biosens. Bioelectron. 2008, 24, 178−183.
2453−2456.
(241) Kim, K.; Lee, Y. M.; Lee, H. B.; Shin, K. S. Silver-Coated Silica
(259) Kennedy, D. A.; Lee, T.; Seely, D. A Comparative Review of
Beads Applicable as Core Materials of Dual-Tagging Sensors
Thermography as a Breast Cancer Screening Technique. Integr. Cancer
Operating via SERS and MEF. ACS Appl. Mater. Interfaces 2009, 1,
Ther. 2009, 8, 9−16.
2174−2180.
(260) Ang, T. L.; Khor, C. J. L.; Gotoda, T. Diagnosis and
(242) Shin, M. H.; Hong, W.; Sa, Y.; Chen, L.; Jung, Y. J.; Wang, X.;
Endoscopic Resection of Early Gastric Cancer. Singapore Med. J. 2010,
Zhao, B.; Jung, Y. M. Multiple Detection of Proteins by SERS-Based
51, 93−100.
Immunoassay with Core Shell Magnetic Gold Nanoparticles. Vib.
(261) Smith, R. A.; Cokkinides, V.; Brawley, O. W. Cancer Screening
Spectrosc. 2014, 72, 44−49.
(243) Song, C. Y.; Min, L. H.; Zhou, N.; Yang, Y. J.; Su, S.; Huang, in the United States, 2009: A Review of Current American Cancer
W.; Wang, L. H. Synthesis of Novel Gold Mesoflowers as SERS Tags Society Guidelines and Issues in Cancer Screening. Ca-Cancer J. Clin.
for Immunoassay with Improved Sensitivity. ACS Appl. Mater. 2009, 59, 27−41.
Interfaces 2014, 6, 21842−21850. (262) Paterlini-Brechot, P.; Benali, N. L. Circulating Tumor Cells
(244) Xiao, R.; Wang, C. W.; Zhu, A. N.; Long, F. Single Functional (CTC) Detection: Clinical Impact and Future Directions. Cancer Lett.
Magnetic-Bead as Universal Biosensing Platform for Trace Analyte 2007, 253, 180−204.
Detection Using SERS-Nanobioprobe. Biosens. Bioelectron. 2016, 79, (263) Schwarzenbach, H.; Hoon, D. S. B.; Pantel, K. Cell-Free
661−668. Nucleic Acids as Biomarkers in Cancer Patients. Nat. Rev. Cancer
(245) Sun, J.; Xianyu, Y.; Jiang, X. Point-of-Care Biochemical Assays 2011, 11, 426−437.
Using Gold Nanoparticle-Implemented Microfluidics. Chem. Soc. Rev. (264) Srinivas, P. R.; Kramer, B. S.; Srivastava, S. Trends in
2014, 43, 6239−6253. Biomarker Research for Cancer Detection. Lancet Oncol. 2001, 2,
(246) Kim, I.; Junejo, I.; Lee, M.; Lee, S.; Lee, E. K.; Chang, S. I.; 698−704.
Choo, J. SERS-Based Multiple Biomarker Detection Using a Gold- (265) Zaenker, P.; Ziman, M. R. Serologic Autoantibodies as
Patterned Microarray Chip. J. Mol. Struct. 2012, 1023, 197−203. Diagnostic Cancer Biomarkers-A Review. Cancer Epidemiol., Bio-
(247) Song, W.; Mao, Z.; Liu, X. J.; Lu, Y.; Li, Z. S.; Zhao, B.; Lu, L. markers Prev. 2013, 22, 2161−2181.
H. Detection of Protein Deposition within Latent Fingerprints by (266) Stearns, V.; Yamauchi, H.; Hayes, D. F. Circulating Tumor
Surface-Enhanced Raman Spectroscopy Imaging. Nanoscale 2012, 4, Markers in Breast Cancer: Accepted Utilities and Novel Prospects.
2333−2338. Breast Cancer Res. Treat. 1998, 52, 239−259.
(248) Li, M.; Cushing, S. K.; Zhang, J. M.; Suri, S.; Evans, R.; Petros, (267) Zhang, A. H.; Sun, H.; Yan, G. L.; Han, Y.; Wang, X. J. Serum
W. P.; Gibson, L. F.; Ma, D. L.; Liu, Y. X.; Wu, N. Q. Three- Proteomics in Biomedical Research: A Systematic Review. Appl.
Dimensional Hierarchical Plasmonic Nano-Architecture Enhanced Biochem. Biotechnol. 2013, 170, 774−786.
Surface-Enhanced Raman Scattering Immunosensor for Cancer (268) Granger, J. H.; Granger, M. C.; Firpo, M. A.; Mulvihill, S. J.;
Biomarker Detection in Blood Plasma. ACS Nano 2013, 7, 4967− Porter, M. D. Toward Development of a Surface-Enhanced Raman
4976. Scattering (SERS)-Based Cancer Diagnostic Immunoassay Panel.
(249) Fu, C. C.; Gu, Y. J.; Wu, Z. Y.; Wang, Y. Y.; Xu, S. P.; Xu, W. Analyst 2013, 138, 410−416.
Q. Surface-Enhanced Raman Scattering (SERS) Biosensing Based on (269) Lee, M.; Lee, S.; Lee, J. H.; Lim, H. W.; Seong, G. H.; Lee, E.
Nanoporous Dielectric Waveguide Resonance. Sens. Actuators, B 2014, K.; Chang, S. I.; Oh, C. H.; Choo, J. Highly Reproducible
201, 173−176. Immunoassay of Cancer Markers on a Gold-Patterned Microarray
(250) Gu, X. F.; Yan, Y. R.; Jiang, G. Q.; Adkins, J.; Shi, J.; Jiang, G. Chip Using Surface-Enhanced Raman Scattering Imaging. Biosens.
M.; Tian, S. Using a Silver-Enhanced Microarray Sandwich Structure Bioelectron. 2011, 26, 2135−2141.

7957 DOI: 10.1021/acs.chemrev.7b00027


Chem. Rev. 2017, 117, 7910−7963
Chemical Reviews Review

(270) Worsley, G. J.; Attree, S. L.; Noble, J. E.; Horgan, A. M. Rapid on Aptamer-Functionalized Core-Shell Nanoparticles with Embedded
Duplex Immunoassay for Wound Biomarkers at the Point-of-Care. SERS Reporters. ACS Appl. Mater. Interfaces 2016, 8, 14389−14395.
Biosens. Bioelectron. 2012, 34, 215−220. (289) Kosaka, N.; Iguchi, H.; Ochiya, T. Circulating microRNA in
(271) Ko, J. H.; Lee, S.; Lee, E. K.; Chang, S. I.; Chen, L. X.; Yoon, S. Body Fluid: a New Potential Biomarker for Cancer Diagnosis and
Y.; Choo, J. SERS-Based Immunoassay of Tumor Marker VEGF Using Prognosis. Cancer Sci. 2010, 101, 2087−2092.
DNA Aptamers and Silica-Encapsulated Hollow Gold Nanospheres. (290) Mitchell, P. S.; Parkin, R. K.; Kroh, E. M.; Fritz, B. R.; Wyman,
Phys. Chem. Chem. Phys. 2013, 15, 5379−5385. S. K.; Pogosova-Agadjanyan, E. L.; Peterson, A.; Noteboom, J.;
(272) Nguyen, A. H.; Lee, J.; Choi, H. I.; Kwak, H. S.; Sim, S. J. O’Briant, K. C.; Allen, A.; et al. Circulating microRNAs as Stable
Fabrication of Plasmon Length-Based Surface Enhanced Raman Blood-Based Markers for Cancer Detection. Proc. Natl. Acad. Sci. U. S.
Scattering for Multiplex Detection on Microfluidic Device. Biosens. A. 2008, 105, 10513−10518.
Bioelectron. 2015, 70, 358−365. (291) Driskell, J. D.; Seto, A. G.; Jones, L. P.; Jokela, S.; Dluhy, R. A.;
(273) Ye, S. J.; Zhai, X. M.; Wu, Y. Y.; Kuang, S. P. Dual-Primer Self- Zhao, Y. P.; Tripp, R. A. Rapid microRNA (miRNA) Detection and
generation SERS Signal Amplification Assay for PDGF-BB Using Classification via Surface-Enhanced Raman Spectroscopy (SERS).
Label-Free Aptamer. Biosens. Bioelectron. 2016, 79, 130−135. Biosens. Bioelectron. 2008, 24, 917−922.
(274) He, Y.; Wang, Y.; Yang, X.; Xie, S. B.; Yuan, R.; Chai, Y. Q. (292) Driskell, J. D.; Primera-Pedrozo, O. M.; Dluhy, R. A.; Zhao, Y.;
Metal Organic Frameworks Combining CoFe2O4 Magnetic Nano- Tripp, R. A. Quantitative Surface-Enhanced Raman Spectroscopy
particles as Highly Efficient SERS Sensing Platform for Ultrasensitive Based Analysis of MicroRNA Mixtures. Appl. Spectrosc. 2009, 63,
Detection of N-Terminal Pro-Brain Natriuretic Peptide. ACS Appl. 1107−1114.
Mater. Interfaces 2016, 8, 7683−7690. (293) Chen, Y.; Chen, G.; Feng, S.; Pan, J.; Zheng, X.; Su, Y.; Chen,
(275) Lin, Y.; Xu, G. H.; Wei, F. D.; Zhang, A. X.; Yang, J.; Hu, Q. Y.; Huang, Z.; Lin, X.; Lan, F.; et al. Label-Free Serum Ribonucleic
Detection of CEA in Human Serum Using Surface-Enhanced Raman Acid Analysis for Colorectal Cancer Detection by Surface-Enhanced
Spectroscopy Coupled with Antibody-Modified Au and Gamma- Raman Spectroscopy and Multivariate Analysis. J. Biomed. Opt. 2012,
Fe2O3@Au Nanoparticles. J. Pharm. Biomed. Anal. 2016, 121, 135− 17, 067003.
140. (294) Guven, B.; Dudak, F. C.; Boyaci, I. H.; Tamer, U.; Ozsoz, M.
(276) Cao, C.; Zhang, J.; Wen, X. L.; Dodson, S. L.; Dao, N. T.; SERS-Based Direct and Sandwich Assay Methods for Mir-21
Wong, L. M.; Wang, S. J.; Li, S. Z.; Phan, A. T.; Xiong, Q. H. Detection. Analyst 2014, 139, 1141−1147.
Metamaterials-Based Label-Free Nanosensor for Conformation and (295) Kang, T.; Kim, H.; Lee, J. M.; Lee, H.; Choi, Y.-S.; Kang, G.;
Seo, M.-K.; Chung, B. H.; Jung, Y.; Kim, B. Ultra-Specific Zeptomole
Affinity Biosensing. ACS Nano 2013, 7, 7583−7591.
(277) Owens, P.; Phillipson, N.; Perumal, J.; O’Connor, G. M.; MicroRNA Detection by Plasmonic Nanowire Interstice Sensor with
Bi-Temperature Hybridization. Small 2014, 10, 4200−4206.
Olivo, M. Sensing of p53 and EGFR Biomarkers Using High Efficiency
(296) Chiado, A.; Novara, C.; Lamberti, A.; Geobaldo, F.; Giorgis, F.;
SERS Substrates. Biosensors 2015, 5, 664−677.
Rivolo, P. Immobilization of Oligonucleotides on Metal-Dielectric
(278) Li, X.; Zhang, Y.; Xue, B.; Kong, X.; Liu, X.; Tu, L.; Chang, Y.;
Nanostructures for miRNA Detection. Anal. Chem. 2016, 88, 9554−
Zhang, H. A SERS Nano-Tag-Based Fiber-Optic Strategy for In situ
9563.
Immunoassay in Unprocessed Whole Blood. Biosens. Bioelectron. 2017,
(297) Zheng, J.; Ma, D.; Shi, M.; Bai, J.; Li, Y.; Yang, J.; Yang, R. A
92, 517−522.
New Enzyme-Free Quadratic SERS Signal Amplification Approach for
(279) Wang, Y.; Vaidyanathan, R.; Shiddiky, M. J. A.; Trau, M.
Circulating microRNA Detection in Human Serum. Chem. Commun.
Enabling Rapid and Specific Surface-Enhanced Raman Scattering 2015, 51, 16271−16274.
Immunoassay Using Nanoscaled Surface Shear Forces. ACS Nano (298) Pang, Y.; Wang, C.; Wang, J.; Sun, Z.; Xiao, R.; Wang, S.
2015, 9, 6354−6362. Fe3O4@Ag Magnetic Nanoparticles for microRNA Capture and
(280) Zhou, L.; Zhou, J.; Feng, Z.; Wang, F. Y.; Xie, S. S.; Bu, S. Z. Duplex-Specific Nuclease Signal Amplification Based SERS Detection
Immunoassay for Tumor Markers in Human Serum Based on Si in Cancer Cells. Biosens. Bioelectron. 2016, 79, 574−580.
Nanoparticles and SiC@Ag SERS-Active Substrate. Analyst 2016, 141, (299) Li, X.; Zheng, F.; Ren, R. Detecting miRNA by Producing
2534−2541. RNA: a Sensitive Assay that Combines Rolling-Circle DNA Polymer-
(281) Iliuk, A. B.; Hu, L. H.; Tao, W. A. Aptamer in Bioanalytical ization and Rolling Rircle Transcription. Chem. Commun. 2015, 51,
Applications. Anal. Chem. 2011, 83, 4440−4452. 11976−11979.
(282) Willner, I.; Zayats, M. Electronic Aptamer-Based Sensors. (300) Zhang, H.; Liu, Y.; Gao, J.; Zhen, J. A Sensitive SERS
Angew. Chem., Int. Ed. 2007, 46, 6408−6418. Detection of miRNA Using a Label-Free Multifunctional Probe. Chem.
(283) Citartan, M.; Gopinath, S. C. B.; Tominaga, J.; Tan, S. C.; Commun. 2015, 51, 16836−16839.
Tang, T. H. Assays for Aptamer-Based Platforms. Biosens. Bioelectron. (301) Jahr, S.; Hentze, H.; Englisch, S.; Hardt, D.; Fackelmayer, F.
2012, 34, 1−11. O.; Hesch, R. D.; Knippers, R. DNA Fragments in the Blood Plasma of
(284) Kaur, H.; Kumar, R.; Babu, J. N.; Mittal, S. Advances in Arsenic Cancer Patients: Quantitations and Evidence for Their Origin From
Biosensor Development - A Comprehensive Review. Biosens. Apoptotic and Necrotic Cells. Cancer Res. 2001, 61, 1659−1665.
Bioelectron. 2015, 63, 533−545. (302) Ito, H.; Hasegawa, K.; Hasegawa, Y.; Nishimaki, T.;
(285) Cottat, M.; D’Andrea, C.; Yasukuni, R.; Malashikhina, N.; Hosomichi, K.; Kimura, S.; Ohba, M.; Yao, H.; Onimaru, M.; Inoue,
Grinyte, R.; Lidgi-Guigui, N.; Fazio, B.; Sutton, A.; Oudar, O.; I.; et al. Silver Nanoscale Hexagonal Column Chips for Detecting Cell-
Charnaux, N.; et al. High Sensitivity, High Selectivity SERS Detection Free DNA and Circulating Nucleosomes in Cancer Patients. Sci. Rep.
of MnSOD Using Optical Nanoantennas Functionalized with 2015, 5, 10455.
Aptamers. J. Phys. Chem. C 2015, 119, 15532−15540. (303) Zhou, Q.; Zheng, J.; Qing, Z.; Zheng, M.; Yang, J.; Yang, S.;
(286) Wu, X.; Fu, P.; Ma, W.; Xu, L.; Kuang, H.; Xu, C. SERS-Active Ying, L.; Yang, R. Detection of Circulating Tumor DNA in Human
Silver Nanoparticle Trimers for Sub-Attomolar Detection of Alpha Blood via DNA-Mediated Surface-Enhanced Raman Spectroscopy of
Fetoprotein. RSC Adv. 2015, 5, 73395−73398. Single-Walled Carbon Nanotubes. Anal. Chem. 2016, 88, 4759−4765.
(287) Wu, P.; Gao, Y.; Zhang, H.; Cai, C. X. Aptamer-Guided Silver- (304) Wee, E. J. H.; Wang, Y. L.; Tsao, S. C. H.; Trau, M. Simple,
Gold Bimetallic Nanostructures with Highly Active Surface-Enhanced Sensitive and Accurate Multiplex Detection of Clinically Important
Raman Scattering for Specific Detection and Near-Infrared Photo- Melanoma DNA Mutations in Circulating Tumour DNA with SERS
thermal Therapy of Human Breast Cancer Cells. Anal. Chem. 2012, 84, Nanotags. Theranostics 2016, 6, 1506−1513.
7692−7699. (305) Kong, K. V.; Leong, W. K.; Lam, Z.; Gong, T. X.; Goh, D.; Lau,
(288) Zhao, J. J.; Zhang, K.; Li, Y. X.; Ji, J.; Liu, B. H. High- W. K. O.; Olivo, M. A Rapid and Label-Free SERS Detection Method
Resolution and Universal Visualization of Latent Fingerprints Based for Biomarkers in Clinical Biofluids. Small 2014, 10, 5030−5034.

7958 DOI: 10.1021/acs.chemrev.7b00027


Chem. Rev. 2017, 117, 7910−7963
Chemical Reviews Review

(306) Li, Y. T.; Qu, L. L.; Li, D. W.; Song, Q. X.; Fathi, F.; Long, Y. (324) Wu, L.; Wang, J.; Feng, L.; Ren, J.; Wei, W.; Qu, X. Label-Free
T. Rapid and Sensitive In-situ Detection of Polar Antibiotics in Water Ultrasensitive Detection of Human Telomerase Activity Using
Using a Disposable Ag-Graphene Sensor Based on Electrophoretic Porphyrin-Functionalized Graphene and Electrochemiluminescence
Preconcentration and Surface-Enhanced Raman Spectroscopy. Biosens. Technique. Adv. Mater. 2012, 24, 2447−2452.
Bioelectron. 2013, 43, 94−100. (325) Zong, S.; Wang, Z.; Chen, H.; Hu, G.; Liu, M.; Chen, P.; Cui,
(307) Tang, L. J.; Li, S.; Han, F.; Liu, L. Q.; Xu, L. G.; Ma, W.; Y. Colorimetry and SERS Dual-Mode Detection of Telomerase
Kuang, H.; Li, A. K.; Wang, L. B.; Xu, C. L. SERS-Active Au@Ag Activity: Combining Rapid Screening with High Sensitivity. Nanoscale
Nanorod Dimers for Ultrasensitive Dopamine Detection. Biosens. 2014, 6, 1808−1816.
Bioelectron. 2015, 71, 7−12. (326) Zong, S.; Wang, Z.; Chen, H.; Cui, Y. Assessing Telomere
(308) Lee, S.; Chon, H.; Lee, J.; Ko, J.; Chung, B. H.; Lim, D. W.; Length Using Surface Enhanced Raman Scattering. Sci. Rep. 2015, 4,
Choo, J. Rapid and Sensitive Phenotypic Marker Detection on Breast 6977.
Cancer Cells Using Surface-Enhanced Raman Scattering (SERS) (327) Zong, S.; Chen, C.; Wang, Z.; Zhang, Y.; Cui, Y. Surface
Imaging. Biosens. Bioelectron. 2014, 51, 238−243. Enhanced Raman Scattering Based In situ Hybridization Strategy for
(309) Dinish, U. S.; Fu, C. Y.; Soh, K. S.; Ramaswamy, B.; Kumar, A.; Telomere Length Assessment. ACS Nano 2016, 10, 2950−2959.
Olivo, M. Highly Sensitive SERS Detection of Cancer Proteins in Low (328) Shi, M. L.; Zheng, J.; Liu, C. H.; Tan, G. X.; Qing, Z. H.; Yang,
Sample Volume Using Hollow Core Photonic Crystal Fiber. Biosens. S.; Yang, J. F.; Tan, Y. J.; Yang, R. H. SERS Assay of Telomerase
Bioelectron. 2012, 33, 293−298. Activity at Single-Cell Level and Colon Cancer Tissues via Quadratic
(310) Kong, K. V.; Lam, Z.; Goh, W. D.; Leong, W. K.; Olivo, M. Signal Amplification. Biosens. Bioelectron. 2016, 77, 673−680.
Metal Carbonyl-Gold Nanoparticle Conjugates for Live-Cell SERS (329) Wu, Z. T.; Liu, Y. Z.; Zhou, X. D.; Shen, A. G.; Hu, J. M. A
Imaging. Angew. Chem., Int. Ed. 2012, 51, 9796−9799. ″Turn-off″ SERS-Based Detection Platform for Ultrasensitive
(311) Wang, X. K.; Chen, L.; Fu, X. L.; Chen, L. X.; Ding, Y. J. Detection of Thrombin Based on Enzymatic Assays. Biosens.
Highly Sensitive Surface-Enhanced Raman Scattering Sensing of Bioelectron. 2013, 44, 10−15.
Heparin Based on Antiaggregation of Functionalized Silver Nano- (330) Yoon, J.; Choi, N.; Ko, J.; Kim, K.; Lee, S.; Choo, J. Highly
particles. ACS Appl. Mater. Interfaces 2013, 5, 11059−11065. Sensitive Detection of Thrombin Using SERS-Based Magnetic
(312) Fu, X.; Cheng, Z.; Yu, J.; Choo, P.; Chen, L.; Choo, J. A SERS- Aptasensors. Biosens. Bioelectron. 2013, 47, 62−67.
Based ;ateral Flow Assay Biosensor for Highly Sensitive Detection of (331) Srivastava, S. K.; Shalabney, A.; Khalaila, I.; Gruner, C.;
HIV-1 DNA. Biosens. Bioelectron. 2016, 78, 530−537. Rauschenbach, B.; Abdulhalim, I. SERS Biosensor Using Metallic
(313) Lee, I. H.; Lee, J. M.; Jung, Y. Controlled Protein Embedment Nano-Sculptured Thin Films for the Detection of Endocrine
onto Au/Ag Core-Shell Nanoparticles for Immuno-Labeling of Disrupting Compound Biomarker Vitellogenin. Small 2014, 10,
Nanosilver Surface. ACS Appl. Mater. Interfaces 2014, 6, 7659−7664. 3579−3587.
(314) Chon, H.; Lee, S.; Yoon, S. Y.; Chang, S. I.; Lim, D. W.; Choo, (332) Neng, J.; Harpster, M. H.; Wilson, W. C.; Johnson, P. A.
J. Simultaneous Immunoassay for the Detection of Two Lung Cancer Surface-Enhanced Raman Scattering (SERS) Detection of Multiple
Markers Using Functionalized SERS Nanoprobes. Chem. Commun. Viral Antigens Using Magnetic Capture of SERS-Active Nanoparticles.
2011, 47, 12515−12517. Biosens. Bioelectron. 2013, 41, 316−321.
(315) Zhao, Y.; Yang, X.; Li, H.; Luo, Y. D.; Yu, R. P.; Zhang, L. L.; (333) Collins, K.; Mitchell, J. R. Telomerase in the Human
Yang, Y. X.; Song, Q. J. Au Nanoflower-Ag Nanoparticle Assembled Organism. Oncogene 2002, 21, 564−579.
SERS-Active Substrates for Sensitive MC-LR Detection. Chem. (334) Shay, J. W.; Wright, W. E. Telomerase: A Target for Cancer
Commun. 2015, 51, 16908−16911. Therapeutics. Cancer Cell 2002, 2, 257−265.
(316) Robson, A. F.; Hupp, T. R.; Lickiss, F.; Ball, K. L.; Faulds, K.; (335) Kim, N. W.; Wu, F. Advances in Quantification and
Graham, D. Nanosensing Protein Allostery Using a Bivalent Mouse Characterization of Telomerase Activity by the Telomeric Repeat
Double Minute Two (MDM2) Assay. Proc. Natl. Acad. Sci. U. S. A. Amplification Protocol (TRAP). Nucleic Acids Res. 1997, 25, 2595−
2012, 109, 8073−8078. 2597.
(317) Ramya, A. N.; Joseph, M. M.; Nair, J. B.; Karunakaran, V.; (336) Hisatomi, H.; Kawaoto, Y.; Ohmura, Y.; Kuwahara, M.;
Narayanan, N.; Maiti, K. K. New Insight of Tetraphenylethylene-Based Yamauchi, T.; Hikiji, K. Detection of Telomerase Activity in
Raman Signatures for Targeted SERS Nanoprobe Construction Hepatocellular Carcinoma by Fluorescence-Based TRAP Method.
Toward Prostate Cancer Cell Detection. ACS Appl. Mater. Interfaces Int. Hepatol. Commun. 1996, 6, 29−35.
2016, 8, 10220−10225. (337) Nakamura, Y.; Hirose, M.; Matsuo, H.; Tsuyama, N.;
(318) Wu, Z. T.; Liu, Y. F.; Liu, Y. Z.; Xiao, H. M.; Shen, A. G.; Kamisango, K.; Ide, T. Simple, Rapid, Quantitative, and Sensitive
Zhou, X. D.; Hu, J. M. A Simple and Universal ″Turn-on″ Detection Detection of Telomere Repeats in Cell Lysate by a Hybridization
Platform for Proteases Based on Surface Enhanced Raman Scattering Protection Assay. Clin. Chem. 1999, 45, 1718−1724.
(SERS). Biosens. Bioelectron. 2015, 65, 375−381. (338) Gil, M. E.; Coetzer, T. L. Real-Time Quantitative PCR of
(319) Yan, J.; Su, S.; He, S. J.; He, Y.; Zhao, B.; Wang, D. F.; Zhang, Telomere Length. Mol. Biotechnol. 2004, 27, 169−172.
H. L.; Huang, Q.; Song, S. P.; Fan, C. Nano Rolling-Circle (339) Delara, J.; Wydner, K. L.; Hyland, K. M.; Ward, W. S.
Amplification for Enhanced SERS Hot Spots in Protein Microarray Fluorescent In-situ Hybridization of the Telomere Repeat Sequence in
Analysis. Anal. Chem. 2012, 84, 9139−9145. Hamster Sperm Nuclear-Structures. J. Cell. Biochem. 1993, 53, 213−
(320) Noble, J.; Attree, S.; Horgan, A.; Knight, A.; Kumarswami, N.; 221.
Porter, R.; Worsley, G. Optical Scattering Artifacts Observed in the (340) Xu, L. G.; Zhao, S.; Ma, W.; Wu, X. L.; Li, S.; Kuang, H.;
Development of Multiplexed Surface Enhanced Raman Spectroscopy Wang, L. B.; Xu, C. L. Multigaps Embedded Nanoassemblies Enhance
Nanotag Immunoassays. Anal. Chem. 2012, 84, 8246−8252. In situ Raman Spectroscopy for Intracellular Telomerase Activity
(321) Sato, S.; Takenaka, S. PCR-Free Telomerase Assay Using Sensing. Adv. Funct. Mater. 2016, 26, 1602−1608.
Chronocoulometry Coupled with Hexaammineruthenium(III) Chlor- (341) Luo, S.-C.; Sivashanmugan, K.; Liao, J.-D.; Yao, C.-K.; Peng,
ide. Anal. Chem. 2012, 84, 1772−1775. H.-C. Nanofabricated SERS-Active Substrates for Single-Molecule to
(322) Maesawa, C.; Inaba, T.; Sato, H.; Iijima, S.; Ishida, K.; Virus Detection In vitro: A Review. Biosens. Bioelectron. 2014, 61, 232−
Terashima, M.; Sato, R.; Suzuki, M.; Yashima, A.; Ogasawara, S.; et al. 240.
A Rapid Biosensor Chip Assay for Measuring of Telomerase Activity (342) Cheng, H. W.; Huan, S. Y.; Yu, R. Q. Nanoparticle-Based
Using Surface Plasmon Resonance. Nucleic Acids Res. 2003, 31, e4. Substrates for Surface-Enhanced Raman Scattering Detection of
(323) Wang, J.; Wu, L.; Ren, J.; Qu, X. Visualizing Human Bacterial Spores. Analyst 2012, 137, 3601−3608.
Telomerase Activity with Primer-Modified Au Nanoparticles. Small (343) Shanmukh, S.; Jones, L.; Driskell, J.; Zhao, Y.; Dluhy, R.;
2012, 8, 259−264. Tripp, R. A. Rapid and Sensitive Detection of Respiratory Virus

7959 DOI: 10.1021/acs.chemrev.7b00027


Chem. Rev. 2017, 117, 7910−7963
Chemical Reviews Review

Molecular Signatures Using a Silver Nanorod Array SERS Substrate. (360) Zhu, Y.; Kuang, H.; Xu, L.; Ma, W.; Peng, C.; Hua, Y.; Wang,
Nano Lett. 2006, 6, 2630−2636. L.; Xu, C. Gold Nanorod Sssembly Based Approach to Toxin
(344) Shao, F.; Lu, Z.; Liu, C.; Han, H.; Chen, K.; Li, W.; He, Q.; Detection by SERS. J. Mater. Chem. 2012, 22, 2387−2391.
Peng, H.; Chen, J. Hierarchical Nanogaps within Bioscaffold Arrays as (361) Chung, E.; Gao, R.; Ko, J.; Choi, N.; Lim, D. W.; Lee, E. K.;
A High-Performance SERS Substrate for Animal Virus Biosensing. Chang, S.-I.; Choo, J. Trace Analysis of Mercury(II) Ions Using
ACS Appl. Mater. Interfaces 2014, 6, 6281−6289. Aptamer-Modified Au/Ag Core-Shell Nanoparticles and SERS Spec-
(345) Liu, T.-Y.; Tsai, K.-T.; Wang, H.-H.; Chen, Y.; Chen, Y.-H.; troscopy in a Microdroplet Channel. Lab Chip 2013, 13, 260−266.
Chao, Y.-C.; Chang, H.-H.; Lin, C.-H.; Wang, J.-K.; Wang, Y.-L. (362) Ma, W.; Sun, M.; Xu, L.; Wang, L.; Kuang, H.; Xu, C. A SERS
Functionalized Arrays of Raman-Enhancing Nanoparticles for Capture Active Gold Nanostar Dimer for Mercury Ion Detection. Chem.
and Culture-Free Analysis of Bacteria in Human Blood. Nat. Commun. Commun. 2013, 49, 4989−4991.
2011, 2, 538. (363) Tang, W.; Chase, D. B.; Sparks, D. L.; Rabolt, J. F. Selective
(346) Wang, J.; Wu, X.; Wang, C.; Rong, Z.; Ding, H.; Li, H.; Li, S.; and Quantitative Detection of Trace Amounts of Mercury(II) Ion
Shao, N.; Dong, P.; Xiao, R.; et al. Facile Synthesis of Au-Coated (Hg2+) and Copper(II) Ion (Cu2+) Using Surface-Enhanced Raman
Magnetic Nanoparticles and Their Application in Bacteria Detection Scattering (SERS). Appl. Spectrosc. 2015, 69, 843−849.
via a SERS Method. ACS Appl. Mater. Interfaces 2016, 8, 19958− (364) Wu, X.; Tang, L.; Ma, W.; Xu, L.; Liu, L.; Kuang, H.; Xu, C.
19967. SERS-Active Au NR Oligomer Sensor for Ultrasensitive Detection of
(347) Guven, B.; Basaran-Akgul, N.; Temur, E.; Tamer, U.; Boyaci, I. Mercury Ions. RSC Adv. 2015, 5, 81802−81807.
H. SERS-Based Sandwich Immunoassay Using Antibody Coated (365) Wang, Y.; Chen, S.; Wei, C.; Xu, M.; Yao, J.; Li, Y.; Deng, A.;
Magnetic Nanoparticles for Escherichia coli Enumeration. Analyst Gu, R. A Femtogram Level Competitive Immunoassay of Mercury(II)
2011, 136, 740−748. Based on Surface-Enhanced Raman Spectroscopy. Chem. Commun.
(348) Yakes, B. J.; Lipert, R. J.; Bannantine, J. P.; Porter, M. D. 2014, 50, 9112−9114.
Detection of Mycobacterium Avium Subsp Paratuberculosis by a (366) She, P.; Chu, Y.; Liu, C.; Guo, X.; Zhao, K.; Li, J.; Du, H.;
Sonicate Immunoassay Based on Surface-Enhanced Raman Scattering. Zhang, X.; Wang, H.; Deng, A. A Competitive Immunoassay for
Clin. Vaccine Immunol. 2008, 15, 227−234. Ultrasensitive Detection of Hg2+ in Water, Human Serum and Urine
(349) Temur, E.; Boyaci, I. H.; Tamer, U.; Unsal, H.; Aydogan, N. A Samples Using Immunochromatographic Test Based on Surface-
Highly Sensitive Detection Platform Based on Surface-Enhanced Enhanced Raman Scattering. Anal. Chim. Acta 2016, 906, 139−147.
Raman Scattering for Escherichia coli Enumeration. Anal. Bioanal. (367) Sun, Z.; Du, J.; Jing, C. Recent Progress in Detection of
Chem. 2010, 397, 1595−1604. Mercury Using Surface Enhanced Raman Spectroscopy - A Review. J.
(350) Driskell, J. D.; Kwarta, K. M.; Lipert, R. J.; Porter, M. D.; Neill, Environ. Sci. 2016, 39, 134−143.
J. D.; Ridpath, J. F. Low-Level Detection of Viral Pathogens by a (368) Sun, B.; Jiang, X.; Wang, H.; Song, B.; Zhu, Y.; Wang, H.; Su,
Surface-Enhanced Raman Scattering Based Immunoassay. Anal. Chem. Y.; He, Y. Surface-Enhancement Raman Scattering Sensing Strategy
2005, 77, 6147−6154. for Discriminating Trace Mercuric Ion (II) from Real Water Samples
(351) Xu, S. P.; Ji, X. H.; Xu, W. Q.; Li, X. L.; Wang, L. Y.; Bai, Y. B.; in Sensitive, Specific, Recyclable, and Reproducible Manners. Anal.
Zhao, B.; Ozaki, Y. Immunoassay Using Probe-Labelling Immunogold Chem. 2015, 87, 1250−1256.
Nanoparticles with Silver Staining Enhancement via Surface-Enhanced (369) Yang, K.; Hu, Y. J.; Dong, N. A Novel Biosensor Based on
Raman Scattering. Analyst 2004, 129, 63−68. Competitive SERS Immunoassay and Magnetic Separation for
(352) Sun, Y.; Xu, L.; Zhang, F.; Song, Z.; Hu, Y.; Ji, Y.; Shen, J.; Li, Accurate and Sensitive Detection of Chloramphenicol. Biosens.
B.; Lu, H.; Yang, H. A Promising Magnetic SERS Immunosensor for Bioelectron. 2016, 80, 373−377.
Sensitive Detection of Avian Influenza Virus. Biosens. Bioelectron. 2017, (370) Ding, X.; Kong, L.; Wang, J.; Fang, F.; Li, D.; Liu, J. Highly
89, 906−912. Sensitive SERS Detection of Hg2+ Ions in Aqueous Media Using Gold
(353) Lin, H. Y.; Huang, C. H.; Hsieh, W. H.; Liu, L. H.; Lin, Y. C.; Nanoparticles/Graphene Heterojunctions. ACS Appl. Mater. Interfaces
Chu, C. C.; Wang, S. T.; Kuo, I. T.; Chau, L. K.; Yang, C. Y. On-line 2013, 5, 7072−7078.
SERS Detection of Single Bacterium Using Novel SERS Nanoprobes (371) Liu, M.; Wang, Z.; Pan, L.; Cui, Y.; Liu, Y. A SERS/
and A Microfluidic Dielectrophoresis Device. Small 2014, 10, 4700− Fluorescence Dual-Mode Nanosensor Based on the Human Telomeric
4710. G-quadruplex DNA: Application to Mercury (II) Detection. Biosens.
(354) Zhang, H.; Harpster, M. H.; Wilson, W. C.; Johnson, P. A. Bioelectron. 2015, 69, 142−147.
Surface-Enhanced Raman Scattering Detection of DNAs Derived from (372) Liu, M.; Wang, Z.; Zong, S.; Chen, H.; Zhu, D.; Wu, L.; Hu,
Virus Genomes Using Au-Coated Paramagnetic Nanoparticles. G.; Cui, Y. SERS Detection and Removal of Mercury(II)/Silver(I)
Langmuir 2012, 28, 4030−4037. using Oligonucleotide-Functionalized Core/Shell Magnetic Silica
(355) Zhan, L.; Zhen, S. J.; Wan, X. Y.; Gao, P. F.; Huang, C. Z. A Sphere@Au Nanoparticles. ACS Appl. Mater. Interfaces 2014, 6,
Sensitive Surface-Enhanced Raman Scattering Enzyme-Catalyzed 7371−7379.
Immunoassay of Respiratory Syncytial Virus. Talanta 2016, 148, (373) Ye, L. L.; Wen, G. Q.; Ouyang, H. X.; Liu, Q. Y.; Liang, A. H.;
308−312. Jiang, Z. L. A Novel and Highly Sensitive Nanocatalytic Surface
(356) Temur, E.; Zengin, A.; Boyaci, I. H.; Dudak, F. C.; Torul, H.; Plasmon Resonance-Scattering Analytical Platform for Detection of
Tamer, U. Attomole Sensitivity of Staphylococcal Enterotoxin B Trace Pb Ions. Sci. Rep. 2016, 6, 24150.
Detection Using an Aptamer-Modified Surface-Enhanced Raman (374) Wang, Y.; Irudayaraj, J. A SERS DNAzyme Biosensor for Lead
Scattering Probe. Anal. Chem. 2012, 84, 10600−10606. Ion Detection. Chem. Commun. 2011, 47, 4394−4396.
(357) Ko, J.; Lee, C.; Choo, J. Highly Sensitive SERS-Based (375) Tan, E.; Yin, P.; Lang, X.; Wang, X.; You, T.; Guo, L.
Immunoassay of Aflatoxin B1 Using Silica-Encapsulated Hollow Gold Functionalized Gold Nanoparticles as Nanosensor for Sensitive and
Nanoparticles. J. Hazard. Mater. 2015, 285, 11−17. Selective Detection of Silver Ions and Silver Nanoparticles by Surface-
(358) Zengin, A.; Tamer, U.; Caykara, T. Fabrication of a SERS Enhanced Raman Scattering. Analyst 2012, 137, 3925−3928.
Based Aptasensor for Detection of Ricin B Toxin. J. Mater. Chem. B (376) Yin, J.; Wu, T.; Song, J.; Zhang, Q.; Liu, S.; Xu, R.; Duan, H.
2015, 3, 306−315. SERS-Active Nanoparticles for Sensitive and Selective Detection of
(359) Liu, J.; Hu, Y.; Zhu, G.; Zhou, X.; Jia, L.; Zhang, T. Highly Cadmium Ion (Cd2+). Chem. Mater. 2011, 23, 4756−4764.
Sensitive Detection of Zearalenone in Feed Samples Using (377) Jin, L. L.; She, G. W.; Mu, L. X.; Shi, W. S. Highly Uniform
Competitive Surface-Enhanced Raman Scattering Immunoassay. J. Indicator-Mediated SERS Sensor Platform for the Detection of Zn2+.
Agric. Food Chem. 2014, 62, 8325−8332. RSC Adv. 2016, 6, 16555−16560.

7960 DOI: 10.1021/acs.chemrev.7b00027


Chem. Rev. 2017, 117, 7910−7963
Chemical Reviews Review

(378) Han, B.; Choi, N.; Kim, K. H.; Lim, D. W.; Choo, J. Connections Related to Cell-Cell Crosstalk Specifically Recognized by
Application of Silver-Coated Magnetic Microspheres to a SERS-Based an Aptamer. Angew. Chem., Int. Ed. 2016, 55, 3914−3918.
Optofluidic Sensor. J. Phys. Chem. C 2011, 115, 6290−6296. (396) Wang, X.; Qian, X. M.; Beitler, J. J.; Chen, Z. G.; Khuri, F. R.;
(379) Lu, G.; Li, H.; Liusman, C.; Yin, Z.; Wu, S.; Zhang, H. Surface Lewis, M. M.; Shin, H. J. C.; Nie, S. M.; Shin, D. M. Detection of
Enhanced Raman Scattering of Ag or Au Nanoparticle-Decorated Circulating Tumor Cells in Human Peripheral Blood Using Surface-
Reduced Graphene Oxide for Detection of Aromatic Molecules. Chem. Enhanced Raman Scattering Nanoparticles. Cancer Res. 2011, 71,
Sci. 2011, 2, 1817−1821. 1526−1532.
(380) Lee, C. H.; Hankus, M. E.; Tian, L.; Pellegrino, P. M.; (397) Bhana, S.; Chaffin, E.; Wang, Y.; Mishra, S. R.; Huang, X.
Singamaneni, S. Highly Sensitive Surface Enhanced Raman Scattering Capture and Detection of Cancer Cells in Whole Blood with
Substrates Based on Filter Paper Loaded with Plasmonic Nanostruc- Magnetic-Optical Nanoovals. Nanomedicine 2014, 9, 593−606.
tures. Anal. Chem. 2011, 83, 8953−8958. (398) Syme, C. D.; Sirimuthu, N. M. S.; Faley, S. L.; Cooper, J. M.
(381) Zhang, L.; Lang, X. Y.; Hirata, A.; Chen, M. W. Wrinkled SERS Mapping of Nanoparticle Labels in Single Cells Using a
Nanoporous Gold Films with Ultrahigh Surface-Enhanced Raman Microfluidic Chip. Chem. Commun. 2010, 46, 7921−7923.
Scattering Enhancement. ACS Nano 2011, 5, 4407−4413. (399) Hoonejani, M. R.; Pallaoro, A.; Braun, G. B.; Moskovits, M.;
(382) Galarreta, B. C.; Tabatabaei, M.; Guieu, V.; Peyrin, E.; Meinhart, C. D. Quantitative Multiplexed Simulated-Cell Identification
Lagugne-Labarthet, F. Microfluidic Channel with Embedded SERS 2D by SERS in Microfluidic Devices. Nanoscale 2015, 7, 16834−16840.
Platform for the Aptamer Detection of Ochratoxin A. Anal. Bioanal. (400) Freitag, I.; Beleites, C.; Dochow, S.; Clement, J. H.; Krafft, C.;
Chem. 2013, 405, 1613−1621. Popp, J. Recognition of Tumor Cells by Immuno-SERS-Markers in a
(383) Zengin, A.; Tamer, U.; Caykara, T. Extremely Sensitive Microfluidic Chip at Continuous Flow. Analyst 2016, 141, 5986−5989.
Sandwich Assay of Kanamycin Using Surface-Enhanced Raman (401) Pallaoro, A.; Hoonejani, M. R.; Braun, G. B.; Meinhart, C. D.;
Scattering of 2-Mercaptobenzothiazole Labeled Gold@Silver Nano- Moskovits, M. Rapid Identification by Surface-Enhanced Raman
particles. Anal. Chim. Acta 2014, 817, 33−41. Spectroscopy of Cancer Cells at Low Concentrations Flowing in A
(384) Dong, N.; Hu, Y.; Yang, K.; Liu, J. Development of Aptamer- Microfluidic Channel. ACS Nano 2015, 9, 4328−4336.
Modified SERS Nanosensor and Oligonucleotide Chip to Quantita- (402) Dinish, U. S.; Song, Z.; Ho, C. J. H.; Balasundaram, G.; Attia,
tively Detect Melamine in Milk with High Sensitivity. Sens. Actuators, B A. B. E.; Lu, X.; Tang, B. Z.; Liu, B.; Olivo, M. Single Molecule with
2016, 228, 85−93. Dual Function on Nanogold: Biofunctionalized Construct for In Vivo
(385) Indrasekara, A. S. D. S.; Paladini, B. J.; Naczynski, D. J.; Photoacoustic Imaging and SERS Biosensing. Adv. Funct. Mater. 2015,
Starovoytov, V.; Moghe, P. V.; Fabris, L. Dimeric Gold Nanoparticle 25, 2316−2325.
Assemblies as Tags for SERS-Based Cancer Detection. Adv. Healthcare (403) Maiti, K. K.; Dinish, U. S.; Samanta, A.; Vendrell, M.; Soh, K.
Mater. 2013, 2, 1370−1376. S.; Park, S. J.; Olivo, M.; Chang, Y.-T. Multiplex Targeted In vivo
(386) Zhang, Q.; Lu, X.; Tang, P.; Zhang, D.; Tian, J.; Zhong, L. Cancer Detection Using Sensitive Near-Infrared SERS Nanotags.
Gold Nanoparticle (AuNP)-Based Surface-Enhanced Raman Scatter- Nano Today 2012, 7, 85−93.
ing (SERS) Probe of Leukemic Lymphocytes. Plasmonics 2016, 11, (404) Qian, X.; Peng, X.-H.; Ansari, D. O.; Yin-Goen, Q.; Chen, G.
1361−1368. Z.; Shin, D. M.; Yang, L.; Young, A. N.; Wang, M. D.; Nie, S. In vivo
(387) Yue, J.; Liu, Z.; Cai, X. Y.; Ding, X. T.; Chen, S. H.; Tao, K.; Tumor Targeting and Spectroscopic Detection with Surface-Enhanced
Zhao, T. B. Bull Serum Albumin Coated Au@Ag Nanorods as SERS Raman Nanoparticle Tags. Nat. Biotechnol. 2008, 26, 83−90.
Probes for Ultrasensitive Osteosarcoma Cell Detection. Talanta 2016, (405) Wang, Y. W.; Khan, A.; Som, M.; Leigh, S. Y.; Wang, D.; Chen,
150, 503−509. Y.; McVeigh, P.; Wilson, B. C.; Liu, J. T. C. In Translational
(388) Lu, W.; Singh, A. K.; Khan, S. A.; Senapati, D.; Yu, H.; Ray, P. Biophotonics 2014; SPIE: Bellingham, WA, 2014.
C. Gold Nano-Popcorn-Based Targeted Diagnosis, Nanotherapy (406) Samanta, A.; Maiti, K. K.; Soh, K. S.; Liao, X.; Vendrell, M.;
Treatment, and In situ Monitoring of Photothermal Therapy Response Dinish, U. S.; Yun, S. W.; Bhuvaneswari, R.; Kim, H.; Rautela, S.; et al.
of Prostate Cancer Cells Using Surface-Enhanced Raman Spectrosco- Ultrasensitive Near-Infrared Raman Reporters for SERS-Based In vivo
py. J. Am. Chem. Soc. 2010, 132, 18103−18114. Cancer Detection. Angew. Chem., Int. Ed. 2011, 50, 6089−6092.
(389) Sha, M. Y.; Xu, H.; Natan, M. J.; Cromer, R. Surface-Enhanced (407) Byrne, J. D.; Betancourt, T.; Brannon-Peppas, L. Active
Raman Scattering Tags for Rapid and Homogeneous Detection of Targeting Schemes for Nanoparticle Systems in Cancer Therapeutics.
Circulating Tumor Cells in the Presence of Human Whole Blood. J. Adv. Drug Delivery Rev. 2008, 60, 1615−1626.
Am. Chem. Soc. 2008, 130, 17214−17215. (408) Danhier, F.; Feron, O.; Preat, V. To Exploit the Tumor
(390) Song, J.; Zhou, J.; Duan, H. Self-Assembled Plasmonic Vesicles Microenvironment: Passive and Active Tumor Targeting of Nano-
of SERS-Encoded Amphiphilic Gold Nanoparticles for Cancer Cell carriers for Anti-Cancer Drug Delivery. J. Controlled Release 2010, 148,
Targeting and Traceable Intracellular Drug Delivery. J. Am. Chem. Soc. 135−146.
2012, 134, 13458−13469. (409) Wang, M.; Thanou, M. Targeting Nanoparticles to Cancer.
(391) Park, H.; Lee, S.; Chen, L.; Lee, E. K.; Shin, S. Y.; Lee, Y. H.; Pharmacol. Res. 2010, 62, 90−99.
Son, S. W.; Oh, C. H.; Song, J. M.; Kang, S. H.; et al. SERS Imaging of (410) Dinish, U. S.; Balasundaram, G.; Chang, Y. T.; Olivo, M.
HER2-Overexpressed MCF7 Cells Using Antibody-Conjugated Gold Actively Targeted In vivo Multiplex Detection of Intrinsic Cancer
Nanorods. Phys. Chem. Chem. Phys. 2009, 11, 7444−7449. Biomarkers Using Biocompatible SERS Nanotags. Sci. Rep. 2015, 4,
(392) Li, M.; Banerjee, S. R.; Zheng, C.; Pomper, M. G.; Barman, I. 4075.
Ultrahigh Affinity Raman Probe for Targeted Live Cell Imaging of (411) Wang, Y. W.; Kang, S.; Khan, A.; Bao, P. Q.; Liu, J. T. C. In
Prostate Cancer. Chem. Sci. 2016, 7, 6779−6785. vivo Multiplexed Molecular Imaging of Esophageal Cancer via Spectral
(393) Ramya, A. N.; Samanta, A.; Nisha, N.; Chang, Y.-T.; Maiti, K. Endoscopy of Topically Applied SERS Nanoparticles. Biomed. Biomed.
K. New Insight of Squaraine-Based Biocompatible Surface-Enhanced Opt. Express 2015, 6, 3714−3723.
Raman Scattering Nanotag for Cancer-Cell Imaging. Nanomedicine (412) Wang, Y.; Kang, S.; Doerksen, J. D.; Glaser, A. K.; Liu, J. T. C.
2015, 10, 561−571. Surgical Guidance via Multiplexed Molecular Imaging of Fresh Tissues
(394) Kim, J. H.; Kim, J. S.; Choi, H.; Lee, S. M.; Jun, B. H.; Yu, K. Labeled With SERS-Coded Nanoparticles. IEEE J. Sel. Top. Quantum
N.; Kuk, E.; Kim, Y. K.; Jeong, D. H.; Cho, M. H.; et al. Nanoparticle Electron. 2016, 22, 6802911.
Probes with Surface Enhanced Raman Spectroscopic Tags for Cellular (413) Karabeber, H.; Huang, R. M.; Iacono, P.; Samii, J. M.; Pitter,
Cancer Targeting. Anal. Chem. 2006, 78, 6967−6973. K.; Holland, E. C.; Kircher, M. F. Guiding Brain Tumor Resection
(395) Zhang, N.; Bing, T.; Shen, L. Y.; Song, R. S.; Wang, L. L.; Liu, Using Surface-Enhanced Raman Scattering Nanoparticles and a Hand-
X. J.; Liu, M. R.; Li, J.; Tan, W. H.; Shangguan, D. H. Intercellular Held Raman Scanner. ACS Nano 2014, 8, 9755−9766.

7961 DOI: 10.1021/acs.chemrev.7b00027


Chem. Rev. 2017, 117, 7910−7963
Chemical Reviews Review

(414) Jokerst, J. V.; Cole, A. J.; Van de Sompel, D.; Gambhir, S. S. (431) Chen, Y.; Ren, J. Q.; Zhang, X. G.; Wu, D. Y.; Shen, A. G.; Hu,
Gold Nanorods for Ovarian Cancer Detection with Photoacoustic J. M. Alkyne-Modulated Surface-Enhanced Raman Scattering-Palette
Imaging and Resection Guidance via Raman Imaging in Living Mice. for Optical Interference-Free and Multiplex Cellular Imaging. Anal.
ACS Nano 2012, 6, 10366−10377. Chem. 2016, 88, 6115−6119.
(415) Cedervall, T.; Lynch, I.; Lindman, S.; Berggard, T.; Thulin, E.; (432) Kang, T.; Yoo, S. M.; Kang, M.; Lee, H.; Kim, H.; Lee, S. Y.;
Nilsson, H.; Dawson, K. A.; Linse, S. Understanding the Nanoparticle- Kim, B. Single-Step Multiplex Detection of Toxic Metal Ions by Au
Protein Corona Using Methods to Quantify Exchange Rates and Nanowires-on-Chip Sensor Using Reporter Elimination. Lab Chip
Affinities of Proteins for Nanoparticles. Proc. Natl. Acad. Sci. U. S. A. 2012, 12, 3077−3081.
2007, 104, 2050−2055. (433) Li, S.; Xu, L.; Ma, W.; Kuang, H.; Wang, L.; Xu, C. Triple
(416) Monopoli, M. P.; Walczyk, D.; Campbell, A.; Elia, G.; Lynch, Raman Label-Encoded Gold Nanoparticle Trimers for Simultaneous
I.; Baldelli Bombelli, F.; Dawson, K. A. Physical-Chemical Aspects of Heavy Metal Ion Detection. Small 2015, 11, 3435−3439.
Protein Corona: Relevance to In vitro and In vivo Biological Impacts of (434) Wu, L.; Wang, Z. Y.; Zong, S. F.; Chen, H.; Wang, C. L.; Xu, S.
Nanoparticles. J. Am. Chem. Soc. 2011, 133, 2525−2534. H.; Cui, Y. P. Simultaneous Evaluation of p53 and p21 Expression
(417) Gref, R.; Luck, M.; Quellec, P.; Marchand, M.; Dellacherie, E.; Level for Early Cancer Diagnosis Using SERS Technique. Analyst
Harnisch, S.; Blunk, T.; Muller, R. H. ’Stealth’ Corona-Core 2013, 138, 3450−3456.
Nanoparticles Surface Modified by Polyethylene Glycol (PEG): (435) He, J. C.; Li, G. K.; Hu, Y. L. Aptamer Recognition Induced
Influences of the Corona (PEG Chain Length and Surface Density) Target-Bridged Strategy for Proteins Detection Based on Magnetic
and of the Core Composition on Phagocytic Uptake and Plasma Chitosan and Silver/Chitosan Nanoparticles Using Surface-Enhanced
Protein Adsorption. Colloids Surf., B 2000, 18, 301−313. Raman Spectroscopy. Anal. Chem. 2015, 87, 11039−11047.
(418) Zavaleta, C. L.; Smith, B. R.; Walton, I.; Doering, W.; Davis, (436) Wang, Y. L.; Ravindranath, S.; Irudayaraj, J. Separation and
G.; Shojaei, B.; Natan, M. J.; Gambhir, S. S. Multiplexed Imaging of Detection of Multiple Pathogens in a Food Matrix by Magnetic SERS
Surface Enhanced Raman Scattering Nanotags in Living Mice Using Nanoprobes. Anal. Bioanal. Chem. 2011, 399, 1271−1278.
Noninvasive Raman Spectroscopy. Proc. Natl. Acad. Sci. U. S. A. 2009, (437) Wang, Y.; Rauf, S.; Grewal, Y. S.; Spadafora, L. J.; Shiddiky, M.
106, 13511−13516. J. A.; Cangelosi, G. A.; Schlucker, S.; Trau, M. Duplex Microfluidic
(419) von Maltzahn, G.; Centrone, A.; Park, J.-H.; Ramanathan, R.; SERS Detection of Pathogen Antigens with Nanoyeast Single-Chain
Sailor, M. J.; Hatton, T. A.; Bhatia, S. N. SERS-Coded Gold Nanorods Variable Fragments. Anal. Chem. 2014, 86, 9930−9938.
as a Multifunctional Platform for Densely Multiplexed Near-Infrared (438) Yang, X.; Gu, C.; Qian, F.; Li, Y.; Zhang, J. Z. Highly Sensitive
Imaging and Photothermal Heating. Adv. Mater. 2009, 21, 3175−3180. Detection of Proteins and Bacteria in Aqueous Solution Using Surface-
(420) Graham, D.; Mallinder, B. J.; Whitcombe, D.; Watson, N. D.; Enhanced Raman Scattering and Optical Fibers. Anal. Chem. 2011, 83,
Smith, W. E. Simple Multiplex Genotyping by Surface-Enhanced
5888−5894.
Resonance Raman Scattering. Anal. Chem. 2002, 74, 1069−1074. (439) Song, C. Y.; Yang, Y. J.; Yang, B. Y.; Min, L. H.; Wang, L. H.
(421) Ando, J.; Fujita, K.; Smith, N. I.; Kawata, S. Dynamic SERS
Combination Assay of Lung Cancer Associated Serum Markers Using
Imaging of Cellular Transport Pathways with Endocytosed Gold
Surface-Enhanced Raman Spectroscopy. J. Mater. Chem. B 2016, 4,
Nanoparticles. Nano Lett. 2011, 11, 5344−5348.
1811−1817.
(422) He, S.; Liu, K.-K.; Su, S.; Yan, J.; Mao, X.; Wang, D.; He, Y.; Li,
(440) Zhang, H.; Ma, X. Y.; Liu, Y.; Duan, N.; Wu, S. J.; Wang, Z. P.;
L.-J.; Song, S.; Fan, C. Graphene-Based High-Efficiency Surface-
Xu, B. C. Gold Nanoparticles Enhanced SERS Aptasensor for the
Enhanced Raman Scattering-Active Platform for Sensitive and
Simultaneous Detection of Salmonella typhimurium and Staph-
Multiplex DNA Detection. Anal. Chem. 2012, 84, 4622−4627.
(423) Nima, Z. A.; Mahmood, M.; Xu, Y.; Mustafa, T.; Watanabe, F.; ylococcus aureus. Biosens. Bioelectron. 2015, 74, 872−877.
Nedosekin, D. A.; Juratli, M. A.; Fahmi, T.; Galanzha, E. I.; Nolan, J. (441) Kim, H.; Seol, M.-L.; Lee, D.-I.; Lee, J.; Kang, I.-S.; Lee, H.;
P.; et al. Circulating Tumor Cell Identification by Functionalized Kang, T.; Choi, Y.-K.; Kim, B. Single Nanowire on Graphene (SNOG)
Silver-Gold Nanorods with Multicolor, Super-Enhanced SERS and as an Efficient, Reproducible, and Stable SERS-Active Platform.
Photothermal Resonances. Sci. Rep. 2015, 4, 4752. Nanoscale 2016, 8, 8878−8886.
(424) Mallia, R. J.; McVeigh, P. Z.; Fisher, C. J.; Veilleux, I.; Wilson, (442) Niu, C. Y.; Zou, B. F.; Wang, Y. Q.; Cheng, L.; Zheng, H. H.;
B. C. Wide-Field Multiplexed Imaging of EGFR-Targeted Cancers Zhou, S. M. Highly Sensitive and Reproducible SERS Performance
Using Topical Application of NIR SERS Nanoprobes. Nanomedicine from Uniform Film Assembled by Magnetic Noble Metal Composite
2015, 10, 89−101. Microspheres. Langmuir 2016, 32, 858−863.
(425) Wu, J.; Liang, D.; Jin, Q.; Liu, J.; Zheng, M.; Duan, X.; Tang, X. (443) Dai, Z. G.; Xiao, X. H.; Wu, W.; Liao, L.; Mei, F.; Yu, X. F.;
Bioorthogonal SERS Nanoprobes for Mulitplex Spectroscopic Guo, S. S.; Ying, J. J.; Ren, F.; Jiang, C. Z. Side-to-Side Alignment of
Detection, Tumor Cell Targeting, and Tissue Imaging. Chem. - Eur. Gold Nanorods with Polarization-Free Characteristic for Highly
J. 2015, 21, 12914−12918. Reproducible Surface Enhanced Raman Scattering. Appl. Phys. Lett.
(426) Jeong, S.; Kim, Y.-i.; Kang, H.; Kim, G.; Cha, M. G.; Chang, 2014, 105, 211902.
H.; Jung, K. O.; Kim, Y.-H.; Jun, B.-H.; Hwang, D. W.; et al. (444) Zhang, Z.; Fu, Y.; Yu, W.; Qin, X.; Xue, Z.; Liu, Y.; Luo, D.;
Fluorescence-Raman Dual Modal Endoscopic System for Multiplexed Yan, C.; Sun, X.; Wang, T. Dynamically Regulated Ag Nanowire
Molecular Diagnostics. Sci. Rep. 2015, 5, 9455. Arrays for Detecting Molecular Information of Substrate-Induced
(427) Wang, Y.; Tang, L.-J.; Jiang, J.-H. Surface-Enhanced Raman Stretched Cell Growth. Adv. Mater. 2016, 28, 9589−9595.
Spectroscopy-Based, Homogeneous, Multiplexed Immunoassay with (445) Zhu, C.; Meng, G.; Zheng, P.; Huang, Q.; Li, Z.; Hu, X.; Wang,
Antibody-Fragments-Decorated Gold Nanoparticles. Anal. Chem. X.; Huang, Z.; Li, F.; Wu, N. A Hierarchically Ordered Array of Silver-
2013, 85, 9213−9220. Nanorod Bundles for Surface-Enhanced Raman Scattering Detection
(428) Lau, H. Y.; Wang, Y.; Wee, E. J. H.; Botella, J. R.; Trau, M. of Phenolic Pollutants. Adv. Mater. 2016, 28, 4871−4876.
Field Demonstration of a Multiplexed Point-of-Care Diagnostic (446) Au, A. K.; Huynh, W.; Horowitz, L. F.; Folch, A. 3D-Printed
Platform for Plant Pathogens. Anal. Chem. 2016, 88, 8074−8081. Microfluidics. Angew. Chem., Int. Ed. 2016, 55, 3862−3881.
(429) Bodelon, G.; Montes-Garcia, V.; Fernandez-Lopez, C.; (447) Selimis, A.; Mironov, V.; Farsari, M. Direct Laser Writing:
Pastoriza-Santos, I.; Perez-Juste, J.; Liz-Marzan, L. M. Au@pNIPAM Principles and Materials for Scaffold 3D Printing. Microelectron. Eng.
SERRS Tags for Multiplex Immunophenotyping Cellular Receptors 2015, 132, 83−89.
and Imaging Tumor Cells. Small 2015, 11, 4149−4157. (448) Chia, H. N.; Wu, B. M. Recent Advances in 3D Printing of
(430) Guarrotxena, N.; Bazan, G. C. Antitags: SERS-Encoded Biomaterials. J. Biol. Eng. 2015, 9, 4.
Nanoparticle Assemblies that Enable Single-Spot Multiplex Protein (449) Ambrosi, A.; Pumera, M. 3D-Printing Technologies for
Detection. Adv. Mater. 2014, 26, 1941−1946. Electrochemical Applications. Chem. Soc. Rev. 2016, 45, 2740−2755.

7962 DOI: 10.1021/acs.chemrev.7b00027


Chem. Rev. 2017, 117, 7910−7963
Chemical Reviews Review

(450) Kadimisetty, K.; Mosa, I. M.; Malla, S.; Satterwhite-Warden, J. (467) Culka, A.; Jehlicka, J.; Edwards, H. G. M. Acquisition of Raman
E.; Kuhns, T. M.; Faria, R. C.; Lee, N. H.; Rusling, J. F. 3D-Printed Spectra of Amino Acids Using Portable Instruments: Outdoor
Supercapacitor-Powered Electrochemiluminescent Protein Immunoar- Measurements and Comparison. Spectrochim. Acta, Part A 2010, 77,
ray. Biosens. Bioelectron. 2016, 77, 188−193. 978−983.
(451) Zhang, H. Z.; Zhang, F. T.; Zhang, X. H.; Huang, D.; Zhou, Y. (468) Mohs, A. M.; Mancini, M. C.; Singhal, S.; Provenzale, J. M.;
L.; Li, Z. H.; Zhang, X. X. Portable, Easy-to-Operate, and Antifouling Leyland-Jones, B.; Wang, M. D.; Nie, S. Hand-held Spectroscopic
Microcapsule Array Chips Fabricated by 3D Ice Printing for Visual Device for In Vivo and Intraoperative Tumor Detection: Contrast
Target Detection. Anal. Chem. 2015, 87, 6397−6402. Enhancement, Detection Sensitivity, and Tissue Penetration. Anal.
(452) Tabakman, S. M.; Lau, L.; Robinson, J. T.; Price, J.; Sherlock, Chem. 2010, 82, 9058−9065.
S. P.; Wang, H.; Zhang, B.; Chen, Z.; Tangsombatvisit, S.; Jarrell, J. A.; (469) Zhang, D. S.-z.; Jiang, Y.; Yang, H.; Zhu, Y.; Zhang, S.; Zhu, Y.;
Utz, P. J.; Dai, H. Plasmonic Substrates for Multiplexed Protein Wei, D.; Lin, Y.; Wang, P.; Fu, Q.; et al. Dual-Encoded Microbeads
Microarrays with Femtomolar Sensitivity and Broad Dynamic Range. through a Host−Guest Structure: Enormous, Flexible, and Accurate
Nat. Commun. 2011, 2, 466. Barcodes for Multiplexed Assays. Adv. Funct. Mater. 2016, 26, 6146−
(453) Banholzer, M. J.; Millstone, J. E.; Qin, L. D.; Mirkin, C. A. 6157.
Rationally Designed Nanostructures for Surface-Enhanced Raman (470) Matschulat, A.; Drescher, D.; Kneipp, J. Surface-Enhanced
Spectroscopy. Chem. Soc. Rev. 2008, 37, 885−897. Raman Scattering Hybrid Nanoprobe Multiplexing and Imaging in
(454) Pieczonka, N. P. W.; Aroca, R. F. Single Molecule Analysis by Biological Systems. ACS Nano 2010, 4, 3259−3269.
Surfaced-Enhanced Raman Scattering. Chem. Soc. Rev. 2008, 37, 946− (471) Yan, B.; Li, B.; Wen, Z. N.; Luo, X. Y.; Xue, L. L.; Li, L. J.
954. Label-Free Blood Serum Detection by Using Surface-Enhanced Raman
(455) Thacker, V. V.; Herrmann, L. O.; Sigle, D. O.; Zhang, T.; Liedl, Spectroscopy and Support Vector Machine for the Preoperative
Diagnosis of Parotid Gland Tumors. BMC Cancer 2015, 15, 650.
T.; Baumberg, J. J.; Keyser, U. F. DNA Origami Based Assembly of
(472) Fu, C. C.; Xu, W. Q.; Chen, G.; Xu, S. P. ″Switch-off″
Gold Nanoparticle Dimers for Surface-Enhanced Raman Scattering.
Biosensing for Chymotrypsin-Catalyzed Reaction by SPR-SERS
Nat. Commun. 2014, 5, 3448.
Spectroscopy. Analyst 2013, 138, 6282−6286.
(456) Zheng, Y.; Thai, T.; Reineck, P.; Qiu, L.; Guo, Y.; Bach, U.
(473) Ma, Y. M.; Liu, H. L.; Qian, K.; Yang, L. B.; Liu, J. H. A
DNA-Directed Self-Assembly of Core-Satellite Plasmonic Nanostruc- Displacement Principle for Mercury Detection by Optical Waveguide
tures: A Highly Sensitive and Reproducible Near-IR SERS Sensor. Adv. and Surface Enhanced Raman Spectroscopy. J. Colloid Interface Sci.
Funct. Mater. 2013, 23, 1519−1526. 2012, 386, 451−455.
(457) Pilo-Pais, M.; Watson, A.; Demers, S.; LaBean, T. H.; (474) Ilkhani, H.; Hughes, T.; Li, J.; Zhong, C. J.; Hepel, M.
Finkelstein, G. Surface-Enhanced Raman Scattering Plasmonic Nanostructured SERS-Electrochemical Biosensors for Testing of
Enhancement Using DNA Origami-Based Complex Metallic Nano- Anticancer Drug Interactions with DNA. Biosens. Bioelectron. 2016,
structures. Nano Lett. 2014, 14, 2099−2104. 80, 257−264.
(458) Liu, R.; Liu, B.; Guan, G.; Jiang, C.; Zhang, Z. Multilayered (475) Lu, W.; Wang, Y.; Cao, X.; Li, L.; Dong, J.; Qian, W.
Shell SERS Nanotags with a Highly Uniform Single-Particle Raman Multiplexing Determination of Lung Cancer Biomarkers Using
Readout for Ultrasensitive Immunoassays. Chem. Commun. 2012, 48, Electrochemical and Surface-Enhanced Raman Spectroscopic Techni-
9421−9423. ques. New J. Chem. 2015, 39, 5420−5430.
(459) Chang, H.; Kang, H.; Yang, J. K.; Jo, A.; Lee, H. Y.; Lee, Y. S.; (476) Sreekanth, K. V.; Alapan, Y.; ElKabbash, M.; Ilker, E.;
Jeong, D. H. Ag Shell-Au Satellite Hetero-Nanostructure for Ultra- Hinczewski, M.; Gurkan, U. A.; De Luca, A.; Strangi, G. Extreme
Sensitive, Reproducible, and Homogeneous NIR SERS Activity. ACS Sensitivity Biosensing Platform Based on Hyperbolic Metamaterials.
Appl. Mater. Interfaces 2014, 6, 11859−11863. Nat. Mater. 2016, 15, 621−627.
(460) Driskell, J. D.; Uhlenkamp, J. M.; Lipert, R. J.; Porter, M. D.
Surface-Enhanced Raman Scattering Immunoassays Using A Rotated
Capture Substrate. Anal. Chem. 2007, 79, 4141−4148.
(461) Fan, R.; Vermesh, O.; Srivastava, A.; Yen, B. K. H.; Qin, L. D.;
Ahmad, H.; Kwong, G. A.; Liu, C. C.; Gould, J.; Hood, L.; et al.
Integrated Barcode Chips for Rapid, Multiplexed Analysis of Proteins
in Microliter Quantities of Blood. Nat. Biotechnol. 2008, 26, 1373−
1378.
(462) Chin, C. D.; Laksanasopin, T.; Cheung, Y. K.; Steinmiller, D.;
Linder, V.; Parsa, H.; Wang, J.; Moore, H.; Rouse, R.; Umviligihozo,
G.; et al. Microfluidics-Based Diagnostics of Infectious Diseases in the
Developing World. Nat. Med. 2011, 17, 1015−1019.
(463) Wu, X.; Chen, J.; Li, X.; Zhao, Y.; Zughaier, S. M. Culture-Free
Diagnostics of Pseudomonas Aeruginosa Infection by Silver Nanorod
Array Based SERS From Clinical Sputum Samples. Nanomedicine
2014, 10, 1863−1870.
(464) Andreou, C.; Hoonejani, M. R.; Barmi, M. R.; Moskovits, M.;
Meinhart, C. D. Rapid Detection of Drugs of Abuse in Saliva Using
Surface Enhanced Raman Spectroscopy and Microfluidics. ACS Nano
2013, 7, 7157−7164.
(465) Najafi, R.; Mukherjee, S.; Hudson, J.; Sharma, A.; Banerjee, P.
Development of a Rapid Capture-Cum-Detection Method for
Escherichia coli O157 From AppleJuice Comprising Nano-Immuno-
magnetic Separation in Tandem with Surface Enhanced Raman
Scattering. Int. J. Food Microbiol. 2014, 189, 89−97.
(466) Saha, A.; Jana, N. R. Paper-Based Microfluidic Approach for
Surface-Enhanced Raman Spectroscopy and Highly Reproducible
Detection of Proteins beyond Picomolar Concentration. ACS Appl.
Mater. Interfaces 2015, 7, 996−1003.

7963 DOI: 10.1021/acs.chemrev.7b00027


Chem. Rev. 2017, 117, 7910−7963

You might also like