You are on page 1of 29

Review

Cite This: Chem. Rev. 2019, 119, 293−321 pubs.acs.org/CR

Highly Sensitive and Multiplexed Protein Measurements


Limor Cohen†,‡,§ and David R. Walt*,†,‡

Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States

Wyss Institute for Biologically Inspired Engineering and §Department of Chemical Biology, Harvard University, Boston,
Massachusetts 02115, United States

ABSTRACT: Proteins are involved in many biological processes. Misfolded, truncated,


or mutated proteins as well as over- or underexpressed proteins have been implicated in
many diseases. Therefore, detection and quantification of proteins is extremely
See https://pubs.acs.org/sharingguidelines for options on how to legitimately share published articles.

important. Conventional techniques such as the enzyme-linked immunosorbent assay,


Western Blot, and mass spectrometry have enabled discovery and study of proteins in
biological samples. However, many important proteins are present at low
Downloaded via IOWA STATE UNIV on January 9, 2019 at 10:57:36 (UTC).

concentrations, rendering them undetectable using conventional techniques.


Furthermore, limited ability to simultaneously measure multiple proteins in a sample
has constrained our ability to fully study the proteome. In this review, we
comprehensively discuss approaches for protein detection. We first discuss the
fundamentals of proteins and protein assays, including affinity reagents, surface functionalization, assay formats, signal detection,
and multiplexing. We then discuss the challenges with these methods and review existing methods for highly sensitive and
multiplexed protein detection. Finally, we review recent advances in protein detection from the literature and discuss challenges
and future directions.

CONTENTS 6.1.1. Meso Scale Discovery (MSD) 305


6.1.2. Single-Molecule Arrays (Simoa) 306
1. Introduction 294 6.1.3. Single-Molecule Counting (SMC) 306
2. Proteins 294 6.1.4. Single-Cell Western Blot 307
3. Fundamentals of Protein Assays 295 6.2. Highly Multiplexed Methods for Protein
3.1. Affinity Reagents 295 Detection 307
3.1.1. Antibodies 295 6.2.1. SOMAscan Assay 307
3.1.2. Aptamers 296 6.2.2. Luminex 307
3.1.3. Other Affinity Reagents 296 6.2.3. Mass Cytometry (CyTOF) 309
3.2. Surface Functionalization 296 7. Advances from the Literature 309
3.3. Assay Formats 298 7.1. Advances in Labeling and Signal Detection 309
3.4. Signal Detection 298 7.1.1. Upconverting Nanoparticles 309
3.5. Multiplexing 299 7.1.2. Photoelectrochemical Detection (PEC) 309
4. Current Commonly Used Methods for Protein 7.1.3. Optical Ring Resonators 310
Detection 299 7.1.4. Surface-Enhanced Raman Scattering
4.1. Detection of Total Protein 299 (SERS) Tags 310
4.2. Enzyme-Linked Immunosorbent Assay 7.2. Miniaturization 310
(ELISA) 300 7.2.1. Microcantilever-Based Assays 310
4.3. Enzyme-Linked Immunospot (ELISPOT) 300 7.2.2. Ultrasmall Containers 311
4.4. Western Blot 301 8. Conclusion and Future Directions 313
4.5. Protein Microarrays 301 Author Information 313
4.6. Flow Cytometry 301 Corresponding Author 313
4.7. Proximity Ligation Assay (PLA) and other ORCID 313
Nucleic Acid-Based Detection Methods 302 Notes 313
4.8. Mass Spectrometry 302 Biographies 313
4.9. Lateral Flow Assay (LFA) 303 References 314
4.10. Surface Plasmon Resonance (SPR) 303
4.11. Optical Imaging 304
5. Challenges with Protein Detection 304
6. Emerging Methods for Ultrasensitive and Highly
Special Issue: Chemical Sensors
Multiplexed Measurements 305
6.1. Ultrasensitive Methods for Protein Detec- Received: April 20, 2018
tion 305 Published: August 28, 2018

© 2018 American Chemical Society 293 DOI: 10.1021/acs.chemrev.8b00257


Chem. Rev. 2019, 119, 293−321
Chemical Reviews Review

1. INTRODUCTION ubiquination, deamidation, oxidation, sulfation, and nitration.


The five major classes of covalent modifications are shown in
Proteins are macromolecules involved in important biological
Figure 1.4 In some cases, detection of a specific post-
functions including replicating genomic information, regulating
transcription, signaling, providing structure, catalyzing reac-
tions, and transporting molecules. Many proteins are
dysregulated and differentially expressed in disease and can
serve as drug targets or biomarkers. Therefore, tools for
measuring and detecting proteins are essential for fully
characterizing these functions.
Several challenges exist with protein detection methods.
First, a sample may contain many interfering molecules that
make it difficult to detect a specific protein. Second, many
proteins may be present at low concentrations in a biological
sample. Third, concentrations of different proteins can vary by
many orders of magnitude. Finally, protein processing, such as
post-translational modifications and spliced variants that result
in different isoforms, can make it difficult or complicated to
detect the specific protein of interest. Common techniques
currently employed for protein detection include the Western
blot, mass spectrometry, and the enzyme-linked immunosorb-
ent assay (ELISA). These techniques have provided a wealth of
information on proteins and their function; however, they may
suffer from shortcomings in throughput, multiplexing capa-
bilities, specificity, and sensitivity. In the past few years, many
tools have been developed to overcome these challenges.
In this review, we comprehensively discuss existing and
innovative analytical tools for protein detection and
quantification. First, we briefly discuss the fundamental
properties of proteins. We then discuss fundamentals of
protein assays, including affinity reagents, surface functional-
ization, assay formats, signal detection, and multiplexing. We
then review commonly used methods for protein detection. Figure 1. Five major types of covalent modifications to protein side
We then discuss the challenges with these methods and review chains: phosphorylation, acylation, alkylation, glycosylation, and
oxidation. Reprinted with permission from ref 4. Copyright 2005
existing methods for highly sensitive and multiplexed protein John Wiley and Sons.
detection. Finally, we review advances in protein detection
techniques from the literature and discuss challenges and translational modification on a protein may be desirable over
future directions. detection of the unmodified protein because it may be the
functional or disease-causing species. Some protein detection
2. PROTEINS methods first start by treating the protein with a glycosylase,
Proteins are a class of biological macromolecules that play which removes glycans. This treatment is designed to reduce
important roles in many biological processes. Proteins are the complexity of the sample but may remove important PTMs
essential for replicating genomic information, regulating gene that are key to understanding the sample being measured.
expression, transcribing mRNA, signaling, catalyzing metabolic These modifications play critical roles in the function of
reactions, and transporting molecules. Disrupted protein proteins and can be dysregulated in disease.5−9 For example,
expression as well as dysfunctional mutated or misfolded approximately 30% of proteins encoded by the human genome
proteins are associated with multiple diseases, such as are phosphorylated, with irregular phosphorylation patterns
Alzheimer’s disease and sickle cell anemia. Thus, detecting associated with many human diseases.10 Thus, it is not only
specific proteins is important. Several features of proteins are important to detect specific proteins but also to detect specific
important for their detection and are discussed in this section. modifications on certain proteins. Additionally, proteins range
First, the proteome consists of tens of thousands of proteins, in size from very small monomeric proteins to large
and thus, detecting a specific protein in a complex matrix is macromolecular complexes, in which proteins interact with
challenging. The Human Genome Project revealed that other molecules such as RNA, DNA, or other proteins.
different proteins can be encoded by the same genes. It has Therefore, it is often challenging to detect a specific protein or
been estimated that the human genome contains approx- a specific modification on a protein in a complex biological
imately 20 000 protein coding genes,1 yet the number of sample.
proteins is considerably higher as a result of variations in DNA Second, some proteins of interest may be present at low
sequence, alternative mRNA splicing, post-translational mod- levels, limiting the ability to detect them. It can be particularly
ifications, and proteolytic cleavage.2 A mammalian cell contains difficult to measure low levels of a given protein in a complex
a total of 109 protein molecules, and a bacterial cell contains a biological sample that contains high levels of other potentially
total of 106 protein molecules.3 Proteins are modified by interfering molecules. Third, it is often necessary to detect
covalent post-translational modifications including phosphor- more than one protein in a single sample. Many proteins work
ylation, acetylation, methylation, acylation, glycosylation, in networks to exert their function. Some proteins are
294 DOI: 10.1021/acs.chemrev.8b00257
Chem. Rev. 2019, 119, 293−321
Chemical Reviews Review

overexpressed, while others are under expressed. Protein 3.1.1. Antibodies. Antibodies, major components of the
measurements on the omics scale are highly advantageous vertebrate immune system, bind to foreign molecules known as
and provide unbiased detection of proteins. Measurements of antigens. Most antigens are proteins. An antibody binds
many proteins simultaneously is challenging because proteins specifically to a structure on the antigen called an epitope.
in a biological sample can differ in concentration by several Several classes of antibodies exist, with Immunoglobulin G
orders of magnitude.11 Additionally, levels of a target protein (IgG) being a major class and most commonly used in
can vary substantially between different biological samples, bioanalytical applications. IgGs have four polypeptide chains
which further complicates detection. Finally, the particular consisting of two heavy chains and two light chains that form a
structural form of a protein may depend on its native cellular 150 kDa molecular complex (Figure 2). The variable domains
context. To analyze proteins, it is often necessary to alter their
native environment or remove them from it completely. For
example, hydrophobic membrane proteins that are embedded
in the lipid bilayer may not be stable in aqueous environments.
Therefore, when removing a protein from its native environ-
ment for analysis, it is important to determine if such
manipulations will affect the analysis.

3. FUNDAMENTALS OF PROTEIN ASSAYS


Methods to analyze proteins can be broadly classified into
three categories. The first category is protein detection using
solution-based assays, which will be the focus of this review.
The second category is protein detection using mass
spectrometry-based methods. The third category is structural Figure 2. IgG structure showing two different molecular depictions
analysis of proteins using methods such as X-ray crystallog- on the left and right sides. IgGs consist of two heavy chains and two
raphy, NMR, Cryo-EM, and circular dichroism (CD). The light chains that are linked by a disulfide bond. IgG contains two
antigen-binding sites. Modified and reprinted with permission from
latter two categories have been thoroughly reviewed elsewhere ref 20. Copyright 2017 Annual Reviews.
and will not be the focus of this review.12,13
A protein assay typically consists of three major of the light and heavy chains associate to form the antigen-
components: an affinity reagent, a signal transducer, and a binding domain, also known as the Fab. In 1993, it was
detector.14 The role of the affinity reagent is to specifically bind
discovered that sharks16 and camelids, such as camels and
to the target protein molecule. This binding enables significant
purification of the protein from the complex sample being llamas,17 produce antibodies that do not have a light chain.
analyzed. The signal transducer then converts the binding These single-domain antibodies are known as Nanobodies or
event into a measurable signal, such as an optical, electro- VHHs (Figure 3).18 Single-domain antibodies have lower
chemical, or mechanical signal. The detector, such as a charge- molecular weights (90 kDa) than traditional antibodies (150
coupled device (CCD), then converts the signal into a digital kDa) and therefore may be better affinity reagents.19
readout. In this section we discuss affinity reagents, surface Antibodies are generally produced by affinity maturation in a
functionalization, assay formats, signal detection, and multi- living organism. Antibody diversity is attained by combinatorial
plexing. biosynthesis based on genetic recombination capable of
3.1. Affinity Reagents
An affinity reagent is a molecule that recognizes a specific
protein. Binding of the protein to the affinity reagent is
converted into a measurable signal. Affinity reagents may need
to be adsorbed or covalently attached to a surface. Approaches
for covalent conjugation of biomolecules have been thoroughly
described elsewehere.15 Additionally, affinity reagents need to
interact with the signal transducer either directly or via a
secondary molecule that can interact with the signal trans-
ducer. These interactions must not affect the performance of
the measurement by sterically hindering binding to the protein
or affecting the performance of the signal transducer.
There are several important characteristics of protein affinity
reagents including high affinity (low dissociation constants),
high specificity (ability to recognize the target protein in a
sample containing many other potentially interfering mole-
cules), cost, stability, and reproducibility. The most commonly Figure 3. Single-domain antibody structure. Single-domain antibodies
are much smaller compared to IgGs. Single-domain antibodies do not
used affinity reagents for proteins are antibodies and aptamers. have a light chain and are derived only from the heavy chain. Single-
Other affinity reagents such as molecularly imprinted domain antibodies are naturally found in camelids and sharks. (v,
polymers, lectins, peptides, and antibody mimetics can also variable; HCAb, heavy-chain-only antibodies; H, heavy chain).
be used. These affinity reagents will be discussed in this Modified and reprinted with permission from ref 21. Copyright
section. 2014 Nature Publishing Group.

295 DOI: 10.1021/acs.chemrev.8b00257


Chem. Rev. 2019, 119, 293−321
Chemical Reviews Review

producing more than 108 different antibodies that have ment. The SELEX process starts with a large combinatorial
different binding specificities.22 The most common antibody library of DNA or RNA oligonucleotides of random sequences.
preparations are polyclonal and monoclonal antibodies. These sequences are added to the target protein, usually
Polyclonal antibodies are generated in vivo via affinity attached to a solid support, and some bind with high affinity.
maturation. Therefore, polyclonal antibodies recognize differ- The sequences that do not bind to the target protein are then
ent epitopes of a given antigen. Monoclonal antibodies are separated from those that bind to the target protein. The
derived from a hybridoma, a fusion of a B cell with an immortal bound sequences are eluted from the protein, amplified, and
cell line, and were first described in 1975.23 Since monoclonal undergo another round of selection. This process is repeated,
antibodies are derived from a clone of identical B cells, they typically between 8 and 15 rounds, with each iteration
recognize a single epitope. subjecting the bound sequences to higher stringency to elicit
In some cases, it is preferable to use one clone over another; stronger binders. The targets are then cloned and sequenced.
for example, when detection of a specific post-translational Aptamers have several advantages for use as affinity reagents
modification is required, a monoclonal preparation is necessary such as low cost, high stability, and high specificity, with
since a polyclonal preparation will detect all of the epitopes on dissociation constants in the femtomolar to picomolar
the protein. On the other hand, in some cases a polyclonal range.36−38 Once an optimal sequence has been determined,
preparation is favorable−for instance, when detecting the they can be easily and reproducibly generated by chemical
presence of all proteins containing a particular epitope, synthesis. Additionally, DNA aptamers are highly stable
regardless of the PTM. A disadvantage of polyclonal antibodies reagents such that bound proteins can be removed either
is the potential lack of reproducibility between different chemically or thermally to regenerate the free aptamer for
reagent lots. Once a polyclonal preparation is depleted an another round of binding. Another advantage of aptamers is
animal must be immunized again to produce more polyclonal that they can be generated against almost any protein, even
antibodies. The second polyclonal batch will likely have a toxic proteins and those that are not immunogenic. Aptamers
different composition and may perform differently than the can be easily integrated into many protein assay formats by
first batch, requiring new assay conditions to be determined substituting for antibodies and are easily labeled and modified
and new calibrations to be performed. with various molecules and functional groups. Aptamers are
Several approaches for in vitro antibody production have highly stable and can also be regenerated for numerous uses.
been developed. These include phage, yeast,24 and ribosome The SOMAscan assay, which is described in section 6.1.1, uses
display,25,26 which have been reviewed elsewhere.24,27,28 These aptamers as affinity reagents.
approaches are advantageous for high-throughput antibody 3.1.3. Other Affinity Reagents. Other than antibodies
production as well as production of antibodies against toxic and aptamers, additional affinity reagents are available
molecules and molecules that do not elicit an immune including molecularly imprinted polymers (MIPs),39,40 protein
response in an animal. Another advantage is the ability to ligands, lectins, and antibody mimetics. The protein itself or
incorporate a tag, such as a his-tag, for antibody immobiliza- other peptide ligands can be used to detect protein−protein
tion and conjugation to various labels. Finally, these interactions. For example, when one needs to detect antibodies
recombinant preparations may reduce the cost of antibody to determine if an immune response has occurred, the protein
production. can be used to bind to the target antibodies. Lectins, such as
Antibody−antigen interactions are particularly strong and wheat germ agglutinin or concanavalin A,41 bind to glycans on
have low KD values. In vivo affinity maturation, with repeated glycoproteins and are commonly used. Alternative molecules
exposure of an animal to the same antigen, typically results in that mimic antibodies, known as antibody mimetics, are
antibodies with affinities in the nanomolar to picomolar proteins that are structurally unrelated to antibodies and can
range,29 but directed evolution of antibodies with affinities in bind to targets specifically through protein-engineering
the femtomolar range have been reported.30 Antibodies are approaches.20
generally highly specific to their target, although nonspecific
3.2. Surface Functionalization
binding can occur. Overall, due to their high affinity and
specificity, antibodies are key reagents for protein detection.31 In many protein assays, the target protein must first come into
Antibodies can be easily integrated into protein assays. contact with an affinity reagent that is immobilized on the
Antibodies have multiple reactive side chains that can serve as surface. Therefore, the performance of a protein assay is highly
attachment sites for conjugation to solid supports, such as dependent on the chemical modification of the surface with an
magnetic beads or nanoparticles, or labels that act as signal affinity reagent.42 Approaches to attach an affinity reagent onto
transducers. Antibodies are usually covalently attached to the surface include noncovalent and covalent immobilization.
surfaces or detection labels. Another approach is to utilize Strategies for covalent immobilization chemistries have been
biotinylated antibodies that can interact with streptavidin, thoroughly described.15 Noncovalent surface attachment can
which has been attached to a surface or a signal transducer. be achieved by direct electrostatic interactions between the
However, covalent conjugation may affect interaction with the surface and the affinity reagent. Alternatively, secondary
target protein. Modifying a molecule that can interact with the molecules such as biotin−streptavidin or attachment via
antibody may overcome this challenge. A secondary antibody, interaction of Proteins A and G to the Fc region of the
Protein A, or Protein G, which bind to the Fc region of the antibody can also be used. Self-assembled monolayers (SAMs)
antibody, may be used. are also commonly used to attach affinity reagents to a
3.1.2. Aptamers. Aptamers are single-stranded DNA or surface.43 Finally, encapsulation of the affinity reagent is
RNA oligonucleotides that can bind to proteins with high another approach. All of these strategies can be used to attach
affinity and specificity.32,33 First described in 1990,34,35 an affinity reagent to the surface (Figure 4).44
aptamers are created by an in vitro process known as There are two different implementations for using surface-
SELEX, systemic evolution of ligands by exponential enrich- bound affinity reagents. The first implementation is one in
296 DOI: 10.1021/acs.chemrev.8b00257
Chem. Rev. 2019, 119, 293−321
Chemical Reviews Review

which the target protein must diffuse to the surface, for


example, when using an affinity reagent immobilized onto the
surface of a traditional microtiter plate. The second
implementation is when the affinity reagent is immobilized
onto the surface of micro- or nanoparticles and the target
protein and the surface are both in solution. Solution-based
assays enhance and accelerate the binding kinetics and capture
efficiency of the target protein due to superior mass transport
of the protein to the surface. Another advantage is the higher
surface to volume ratio of micro- and nanoparticles, which
further improves the capture efficiency. Another consideration
is the orientation of the affinity reagent on the surface and the
surface density or number of affinity reagents per area. Both
have been shown to affect the binding efficacy of the target
protein.46,47 Recently, several studies have assessed the
relationship between capture efficiency and antibody function-
alization of gold nanoparticles.45,48−51 Due to their favorable
properties, nanomaterials are becoming more popular for use
in protein detection techniques. These nanomaterials include
carbon nanotubes, polymer nanowires, and zinc oxide
nanorods.52−55
Reduction of nonspecific binding to the surface is also
important. Methods for passivating the surface include using
BSA56 or various neutral or hydrophilic macromolecules such
as poly(hydroxyethyl methacrylate), poly(acrylamide), and
Figure 4. Five strategies used for bioconjugation of affinity reagents to poly(ethylene glycol).57−60
surfaces via (i) electrostatic interactions, (ii) direct interaction with Finally, another consideration is the ability to regenerate the
the surface, (iii) secondary interactions, (iv) covalent immobilization, binding capabilities of the surface for multiple independent
and (v) encapsulation. Reprinted with permission from ref 45.
Copyright 2013 American Chemical Society.
measurements of proteins.61 Regeneration is achieved by
overcoming the attractive forces between the affinity reagent
and the bound protein, ideally without destroying the binding
capabilities of the former. Strategies to regenerate the binding

Figure 5. Immunoassay formats. (a) Label-free immunoassay. Binding of the target protein to capture antibodies immobilized onto a surface results
in a measurable signal. (b) Sandwich immunoassay in which the detection antibody is labeled directly with a tag such as a fluorophore. (c)
Sandwich immunoassay in which an enzyme is used to amplify the signal. (d) Competitive immunoassay in which the target protein competes with
the labeled protein from binding to the capture reagent. (a−d) Heterogeneous immunoassays in which the capture reagent is immobilized onto a
surface and the unbound sample components are washed away. (e) Homogeneous immunoassay in which the reagents are in solution and are not
separated from other unbound sample components.

297 DOI: 10.1021/acs.chemrev.8b00257


Chem. Rev. 2019, 119, 293−321
Chemical Reviews Review

capabilities of surfaces have been developed;62 however, it is Optical detection is most commonly used for signal
still quite challenging to regenerate the surface because the transduction. Optical detection is a signal transduction method
performance is compromised due to reduced binding affinity, based on measuring changes in the amount of light absorbed
increased nonspecific binding, and thus lower sensitivity. Some or emitted at a specific wavelength, typically in the ultraviolet
affinity reagents, such as aptamers, may be more amenable to (UV) or infrared (IR) regions. The affinity reagent is labeled
regeneration than antibodies. The main benefits of surface with a molecule that can be optically detected. Since the ability
regeneration are reduced cost and the ability to monitor to detect the protein analyte depends on detecting the optical
changes in concentration over time without changing the label, the label needs to either have a high extinction coefficient
affinity capture surface. for absorbance measurements or have a high quantum yield for
3.3. Assay Formats fluorescence emission measurements. In addition, there should
be a sufficient degree of labeling on the affinity reagent.
The two most common formats for protein assays are Fluorescent labels are more commonly used in optical
noncompetitive and competitive assays. Noncompetitive assays detection. Two key properties of fluorophores are their Stokes
can be further broken down into label-free assays (Figure 5a) shift and quantum yield. A larger Stokes shift results in better
and sandwich assays (Figure 5a−c). Both label-free and signal separation from the excitation wavelength. The quantum
sandwich assays require a capture reagent that specifically yield, along with the molar extinction coefficient, determines
binds to the target protein. In a label-free assay, binding of the the overall brightness of the fluorophore.68 Numerous
target protein to the capture reagent results in a measurable fluorescent dyes and their derivatives have been developed to
change. In a sandwich assay, a second affinity reagent binds to increase the brightness and quantum yield.69 The most
a different epitope on the target protein, forming a “sandwich.” common fluorescent core structures are water-soluble con-
The second affinity reagent is labeled with a tag, such as a jugated organic molecules that are derivatives of cyanine,
fluorophore (Figure 5b), or an enzyme (Figure 5c) that fluorescein, rhodamine, and coumarin. Derivatives with
produces molecules that can be detected. In a competitive aromatic ring constituents, such as electron-donating or
assay (Figure 5d), the surface is coated with a capture reagent -withdrawing groups, can have drastic effects on fluorescence
that binds to the target protein. A sample containing the target and quantum yield. Fluorescent molecules other than small
protein and a known amount of labeled target protein are organic dyes can also be used for optical detection. These
added. The target protein competes with the labeled protein include naturally fluorescent protein molecules, such as
from binding to the capture reagent. When low levels of the phycoerythrin and green fluorescent protein (GFP). However,
target proteins are present relative to the labeled protein, the these molecules are larger than organic dyes and may sterically
labeled protein can bind to the capture reagents to produce a hinder the protein from binding to the affinity reagent.
signal. With increasing levels of the target protein, binding of There are several limitations to detecting a fluorophore that
the labeled protein to the capture reagent decreases. Therefore, has been directly conjugated to an affinity reagent. First, the
the concentration of the target protein is inversely proportional protein-binding site on the affinity reagent may be unavailable
to the signal. A competitive assay for protein detection is less for binding to the protein. Second, when multiple fluorophores
commonly used than the noncompetitive assay. However, it is are proximal to each other, quenching could occur. Finally, a
often used to detect antibodies or when only one affinity limited number of fluorophores can be attached to an affinity
reagent is available. reagent either because more labels will affect the binding
Protein assays can be homogeneous (Figure 5e) or affinity or because there are a limited number of sites where
heterogeneous (Figures 5a−d). In a homogeneous assay, the the label can react. Typically, only 8−10 fluorophores can be
reagents are in solution and do not require separation from conjugated to an antibody.
other unbound sample components, while in a heterogeneous In general, amplifying the signal leads to enhanced
assay, the capture reagent is immobilized onto a surface and sensitivity. Various approaches to signal amplification have
the unbound sample components are washed away. Hetero- been implemented. One approach is by using an enzyme that
geneous assays are more commonly used than homogeneous generates many detectable signal molecules. Enzymatic turn-
assays. over amplifies the signal and increases the sensitivity because
3.4. Signal Detection
the number of detectable molecules is substantially higher than
the number of protein molecules captured. Commonly used
Interaction of a protein with an affinity reagent must be enzymes include alkaline phosphatase (AP), horseradish
converted into a measurable signal. Signal transduction peroxidase (HRP), and β-galactosidase. These enzymes turn
approaches fall into two general categories: chemical and over many substrate molecules to generate many fluorescent
physical (or label-free) transduction. Chemical transduction is molecules. A second approach is to use nanomaterials.70−72
a change in chemical composition coupled to a binding event, Nanomaterials have increased surface area such that they can
for example, when an enzyme catalyzes the formation of a be conjugated to both an affinity reagent and many labels, such
fluorescent product. Physical transduction is a measurable as fluorophores or enzymes. Nanomaterials that are inherently
change in the physical properties in response to the interaction brighter, such as carbon dots73−75 or polymer dots,76−78 can
of the affinity reagent with the protein. Because physical also be used as optical labels. Fluorescent silica nanoparticles in
transduction methods do not require a chemical label, they are which standard organic dyes are incorporated can also be used
also known as label-free transduction. Several reviews have to enhance the signal.79−81
been written that provide more detail on optical label-free Fluorescent molecules are sensitive to changes in the
detection methods including theoretical discusions.63−67 chemical environment. Buffer composition, pH, and solvent
Chemical and label-free signal transduction approaches polarity can affect the quantum yield. Overlabeling an affinity
relevant to protein detection include optical, electrochemical, reagent with a fluorescent molecule can also change the
and mechanical signal transduction. quantum yield due to quenching resulting from interactions
298 DOI: 10.1021/acs.chemrev.8b00257
Chem. Rev. 2019, 119, 293−321
Chemical Reviews Review

between the dyes. Additionally, nonspecific binding can arise


between organic dyes containing aromatic groups and other
components in the assay such as surfaces.82 Such interactions
can be minimized by adding negative charges to the
fluorophore, such as PEG or sulfonate modifications, to
increase the hydrophilicity.15 Naturally occurring fluorophores
in a biological sample, such as aromatic amino acid residues,
NADH, and flavins, can also cause interference and increase
the background. Figure 6. Approaches for multiplexing via (a) a conventional
Other commonly used methods for optical detection, microarray in which the identity of the target protein is known
particularly as part of the enzyme-linked immunosorbent based on the predetermined position of the affinity reagent and (b) a
assay (ELISA), include electrochemiluminescence (ECL), suspension array in which the affinity reagents are attached to
encoded particles. Each optically distinct population of beads is
chemiluminescence, and colorimetric-based assays. In ECL, conjugated to capture antibodies to a specific protein. Printed with
an electron transfer reaction at the surface of an electrode permission from ref 108. Copyright 2006 John Wiley and Sons.
produces excited states that can then emit light. The MSD
assay uses ECL for protein detection and is discussed in
example, the SOMAscan assay, which is described in more
section 6.1.1.
detail in section 6.2.1, can detect approximately 1300 different
Chemiluminescence is a result of a chemical reaction that
proteins simultaneously using a nucleic acid microarray. The
produces light. In a chemiluminescent reaction, oxidation of an
second format employs encoded micro- or nanoparticles, in
organic dye, such as luminol, with a strong oxidant, such as which each uniquely encoded particle population is function-
hydrogen peroxide, in the presence of a catalyst produces a alized with an affinity reagent specific to a given protein. There
light-emitting molecule. The emission lifetime of the molecule are several ways to encode micro- and nanoparticles, including
can last from several seconds to hours. Commonly used optical encoding with fluorescent dyes or Raman tags.103−105
chemiluminescent reagents for protein detection are the Luminex has developed a 100-plex assay by optically encoding
enzyme horseradish peroxidase and luminol or its derivatives. microspheres with combinations of fluorescent dyes.106,107
Chemiluminescent detection does not require an excitation Other ways to encode are based on graphical, physical,
source, and therefore, there is no background signal due to magnetic, and thermal properties. These methods have been
excitation of other components in the sample matrix. A recently reviewed.108−110 Finally, encoding using nucleic acid
disadvantage is that the generated chemiluminescent signal can barcodes can provide high multiplexing capabilities.111
be relatively weak; therefore, approaches to enhance A major challenge with multiplexed protein detection assays
chemiluminescent intensity have been developed.83 These is nonspecific binding and cross-reactivity between affinity
approaches include modification of luminol or addition of reagents, secondary labels, and other components in the
various organic compounds, such as phenols, that enhance biological sample.112 Despite many efforts, measuring multiple
chemiluminescence. Quantum dots and metallic nanoparticles protein analytes with high sensitivity, specificity, and accuracy
have also been used to enhance chemiluminescence.84−86 For is still a major challenge.
example, it has been shown that gold nanoparticles can catalyze
chemiluminescence of luminol.87 Additionally, carbon dots 4. CURRENT COMMONLY USED METHODS FOR
have been shown to have chemiluminescent properties.88 PROTEIN DETECTION
Colorimetric signal generation is based on formation of a
This section discusses the most widely used techniques for
colored precipitate, typically by an enzyme.89 Nanoparticle- protein measurements, particularly for life sciences research
based nonenzymatic colorimetric detection has also been and clinical diagnostics. Many of these techniques were
demonstrated.90,91 Nanoparticle-based colorimetric assays can developed in the 1960s and 1970s and are still widely used
exhibit high sensitivities for protein markers,92−95 down to the today. These techniques include direct detection of total
attomolar range.96 Advantages of colorimetric assays include protein, the enzyme-linked immunosorbent assay (ELISA),
relative simplicity, low cost, and ability to detect markers with enzyme-linked immunospot (ELISPOT), Western blot,
the naked eye.97 protein microarrays, flow cytometry, proximity ligation assays
In addition to an optical label, optical detection tools often (PLAs) and other nucleic acid-based methods, mass
require components such as a light source, optical filters, and spectrometry, lateral flow assays, surface plasmon resonance
detectors, such as photodiodes, phototransistors, or photo- (SPR), and optical imaging. Improvements in protein
multiplier tubes, which convert the incident light into an detection methods are commonly based on improving these
electrical signal. Additional information on instrumentation for basic techniques. Other commonly used techniques, such as X-
optical detection can be found in the literature.68 Electro- ray crystallography, CD, NMR, and electron microscopy, will
chemical methods for protein detection are widely used in the not be covered. These techniques are used to analyze the
literature and have been recently reviewed.98−100 structure of a specific protein or interactions between two
3.5. Multiplexing proteins and require purified protein samples. This review
Measuring multiple different proteins in a single sample focuses on measuring unknown levels of proteins in a
simultaneously is advantageous for high-throughput analysis as biological sample. Several reviews have been recently written
well as reduced time and cost. There are two commonly used on various aspects of protein detection techniques.113−120
multiplex assay formats (Figure 6). The first format spatially 4.1. Detection of Total Protein
separates the capture reagents using a conventional microarray, Direct detection and quantification of total protein is
in which the identity of the capture reagent at each position in important for many applications. Methods using UV and
the array is predetermined by its spatial location.101,102 For visible spectroscopy are commonly used for fast quantification
299 DOI: 10.1021/acs.chemrev.8b00257
Chem. Rev. 2019, 119, 293−321
Chemical Reviews Review

of total protein concentration relative to a standard or based methods are advantageous for direct and simple detection and
on known protein extinction coefficients. The aromatic amino quantification of total protein and are widely in use.
acids, tryptophan and tyrosine and to a lesser extent 4.2. Enzyme-Linked Immunosorbent Assay (ELISA)
phenylalanine, absorb light at 280 nm, a property which
enables quantification based on absorbance. This technique is The most common tool for protein detection is the
easy to use, but disadvantages include interference from other immunoassay. The immunoassay was first introduced in
sample components, such as nucleic acids, and low sensitivity 1960 to measure plasma insulin using radioactive labels.126
due to low extinction coefficients of amino acid residues. Due to safety concerns associated with the use of radioactive
Additionally, different proteins have different absorbance labels, including the need for special facilities and disposal of
spectra depending on the structure and the number of waste, alternative methods to replace radioactive labels were
aromatic amino acid residues, which can complicate necessary. In 1971, an enzyme-based immunoassay was
quantification and reduce accuracy. developed by two different groups independently in which
Another strategy for total protein quantification is based on the radioactive label was replaced by an enzyme label.127
colorimetric protein assays. There are two general approaches. Engvall and Perlmann developed the enzyme-linked immuno-
The first approach is based on chelation of copper by proteins sorbent assay (ELISA) for antibodies in rabbit serum using
and includes the biuret assay, the bicinchoninic acid assay alkaline phosphatase,128 and van Weemen and Schuurs
(BCA assay), and the Lowry protein assay. In the biuret assay, developed the enzyme immunoassay (EIA) for human
peptides with three or more amino acid residues form a chorionic gonadotropin in urine using horseradish perox-
colored chelated complex with Cu2+ under alkaline conditions. idase.129 In an ELISA, the enzyme label is used to amplify the
The color intensity is proportional to the number of amino signal by catalyzing the formation of many thousands of
acid residues. The biuret test is a simple and fast quantitative molecules of a detectable product. In this manner, each protein
test to measure total protein concentration, with low binding event is amplified.
sensitivities in the mg/mL range. The BCA assay, developed Currently, the enzyme-linked immunosorbent assay
(ELISA) is the gold standard tool for protein detection and
in 1985, can be used to enhance sensitivity.121 It is based on
quantification. Commonly used enzymes include horseradish
the biuret reaction, in which proteins first form a chelated
peroxidase (HRP), alkaline phosphatase (AP), and β-
complex with Cu2+. Cu2+ is then reduced and chelated with
galactosidase. Many different substrates for these enzymes
BCA to form a colored complex. Another widely used
have been developed, and the products can be detected using
technique is the Lowry test, which was developed in 1951.
various detection methods including colorimetric, fluorescent,
The Lowry test is also based on the biuret reaction and
chemiluminescent, and electrochemical detection.
involves the reduction of copper (Cu2+ to Cu+) by proteins in
The main advantages of the ELISA include the ability to
alkaline solutions and formation of a colored compound via
measure proteins quantitatively with relatively high sensitivity,
reduction of a color-enhancing Folin phenol reagent.122 The
in the pg/mL range, and with a wide dynamic range that spans
colored compound is produced and can be measured at a long
4 orders of magnitude. Although ELISAs are rather time
wavelength (750 nm), which is advantageous since fewer
consuming to carry out because they involve relatively long
compounds absorb light at that wavelength and therefore the
incubation times and extensive washing, they are easy to use.
background is lower.
ELISAs are widely used for both clinical diagnostics and basic
The second approach is based on protein-binding dyes. The
research. For example, ELISAs are commonly used in the clinic
most commonly used assay is the Bradford assay, developed in
to detect C-reactive protein (CRP), a protein that plays an
1976, which utilizes the dye Coomassie G-250.123 The important role in activating the immune system.130 CRP levels
Coomassie dye binds selectively to certain amino acids and in the blood are often elevated in subjects with inflammation;
tertiary protein structures. Therefore, there can be variations therefore, elevated CRP levels can be indicative of autoimmune
between dye-binding efficiency to different proteins, which can disease or bacterial infection. Another example in which
reduce accuracy. For the dye to bind to proteins, the protein ELISAs are used in the clinic is for detecting antibodies against
mass must be high, and therefore, this method is generally used HIV in blood. HIV antibodies are detectable in the blood 3
for proteins with high molecular weights. The Bradford assay is months following infection. However, it has been shown that
rapid, simple, and widely used, but its accuracy is limited. ultrasensitive immunoassays can detect HIV earlier than the 3-
Fluorescent dyes can also be used due to their favorable month period. One example of such assay is the biobarcode
properties including increased sensitivity, decreased back- assay, which is discussed in more detail in section 4.7.131
ground, and wider dynamic range. In this method, a Approaches to improve protein detection are primarily based
nonfluorescent dye interacts with proteins either covalently on improving the performance of ELISAs.
or noncovalently. This interaction causes the dye to become
fluorescent. 1 2 4 For example, the CBQCA (3-(4- 4.3. Enzyme-Linked Immunospot (ELISPOT)
carboxybenzoyl)quinoline-2-carboxaldehyde) assay is based The enzyme-linked immunospot (ELISPOT), which was
on binding of a nonfluorescent dye to primary amines of developed in 1983, is used to detect secreted proteins by live
proteins in the presence of cyanide or thiols. The binding cells in vitro.132 ELISPOT is conceptually similar to an ELISA.
causes the dye to become fluorescent, and the fluorescence Briefly, antibodies are coated onto the surface of a microtiter
intensity is proportional to the amount of total protein. plate. A sample containing cells that secrete proteins is added,
The methods described above vary in their ability to detect and the target protein then binds to the antibodies. A second
and quantify different proteins, are subject to interference from enzyme-labeled detection antibody is then added. The enzyme
other molecules in the sample, and may require careful produces a colored precipitate. The assay is easy to perform on
calibration; therefore, the particular choice of protein assay is a large number of samples. This method is commonly used to
highly dependent on the sample type.125 Nevertheless, these detect cytokine secreting cells. ELISPOT assays have been
300 DOI: 10.1021/acs.chemrev.8b00257
Chem. Rev. 2019, 119, 293−321
Chemical Reviews Review

used to detect interferon gamma release for tuberculosis


diagnosis.133
4.4. Western Blot
The Western blot, which was developed by three different
groups independently between 1979 and 1981, is a widely used
technique in life science research to identify specific
proteins.134−136 In a Western blot, a sample is separated by
size using gel electrophoresis, transferred to a solid membrane,
blotted with a labeled antibody specific to the target protein,
and then visualized.137 Western blots can be used for
quantitative detection of proteins by using known standards. Figure 7. Protein microarrays. (a) In a planar microarray, capture
A major advantage of Western blots is the added layer of antibodies specific to different proteins are immobilized at known and
specificity, since proteins are initially separated by size. This predefined positions for multiplexed detection. Target protein binds
additional specificity contrasts with ELISA, which only detects to the capture antibody and then to a labeled detection antibody.
the label and does not provide any information on the Signal intensity is correlated with the presence and amount of the
molecular size of the protein. Even though the Western blot is target protein. (b) In a reverse phase protein microarray, the sample is
rather time consuming and can only detect high protein deposited onto the surface of the microarray and the target protein is
detected by probing with an antibody. Signal intensity is correlated
concentrations, it is widely used due to its high specificity and
with the presence of the target protein. Modified and reprinted with
simplicity, since it requires no complex instrumentation. A permission from ref 117. Copyright 2015 Nature Publishing Group.
variation of the Western blot is the dot blot, which is similar to
the Western blot but does not separate protein by electro-
phoresis. Recently, the single-cell Western blot was developed ProtoArray from ThermoFisher. Protein microarrays can be
for measuring proteins with high sensitivity on a single-cell used to study protein−protein interactions and enzymatic
level. This method is described in section 6. The Western blot activity and for biomarker discovery.141 One of the earliest
is most widely used for basic research and is commonly used to implementations of protein microarrays was for analyzing the
detect protein isoforms, truncated proteins, post-translational biochemical activity of yeast proteins, including identifying
modifications, and antibodies. Western blots are also used in proteins that interact with calmodulin and phospholipids and
the clinic. For example, HIV diagnosis is typically a two-step analyzing protein kinases.142,143 More recently, protein micro-
process in which the first step is detecting antibodies against arrays were used for high-throughput detection of antibodies
HIV in the blood using ELISA and the second step is against Plasmodium falciparum (Pf) in plasma before and after
confirming the ELISA result using a Western blot.138,139 This malaria season.144 Additionally, protein microarrays have been
combined approach leads to 99% clinical sensitivity and used for analyzing protein interactions with other molecules
specificity. including both nucleic acids145 and small molecules.146 Protein
4.5. Protein Microarrays microarrays are also used for clinical diagnostics.147 One
example is the Meso Scale Discovery (MSD) system. Another
Protein microarrays are used for multiplexed detection of
example is the SOMAscan assay, which uses a nucleic acid
proteins using reagents immobilized on a planar surface
microarray for protein detection. These methods are further
typically made of silicon, glass, or nitrocellulose. Reagents can
described in section 6.
be immobilized onto the surface of the microarray by various
approaches including contact printing or in situ synthesis of 4.6. Flow Cytometry
reagents.140 There are two types of protein microarrays. The Flow cytometry was first introduced in the 1960s and is used
first is based on a sandwich immunoassay (Figure 7A) in which to separate cells based on size and protein surface markers. In
capture antibodies are immobilized onto the surface of the flow cytometry, fluorophore-labeled antibodies bind to specific
microarray. The target protein first binds to the capture cell-surface markers. The cells are then flowed through a flow
antibodies, labeled with a detection antibody, and detected. channel. Lasers are used to excite the fluorophores, and the
This type of microarray can be used for multiplexed detection emission is measured at two different angles, providing the
of proteins by immobilizing antibodies specific to different ability to differentiate one cell at a time based on size and
proteins at known and predefined positions. In the second type surface markers. The main advantage of flow cytometers is
of microarray, also known as a reverse phase array (Figure 7B), their high throughput, namely, their ability to analyze
a sample is deposited onto the surface of the microarray and thousands of cells per second. Another advantage is the ability
the target protein is detected by probing with an antibody. to sort using fluorescence-activated cell sorting (FACS) for
Reverse phase arrays are commonly used to detect protein further downstream analysis of cells. Flow cytometry is
phosphorylation levels or antibodies. In protein microarrays, commonly used by immunologists, particularly to detect cell-
the detected signal is usually fluorescent, chemiluminescent, surface proteins known as clusters of differentiation (CDs).
electrochemiluminescent, or colorimetric. Typically, the bind- For example, analysis of cell-surface markers using flow
ing kinetics of protein microarrays are less favorable than the cytometry was instrumental for characterizing B-cell lineage
binding kinetics of solution-based assays using microspheres development. Hardy et al. showed that progenitor and
since the proteins must diffuse to the surface of the microarray precursor B cells in the bone marrow are a complex mixture
in order to bind to the capture reagent. This mass transport of cells at different stages of development.148 Flow cytometry
limitation may result in reduced sensitives for protein also has applications in clinical diagnostics of immunodefi-
microarrays. ciency and hematopoietic disorders based on cell-surface
Protein microarrays are commonly used for protein protein markers.149 In 2017, the FDA approved for the first
detection and are available commercially. One example is the time a flow-cytometry-based assay for blood cancer diag-
301 DOI: 10.1021/acs.chemrev.8b00257
Chem. Rev. 2019, 119, 293−321
Chemical Reviews Review

nostics.150 This assay detects several cell-surface markers in cleotides complementary to the oligonucleotides connecting
peripheral blood, bone marrow, and lymph node samples for the two antibodies are added and two restriction sites are
diagnosis of leukemia, non-Hodgkin lymphoma, multiple generated for digestion. The resulting DNA strands are
myeloma, myelodysplastic syndrome (MDS), and myelopro- circularized by a second ligation and then amplified by RCA.
liferative neoplasms (MPN). The amplified products are then detected by hybridization with
Other than cells, flow cytometers can also be used to detect fluorescently labeled probes and counted using a microfluidic
specific proteins using microspheres in a sandwich assay device. An alternative to PLA is the proximity extension assay
format. Microspheres are coated with specific antibodies, (PEA), in which the DNA ligase is replaced by a DNA
incubated with the target protein, and then labeled with a polymerase. PEA aims to reduce the interfering effects of DNA
second fluorescently labeled detection antibody. Flow ligase in biological samples.158 Multiplexed PLAs have been
cytometry is becoming increasingly popular for protein developed by using probes with different sequences and
quantification due to the high multiplexing capabilities. lengths, allowing for development of a 24 plex assay.159 Both
Fluorescent labels are now being replaced with metal ions in PLA and PEA provide sensitive methods for protein detection
a new technique known as mass cytometry. These new with subfemtomolar sensitivity using a low sample volume.
techniques are discussed in more detail in section 6. The main These assays can be used in solution-based assays to measure
limitation of flow cytometry is high cost, the need for a skilled protein levels and detect protein−protein interactions. They
operator, and relatively complex instrumentation. can also be used for protein analysis in tissue samples. Nucleic
4.7. Proximity Ligation Assay (PLA) and other Nucleic acid assays have been used for highly sensitive protein assays
Acid-Based Detection Methods with detection limits in the attomolar range.160,161 Recently,
An approach for signal generation and amplification is to new methods have been developed for DNA-based protein
convert protein binding into a nucleic acid signal.151,152 The detection, including use of DNA nanoswitches,162 digital
main advantage is that highly sensitive detection of nucleic droplet PCR,163,164 and incorporation of nanoparticles.165
acids is possible because DNA can be amplified. By combining Another method for ultrasensitive protein detection is the
the benefits provided by antibodies and the amplification biobarcode assay developed in the Mirkin laboratory (Figure
advantage provided by PCR, immuno-PCR was introduced 9).160,161,166,167 First, antibody-functionalized magnetic par-
and enabled protein detection with greater sensitivity than the ticles bind to the target protein. Then gold nanoparticles,
conventional immunoassay. In this approach, the target protein which are functionalized with both detection antibodies and
binds to capture antibodies immobilized on a surface. A oligonucleotide barcodes, bind to the target protein to form a
DNA−antibody conjugate then binds to the target protein. sandwich complex. The oligonucleotide barcodes are then
The DNA is amplified by PCR and then detected by gel released. Prior to detection, the biobarcodes can also be
electrophoresis.153 Further improvements to immuno-PCR amplified by PCR. The biobarcodes hybridize to probes on a
have been realized by using a sandwich assay consisting of a microarray that are complementary to one-half of the
capture antibody and a detection antibody that is conjugated biobarcode sequence. Following hybridization to complemen-
to an oligonucleotide label, amplifying the oligonucleotide tary probes on the microarray, nanoparticles functionalized
label and quantifying the label using qPCR.154 Another with oligonucleotides complementary to the other half of the
method called the proximity ligation assay (PLA) is used for biobarcode sequence hybridize. Silver amplification is used to
protein detection (Figure 8).155,156 In PLA, two biotinylated enhance the signal, and gray spots can be detected using a
Verigene instrument. The assay results in high sensitivity in the
attomolar range. The biobarcode assay has been used for the
sensitive detection of prostate specific antigen (PSA) in the
blood of patients who have undergone radical prostatec-
tomy.168 Following radical prostatectomy, PSA is usually not
detectable using a conventional ELISA. Since increasing PSA
levels can be indicative of recurrence, high-sensitivity assays
Figure 8. Proximity ligation assay (PLA). In PLA, the sample is first such as the biobarcode assay can be used to detect recurrence
incubated with antibodies conjugated to oligonucleotide probes (1). earlier.
Components for ligation and detection are added to the sample (2), Multiplexed detection can be achieved by using gold
and quantitative PCR is used for detection (3). Modified and nanoparticles each functionalized with a unique biobarcode
reprinted with permission from ref 156. Copyright 2004 National
Academy of Science. sequence (Figure 9).111 These unique biobarcodes hybridize to
complementary probes at predetermined positions on a
microarray. Another method that uses a DNA microarray for
antibodies bind to different epitopes of the target protein multiplexed protein detection is the SOMAscan assay, which is
analyte. Two different streptavidin-labeled oligonucleotides are further discussed in section 6.2.1.
bound to each of the two biotinylated antibodies. PLA is
4.8. Mass Spectrometry
designed to give a signal only when a protein is present due to
the proximity of the epitopes within the target protein. When Mass spectrometry is a common tool for proteomics analysis. It
both oligo-labeled antibodies bind to the protein, a connector is probably the most comprehensive method for analyzing
oligonucleotide can hybridize to both of the oligonucleotides, a complex mixtures because it provides a full profile of the
ligation step is performed, resulting in a DNA template that protein composition. Conventional tandem mass spectrometry
can be amplified and quantified using qPCR.156 A variation of digests proteins into peptides, and these peptides can then be
PLA utilizes rolling circle amplification (RCA) for single- used to derive the sequence of the original protein molecule.
molecule detection using digital PCR that enhances the This strategy, known as “bottom-up” proteomics, provides
sensitivity of the method.157 More specifically, two oligonu- invaluable information about proteins present in complex
302 DOI: 10.1021/acs.chemrev.8b00257
Chem. Rev. 2019, 119, 293−321
Chemical Reviews Review

Figure 9. Biobarcode assay. Target protein binds to magnetic microparticles (MMPs) functionalized with capture antibodies. Each population of
nanoparticles (NP) is functionalized with unique biobarcodes and detection antibodies specific to a target protein. Nanoparticles bind to the target
protein. Biobarcodes are then released and hybridize to probes complementary to one-half of the sequence. Gold nanoparticles then hybridized to
the other half of the sequence. Silver is used to enhance the signal, and the signal intensity is measured. Modified and reprinted with permission
from ref 166. Copyright 2006 Nature Publishing Group.

samples.169 However, since the same peptide sequence may be


present in many different proteins, it is often challenging to
identify different proteins, such as those that arise from
different gene product and splice variants. Another strategy
that may overcome this challenge is “top-down” proteomics, in
which whole, intact proteins are identified directly using
tandem mass spectrometry. This approach enables analysis of
different types of proteins; however, it is still challenging to
analyze low-abundance proteins, and quantification is also
difficult. Additional information on mass spectrometry
proteomics techniques and advances is provided in other
reviews.12,13,170−173
4.9. Lateral Flow Assay (LFA)
Lateral flow assays (LFAs) are commonly used for clinical
diagnostics and point of care detection of proteins in various
sample matrices including blood and urine.174,175 The most
commonly used assay format is the sandwich assay. An LFA
assay consists of several zones, and a typical configuration is
shown in Figure 10. First, the sample is added to the sample
application pad. The sample then moves toward the conjugate Figure 10. Sandwich lateral flow assay (LFA). (Top) In a sandwich
pad, which houses an antibody conjugated to a label. LFA, the sample is first added to the sample application pad and then
Commonly used labels for LFAs are latex particles or gold migrates toward the conjugate pad, which houses antibodies
conjugated to a label. Protein binds to the labeled antibodies, and
nanoparticles, which can form colored precipitates. Next, the sample then moves to a band that contains immobilized capture
sample, which contains the protein bound to a labeled antibodies. Sandwich complex is then formed. Control band is
antibody, moves to a band that contains immobilized capture immobilized with antibodies that capture the labeled antibody to
antibodies. A sandwich complex is then formed. Often there is ensure the test is performing correctly. Excess reagents then migrate
a second control band, which is immobilized with antibodies past the bands and are trapped in the absorbent pad. (Bottom) Signal
that capture the labeled antibody, to ensure the test is intensity of the band is proportional to the concentration of the target
performing correctly. Excess reagents then migrate past these protein. Reprinted with permission from refs 175 and 177. Copyright
2016 Elsevier and 2009 MDPI.
bands and are trapped in the wick or absorbent pad. The signal
intensity of the band is proportional to the concentration of
the target protein. The results can be visualized with the naked which measures the presence of the protein human chorionic
eye or with a reader. Usually LFAs provide qualitative or gonadotropin (hCG) in urine.176
semiquantitative protein measurements. One limitation is that 4.10. Surface Plasmon Resonance (SPR)
when very high levels of protein target are present in the Surface plasmon resonance (SPR) is a label-free method that
sample, the antibody binding sites become saturated. This can be used for protein detection. SPR occurs when
results in a high-dose hook effect, which produces a lower conduction electrons undergo oscillations at metallic surfaces
signal intensity than expected. upon exposure to light.178,179 SPR detection of proteins is
LFAs are important for clinical diagnostics. A major based on immobilization of an affinity reagent onto a metal
advantage of LFAs is that they are cheap and rapid, with surface. The affinity reagent then specifically binds to the
results provided in minutes, do not require complex protein of interest in the sample. The binding event causes a
instrumentation, and are compatible with point of care measurable change in the refractive index. SPR intensity
applications. The most common use is the pregnancy test, depends on the dielectric properties of the medium as well as
303 DOI: 10.1021/acs.chemrev.8b00257
Chem. Rev. 2019, 119, 293−321
Chemical Reviews Review

size, composition, and shape of the metallic structure.180 slide, enable high-throughput analysis of many samples. Tissue
Additionally, SPR is highly dependent on the angle of microarrays have been used for the Human Protein Atlas for
incidence of light, the wavelength of light, and the refractive analysis of protein expression in normal and cancer human
index of the sample. SPR is commonly used to study the tissue.1,192,193
binding kinetics of protein−protein interactions, such as Live cell imaging is becoming more popular to study protein
binding of proteins to antibodies.181 Additionally, SPR has localization and dynamics in live cells. Since antibodies cannot
been used to detect live malignant cells.182 Gold nanoparticles penetrate the cell membrane, proteins are genetically tagged
conjugated to antiepidermal growth factor receptor (anti- with fluorescent molecules, such as green fluorescent protein
EGFR) antibodies were added to cultures of nonmalignant and (GFP). Other systems that are based on SNAP, CLIP, and
malignant cells. The anti-EGFR antibody-conjugated nano- Halo tags have also been used.194
particles bound to the surface of the malignant cells with The study and detection of proteins in the context of their
substantially higher affinity than to the nonmalignant cells and cellular environment can be enhanced by improvements in
were detected by SPR. More recently, SPR has been used to fluorescence microscopy. Super-resolution microscopy can
detect exosomes, nanometer-sized extracellular vesicles, based resolve features less than the diffraction limit of light, which
on surface protein markers.183 is about 200 nm in the lateral direction and 500 nm in the axial
SPR is advantageous due to its ability to generate direction. These techniques include stimulated emission
quantitative measurements with a lack of labeling variations depletion (STED), photoactivation localization microscopy
associated with optical labels such as fluorescent dyes. (PALM), stochastic optical reconstruction microscopy
Localized surface plasmon resonance (LSPR) of metal (STORM), structured illumination microscopy (SIM), satu-
nanostructures can be used to enhance the signal.184,185 rated structured illumination microscopy (SSIM), ground state
Additionally, nanoparticle labels on detection antibodies in a depletion (GSD), and reversible saturated optical (fluores-
sandwich assay format can also be used to enhance the cence) transitions (RESOLFT) and have been thoroughly
signal.186,187 reviewed.195−198
4.11. Optical Imaging
5. CHALLENGES WITH PROTEIN DETECTION
The subcellular localization of proteins is important for The performance of a protein detection technique is assessed
function due to the unique chemical environment and by various parameters including sensitivity, specificity, dynamic
proximity to other interacting molecules. Delocalization of range, multiplexing capabilities, reproducibility, ease of use,
proteins may result in a lack of function and disease. Therefore, and cost. Currently, a major analytical challenge with protein
the spatial distribution of proteins in their native cellular detection techniques is measuring multiple proteins, which are
context is critical for understanding their function. Optical present in complex biological samples, with high sensitivity and
imaging-based techniques are instrumental for studying a wide dynamic range. In this section, we will discuss some of
proteins in their native cellular environment. the current challenges, including sensitivity, specificity, and
Immunohistochemistry (IHC), first developed in 1941,188 is multiplexing capabilities.
used to detect proteins and their cellular localizations in tissues High-sensitivity measurements of proteins are particularly
and cells. Traditionally, a sample is first fixed or frozen; then important for many applications. One example is the study of
the antibody binds to the target protein and is then labeled to single cells. Single-cell transcriptomics has been widely used to
produce a colored product that can be optically detected. study heterogeneity between different cells and for under-
Antibody binding can be observed either by using a standing important biological processes such as stem cell
fluorophore or an enzymatic reaction that produces a colored differentiation and cancer. However, our understanding of
product. IHC is a widely used technique for protein detection, protein levels and dynamics in single cells is limited.
particularly in clinical diagnostics, where it is used for disease Additionally, the relationship between mRNA levels and
diagnosis, prognosis, and treatment.189 Another similar protein levels on a single-cell level has been studied164,199−201
technique, immunocytochemistry (ICC), is used to analyze but is still not fully understood. Another example is
proteins in cultured cells or cells in which the extracellular measurement of blood protein biomarkers for clinical
matrix has been removed. Since sample preparation is different diagnostics, including disease diagnosis, monitoring, and
for IHC and ICC, a given antibody may not be compatible treatment. It is particularly challenging to detect low-
with both methods since the protein epitope may not be fully abundance proteins in the presence of high-abundance
exposed and cannot bind to the antibody. For IHC, a process proteins in the blood. Less than 10% of plasma proteins
known as antigen retrieval is commonly used to unmask the have been quantitatively measured due to limitations in
epitope by heating or digesting the sample with proteases. In analytical sensitivity.202,203 Therefore, high-sensitivity measure-
many cases a combination of both heating and digestion is ments of proteins are critical.
used. This process unmasks the protein epitope and allows the The sensitivity, or detection limit, is typically calculated
antibody to bind to the protein.190 based on the background signal of the assay.204,205 The
The main advantage of these techniques over other protein background signal is due to nonspecific binding of the affinity
detection methods is the ability to correlate the presence of a reagents, labeling reagents, and components in the biological
protein with its location in a tissue or cell. This feature is very sample with each other or with the surface. The detection limit
important for studying cell function in normal and pathological is usually calculated based on measuring a pure protein in a
tissues and for differentiating between healthy and diseased buffer solution. However, many biological samples contain
tissue. Recently, Thul et al. used immunofluorescence interfering substances that may increase the background signal,
microscopy to map the subcellular location of approximately and therefore, it is important to assess the detection limit in the
12 000 human proteins.191 Tissue microarrays, in which biological sample matrix. To reduce nonspecific binding arising
hundreds of different tissue samples are assembled on a single from sample components, sample separation techniques can be
304 DOI: 10.1021/acs.chemrev.8b00257
Chem. Rev. 2019, 119, 293−321
Chemical Reviews Review

employed including desalting, filtration, and affinity depletion measuring multiple proteins simultaneously with high
of other interfering molecules. As a result, the particular type of sensitivity. In this section, we will review techniques that are
sample is also important since different samples may contain currently being implemented to overcome limitations in both
different compositions of interfering substances. Another way sensitivity and multiplexing.
to overcome nonspecific binding is by optimizing buffer 6.1. Ultrasensitive Methods for Protein Detection
components, such as adding blockers. An important parameter
in protein detection is conversion of the target protein binding 6.1.1. Meso Scale Discovery (MSD). The Meso Scale
with the affinity reagent into a measurable signal. Discovery System (MSD) is based on an electrochemilumi-
Detection of a specific protein in a complex biological matrix nescent (ECL) sandwich immunoassay that can measure up to
is often challenging. Biological samples contain many protein 10 different proteins simultaneously (Figure 11).207
and nonprotein analytes that may interfere with detection of
the target protein. Some nonprotein target analytes may be
present at considerably higher levels than the target protein,
further complicating detection. Additionally, nontarget pro-
teins with similar structures may bind to the affinity reagents
and interfere with accurate quantification. High-affinity
reagents that are specific to the target protein and do not
bind to other nonanalyte molecules are necessary to enhance
specificity. Another challenge arises for some types of assays in
which an affinity reagent may specifically bind to a target
protein in a certain method but not in another. For example,
proteins are detected in their native form for sandwich
immunoassays and flow cytometry, but they are denatured in
Western blots and immunohistochemistry. Due to differences
in protein conformation and epitope structure, affinity reagents
that target the same protein may not be usable across different
methods. Therefore, detecting a specific protein target in a
biological sample is often quite challenging.
Another challenge with protein detection methods is
multiplexing. It is advantageous to simultaneously detect
multiple proteins in a single sample for increased throughput,
lower cost, reduced sample volume, and faster assay time. A
major challenge with multiplexed assays is cross-reactivity
between components in the assay, including affinity reagents
and other proteins, which often complicates detection and
Figure 11. Meso scale discovery (MSD) assay. (a) Generation of
accurate quantification. In many samples, the concentrations of electrochemiluminescence (ECL) based on oxidation of ruthenium at
different proteins in a biological sample can span several orders an electrode in the presence of tripropylamine (TPA). (b) ECL
of magnitude. Therefore, it is advantageous to have a wide ruthenium label used to modify antibodies. (c) MSD assay plate
dynamic range to measure both high- and low-abundance consisting of an injection-molded plate top and a bottom plate
proteins simultaneously in a single sample. A wide dynamic consisting of screen-printed carbon ink electrodes. (d) Well
range is also useful for measuring a single protein in different containing electrodes and antibodies. Each well contains 10 spatially
biological samples since some samples may have substantially defined positions that house capture antibodies specific to different
different levels of a given protein. Multiplexed assays also often proteins for multiplexed detection. (e) Sandwich immunoassay with
reduce the sample volume requirements. This feature is ECL detection. (f) Sector PR 400 plate reader. Reprinted with
permission from ref 207. Copyright 2007 Elsevier.
particularly important when sample volume is limited, for
example, when measuring proteins in cerebrospinal fluid,
tumor tissue samples, or blood obtained from infants. Electrochemiluminescence (ECL) involves a chemical
Therefore, a major analytical challenge is measuring multiple reaction that generates intermediates, which undergo an
proteins in a complex biological sample with both high electron transfer reaction at the electrode surface to produce
sensitivity and a wide dynamic range. excited states that emit light.208−211 Inorganic molecules, such
Finally, other analytical parameters of the assay, including as tris(2,2′-bipyridine)ruthenium(II) (Ru(bpy)32+), are widely
precision, accuracy, and reproducibility, are important. A final used to generate ECL. Due to its low molecular mass, many
consideration is whether a simple binary result, such as the Ru(bpy)32+ molecules can be attached to an antibody without
presence or absence of the target protein, is sufficient or if hindering the antibody’s ability to bind to the target protein.
quantitation is required. For detailed discussion of analytical Additionally, when a photon is emitted, the ground state is
parameters of immunoassays, the reader is referred to a regenerated, and therefore, a single label can undergo many
literature review.206 reaction cycles to produce numerous photons. Overall, signal
transduction using ECL is advantageous for several reasons
6. EMERGING METHODS FOR ULTRASENSITIVE AND including high sensitivity, wide dynamic range, simplicity, and
HIGHLY MULTIPLEXED MEASUREMENTS stability. A biological sample may contain molecules that are
Emerging new techniques have been developed to overcome autofluorescent and increase the background signal; therefore,
the challenges discussed in the previous section. A major a major advantage of ECL over fluorescence detection is the
analytical challenge with protein detection techniques is lack of a need for an excitation source. This results in lower
305 DOI: 10.1021/acs.chemrev.8b00257
Chem. Rev. 2019, 119, 293−321
Chemical Reviews Review

Figure 12. Single-molecule array (Simoa) immunoassay. (a) Single protein molecule is first captured on beads, which are functionalized with
capture antibodies. Protein is then labeled with a biotinylated detection antibody and streptavidin-ß-galactosidase. (b) Excess beads are added to a
sample containing a low concentration of protein so that only one or zero protein molecules bind to each bead. In the presence of enzyme
substrate, beads are then loaded onto an array of femtoliter-sized wells such that only one bead can fit per well. Array is sealed with oil, and
fluorescence images of the array are then obtained. (c) Scanning electron micrograph image of a microwell array containing beads inside wells of
femtoliter volume. (d) Fluorescence images of a microwell array following generation of signal from a single enzyme molecule. Reprinted with
permission from ref 219. Copyright 2010 Nature Publishing Group.

background signals by eliminating detection of fluorescence antibody-coated beads are incubated with the blood sample,
that is due to other components in the sample. Enhanced ECL most of the beads will bind zero protein molecules while a
signals can also be generated, and several reviews have been small number of beads will bind one protein molecule. A
written on the topic.212−216 negligible number of beads will bind more than one protein
In the MSD assay, which is based on ECL detection, capture molecule based on the Poisson distribution. The beads are
antibodies are first screen-printed onto the bottom of a 96- or then incubated with a biotinylated detection antibody and
384-well plate at predetermined positions. After binding of the streptavidin-ß-galactosidase, forming an enzyme-labeled im-
target protein to the capture antibody, a second detection munocomplex. The beads are then loaded onto an array of
antibody that is labeled with ruthenium binds to the target femtoliter-sized wells, in which each well is physically able to
protein. When the ruthenium complex is placed at an oxidizing hold exactly one bead. The wells are filled with a fluorogenic
electrode in the presence of tripropylamine (TPA), an ECL substrate and sealed with oil. Beads containing a single
signal is generated. The MSD assays have sensitivities in the immunocomplex will result in enzymatic conversion of many
low pg/mL range and a wide dynamic range of 4 orders of substrate molecules into product. Because the fluorescent
magnitude.217 MSD assays are commonly used to measure product is confined to an extremely small volume of
protein biomarkers in blood for clinical applications. approximately 50 fL, a high local concentration of fluorescent
6.1.2. Single-Molecule Arrays (Simoa). Digital ELISA product results that is easily detected with an imaging detector.
using single-molecule arrays (Simoa), which is being Hundreds of thousands of wells are interrogated simulta-
commercialized by Quanterix Corp., is an ultrasensitive neously, and the ratio of beads containing an enzyme label to
method for protein detection using a bead-based sandwich the number of total beads in wells corresponds to the target
ELISA.218 In a conventional sandwich ELISA, the fluorescent protein concentration in the original sample. In this manner,
product of the enzyme−substrate reaction diffuses into a large single-protein molecules can be detected using a digital
volume of approximately 50−100 μL. Therefore, millions of readout. A wider dynamic range beyond the digital range can
enzyme-labeled immunocomplexes are required to generate a be obtained by using the average fluorescence intensity of the
measurable fluorescent signal above the background. In digital active beads. This results in a dynamic range that spans over 4
ELISA, the fluorescent product of the enzyme−substrate orders of magnitude.221 Simoa immunoassays have been
reaction is confined into femtoliter-sized wells, which are developed for multiplexed detection of six different proteins
termed single-molecule arrays (Simoa).219 Using this ap- using dye-encoded beads.222 Simoa immunoassays have been
proach, the presence of a single-enzyme molecule can be used for ultrasensitive detection of biomarkers with applica-
detected.220 tions in neurology,223 cardiology,224 oncology,225,226 and
In a Simoa immunoassay (Figure 12), antibody-coated immunology.227
capture beads are added in excess to a sample containing low 6.1.3. Single-Molecule Counting (SMC). The single-
concentrations of target protein molecules. On the basis of molecule counting (SMC) platform, also known as the Erenna
Poisson statistics, either one or zero target protein molecules immunoassay system from Singulex Inc., is capable of high-
will bind to each bead. To illustrate this concept, a 100 μL sensitivity protein measurements with detection limits in the
amount of blood sample with 1 fM of the target protein sub pg/mL range.228,229 SMC is based on a traditional
contains approximately 60 000 protein molecules. If 500 000 sandwich immunoassay, in which a capture antibody
306 DOI: 10.1021/acs.chemrev.8b00257
Chem. Rev. 2019, 119, 293−321
Chemical Reviews Review

immobilized on the surface of either a microtiter plate or allowed to settle into a well. The cells are then lysed. The
magnetic beads binds to the target protein. A second detection proteins are loaded electrophoretically into the surrounding
antibody that is conjugated to a fluorophore then binds to a gel, and all of the Western blot steps including electrophoresis,
different epitope on the target protein, forming a sandwich blotting, and probing are performed. This technique has
complex. Following several washing steps, the fluorescently several advantages including the ability to assay low volumes
labeled detection antibody is eluted from the immunocomplex and low concentrations of proteins. An additional advantage is
into a small volume. The eluate is detected using a laser in a the ability to integrate antibody probing with gel electro-
small interrogation area. As the fluorescently labeled detection phoresis. This method allows detection of multiple protein
antibodies flow through the interrogation area, they are isoforms as well as truncated proteins, which may be of
detected as positive or negative events using a confocal importance for disease detection. Using this approach,
imaging system. The emitted photons are counted over defined multiplexed detection of 12 different proteins has been
time intervals, and if the photon count is sufficiently higher accomplished.232
than the background, the signal is considered positive. By 6.2. Highly Multiplexed Methods for Protein Detection
counting the number of positive events, a digital readout is
obtained. At higher protein concentrations, an analog mode is 6.2.1. SOMAscan Assay. Oligonucleotides containing
used in which the total number of photons is used. This modified nucleic acid side chains can bind to proteins with
provides a wide dynamic range of approximately 4 orders of high affinity. Various side-chain modifications for deoxyuridine
magnitude. The original system was designed for single-plex triphosphate (dUTP) have been developed to enhance binding
assays; however, recently, a three-plex assay was developed affinity. These modifications mimic the hydrophobic amino
using three labels and three different lasers.230 acid residues tryptophan and phenylalanine, which are found in
6.1.4. Single-Cell Western Blot. A sensitive protein many protein−protein binding regions. The modified aptamers
detection method for one specific application is the single-cell can achieve high affinities, with dissociation constants of less
Western blot (Figure 13).231,232 Many protein detection than 1 nM. These high-affinity reagents are termed slow off-
rate-modified aptamers, abbreviated as SOMAmers.234,235 On
the basis of these SOMAmers, SomaLogic developed the
SOMAscan assay for highly multiplexed protein detection. The
principles of the assay are shown in Figure 14. For protein
detection, SOMAmers are designed with three tags: a
fluorophore, a photocleavable linker, and biotin. The
SOMAmer reagents first bind to beads functionalized with
Figure 13. Single-cell Western blot. First, single cells settle into wells streptavidin via biotin−streptavidin interaction. Then the
in a microwell array made of a photoactive gel. Dimensions of the beads are incubated with the sample that contains the target
wells are such that only one cell can fit inside each well. Cells are then protein, and the target protein binds to the SOMAmer reagent
lysed with a denaturing RIPA buffer. After polyacrylamide gel attached to the beads. The unbound proteins are washed away,
electrophoresis (PAGE), the proteins are immobilized onto the gel and then the proteins bound to the SOMAmer are tagged with
with UV light followed by probing with a primary antibody (1° Ab) biotin. Ultraviolet light is then used to break the photo-
and then secondary labeled antibody (2° Ab*). Fluorescent images of cleavable linker and release the SOMAmer reagent. Some of
the array are obtained to detect the target protein. Chemical stripping
the released SOMAmer reagents are associated with a
and reprobing enables detection of multiple proteins. Modified and
reprinted with permission from ref 233. Copyright 2014 Nature biotinylated target protein. A polyanionic competitor is then
Publishing Group. added to further reduce nonspecific binding. Streptavidin-
functionalized beads are then used to recapture the
SOMAmer-biotinylated target protein complex. Additional
methods use antibodies to capture a specific protein. While washing steps remove nonspecifically bound SOMAmers. The
antibodies are enabling for protein detection, limitations exist SOMAmer reagent is then released in a denaturing buffer and
due to antibody specificity and cross-reactivity with other hybridized to an array of single-stranded complementary DNA
similar proteins. The Western blot, one of the most commonly probes. Fluorescence intensity is then related to the protein
used techniques for protein detection, first separates proteins levels in the sample. The SOMAscan assay has been used for
based on molecular mass using gel electrophoresis prior to biomarker discovery, primarily in biological samples such as
antibody probing and is thus more specific for the target blood.236
protein. In the traditional Western blot, thousands of cells are The main advantage of the SOMAscan assay is its high
needed to overcome the background signal. There is increasing multiplexing capabilities. SomaLogic has generated SO-
interest in analyzing proteins on the single-cell level, for MAmers specific to approximately 1300 human proteins with
example, for identification of rare circulating tumor cells sensitivities in the low-picomolar to high-femtomolar
(CTCs) in the blood. Microfluidic devices offer an advantage range.237,238 A major goal of the company is to develop
by reducing the amount of required sample, facilitating additional SOMAmers that recognize many different proteins
separation of rare CTCs from the more abundant red and to enable proteomics-level measurements. However, since the
white blood cells. However, a major challenge is downstream technology is still in its early stages, additional analytical
analysis of the isolated CTCs on a single-cell level. variables must be determined, such as cross-reactivity and assay
The single-cell Western blot addresses some of these variability.239 Nevertheless, this platform enables highly
limitations. The device is comprised of a microscope slide multiplexed protein measurements with sufficient sensitivity
and a layer of polyacrylamide gel that is patterned with an array for unbiased protein measurements.
of microwells, in which each well can physically hold only a 6.2.2. Luminex. The Luminex xMAP system is based on a
single cell. A single cell is loaded onto the microwell array and sandwich immunoassay format that incorporates flow
307 DOI: 10.1021/acs.chemrev.8b00257
Chem. Rev. 2019, 119, 293−321
Chemical Reviews Review

Figure 14. SOMAscan assay. (a) SOMAmers, which are functionalized with a fluorophore, photocleavable linker, and biotin, are immobilized onto
streptavidin (SA)-coated microspheres and incubated with a sample. (b) Target protein binds to the SOMAmer reagents. (c) Target protein is
tagged with biotin. (d) UV light is used to cleave the photocleavable linker on the SOMAmer and release the SOMAmer−protein complex from the
microspheres. (e) Addition of a buffer containing a polyanionic competitor disrupts nonspecific binding. (f) SOMAmer−protein complexes are
captured on a new set of streptavidin-coated microspheres via the biotin on the target protein. (g) Denaturing buffer is used to release the
SOMAmers from the microspheres. (h) SOMAmers hybridize to complementary probes on a microarray and are detected by fluorescence.
Fluorescence intensity is correlated to the target protein concentration. Reprinted with permission from ref 236. Copyright 2014 Elsevier.

Figure 15. Mass cytometry (CyTOF). Antibodies labeled with heavy isotopes bind to the target proteins. Each cell is nebulized and introduced into
an inductively coupled plasma (ICP) and ionized. Abundant ions are removed, and remaining heavy elements are analyzed. Signal of each isotope
label is correlated to the presence of the target protein. Reprinted with permission from ref 244. Copyright 2012 Cell Press.

cytometry as the detection platform. Luminex assays can intensity corresponding to the amount of target protein bound
achieve high multiplexing capabilities of 100 analytes by to the bead. A major advantage of the Luminex system is its
entrapping different ratios of dyes into microspheres to obtain high-multiplexing capabilities. However, commercially avail-
optically distinct populations. Each optically distinct popula- able kits typically do not exceed simultaneous measurements of
tion of microspheres is attached to capture antibodies specific more than 30 proteins due to cross-reactivity. The sensitivity of
to a given protein. Following binding of the target protein, a Luminex assays is generally between 1 and 10 pg/mL range,
second antibody conjugated to phycoerythrin (PE) binds to and the dynamic range spans approximately 3−4 orders of
the protein. The microspheres are then analyzed using a flow magnitude. The Luminex system is widely used to measure
cytometer to detect the internal fluorescent labels to identify proteins for various research and approved in vitro diagnostics
the microsphere’s specificity and measure the PE emission (IVDs) applications including infectious diseases, human
308 DOI: 10.1021/acs.chemrev.8b00257
Chem. Rev. 2019, 119, 293−321
Chemical Reviews Review

leukocyte antigen (HLA) detection, and more.240 Another 7.1.1. Upconverting Nanoparticles. Lanthanides exhibit
similar platform is the cytometric bead array (CBA), a bead- anti-Stokes luminescence or upconversion luminescence.250
based sandwich immunoassay based on flow cytometry, that is Upconversion is a nonlinear optical process in which
used for multiplexed detection of proteins.241,242 absorption of two or more photons leads to emission of a
6.2.3. Mass Cytometry (CyTOF). Mass cytometry was photon at a shorter wavelength. Upconverting nanoparticles
developed in 2009 to allow for protein detection on a single- can be used in a sandwich assay for protein detection, in which
cell level.243 Mass cytometry, also known as CyTOF, combines the detection antibody is labeled with an upconverting
the principles of flow cytometry and atomic mass spectrom- nanoparticle. The label is excited in the infrared region and
etry, enabling analysis of 40 parameters per cell with very high emits in the visible region for detection (Figure 16).251
throughput.244,245 The work flow of single-cell mass cytometry
(Figure 15) includes coupling antibodies to a stable non-
radioactive rare-earth metal isotope. The labeled antibodies
bind to the target protein of interest. The labeled cell is then
nebulized into a droplet and introduced into an inductively
coupled argon plasma (ICP). Once the cell enters the plasma,
the sample is vaporized and the cellular material in the droplet
is ionized, including the abundant isotopes in the cell such as
carbon, nitrogen, and oxygen. The ionized sample is passed
through a mass filter, which allows only the heavy reporter ions
through, and these ions are analyzed by the mass spectrometer.
The mass spectrometer then measures the composition of the
metal isotopes. This number is correlated to the abundance of
antibody bound targets in the original sample. Mass cytometry
has been used for various applications. Examples include Figure 16. Upconverting nanoparticles. Absorption of near IR light
single-cell analysis of immune and drug responses246 and (NIR) and energy transfer lead to upconverted blue, green, or red
analysis of T cells in healthy and HIV-infected subjects.247 emissions. Reprinted with permission from ref 251. Copyright 2009
Recently, metal isotopes have been used for multiplexed John Wiley and Sons.
detection of proteins in tissue samples.248,249
The main advantage of mass cytometry is the ability to Upconverting nanoparticles have several favorable optical
overcome the need for fluorophores by labeling antibodies properties including high stability and quantum yield. Addi-
with rare-earth heavy metals. The use of heavy metals provides tionally, background arising from absorbing molecules in a
an advantage for multiplexed detection since there are about biological sample is low since most biomolecules in a sample
100 isotopes that are potentially available for mass cytometry absorb in the UV region and not in the IR region.
and only 18 fluorophores for flow cytometry due to the Upconverting nanoparticles have been used as optical labels
spectral overlap of fluorophores. Additionally, the signal from for multiplexed detection and optical encoding.252−254 Several
the heavy rare-earth labels has low background because there reviews have been written on their properties and applica-
are no rare earths present in biological samples. Therefore, the tions.255−262
measured signal is highly specific to the rare-earth labels. This 7.1.2. Photoelectrochemical Detection (PEC). In
contrasts with optical detection of fluorophores, in which the photoelectrochemical (PEC) signal transduction, light is used
biological sample may contain molecules that are autofluor-
to excite photoactive species, which produce a measurable
escent and interfere with detection of the fluorescent label.
electrical signal (Figure 17). Since the excitation source
Another advantage of mass cytometry is its very high
(optical) is different than the detected signal (electrical), PEC-
throughput, with an ability to assay millions of single cells.
The main disadvantages of the method are that it can provide
only relative quantification and has low sensitivity. Several
other disadvantages are a lack of ability to sort the cells since
they are vaporized, challenges with quality control, statistical
analysis of complex data, and limited availability of commercial
rare-earth metal-labeled antibodies.

7. ADVANCES FROM THE LITERATURE


7.1. Advances in Labeling and Signal Detection
A major consideration for protein assays is converting the
binding of the target protein to an affinity reagent into a
measurable optical, electrochemical, or mechanical signal. In
general, enhancing the signal leads to higher sensitivities.
Various approaches to enhance the signal of labeling reagents
have been developed. In this section, we discuss several novel Figure 17. Schematic of photoelectrochemical detection of proteins.
and promising approaches for labeling and signal detection, Affinity reagent immobilized on the surface binds to the target
including upconverting nanoparticles, photoelectrochemical protein, resulting in steric hindrance that decreases the intensity of the
detection, optical ring resonators, and surface-enhanced photocurrent. Reprinted with permission from ref 265. Copyright
Raman scattering (SERS) tags. 2015 American Chemical Society.

309 DOI: 10.1021/acs.chemrev.8b00257


Chem. Rev. 2019, 119, 293−321
Chemical Reviews Review

based signal detection could have potentially high sensitivity single molecules.275−278 Additionally, multiplexed detection for
due to reduced background.263−266 Therefore, even if bio- five protein analytes has been demonstrated using arrays of
logical molecules in the sample are excited, they will likely not microring resonators.279 Additional information on WGM is
be detected. There are two essential components of PEC provided in refs 280 and 281.
assays for protein detection: an affinity reagent that is near the 7.1.4. Surface-Enhanced Raman Scattering (SERS)
signal transducer and a photoactive species. Typical molecules Tags. A molecule in a sample can be identified based on its
used for PEC signal generation are organic or inorganic Raman spectra. Raman spectroscopy on its own is not
semiconductors, quantum dots,267,268 or secondary enzyme sufficient to detect protein molecules in most cases because
labels, such as glucose oxidase, horseradish peroxidase, alkaline Raman signals are weak. Therefore, surface-enhanced Raman
phosphatase, and acetylcholine esterase, that can produce scattering (SERS) is often used in which a metal surface,
photocurrent due to catalysis. generally composed of gold or silver, enhances the Raman
Development of PEC-based assays for protein detection has spectral lines. SERS tags, metal nanoparticles composed of a
gained momentum in recent years. Two general approaches Raman reporter that has strong Raman scattering, a protective
have been used. The first is label-free detection in which shell, and an affinity reagent, have been used to enhance the
binding of the target protein to an affinity reagent results in sensitivity of Raman-based protein assays. SERS tags have
steric hindrance that decreases the intensity of the photo- several advantages including high sensitivity,282 spectral lines
current.269,270 The second approach is based on a typical with narrow line widths, and multiplexing capabilities.
sandwich-assay format in which an affinity reagent binds to the Furthermore, SERS tags have high stability and do not
target protein, and subsequent labeling with a PEC active photobleach. SERS tags can be detected with NIR lasers to
species results in measurable photocurrent. High-sensitivity minimize background autofluorescence arising from compo-
PEC protein assays with detection limits in the subpg/mL nents in a biological sample.283 A multiplexed sandwich assay
range have been developed.271 Recently, multiplexed protein using SERS tags as labels is shown in Figure 19.284 Different
detection methods based on an antibody array or two different approaches have been developed for protein detection using
enzyme labels that produce distinguishable PEC signals were SERS.285−287 Several excellent reviews have been written on
developed.272−274 PEC assays are an emerging class of protein use of SERS tags and bioassays.288−291
assays due to their potential for high sensitivity and relative 7.2. Miniaturization
simplicity. Miniaturized assays are advantageous due to size compatibility
7.1.3. Optical Ring Resonators. In an optical ring between the target protein and the assay format. Major
resonator, light undergoes total internal reflection along a advantages of these systems are low volume requirements and
curved boundary in a structure known as an optical enhanced sensitivity. In this section, we discuss miniaturized
microcavity, resulting in light propagation in circulating approaches for protein detection including mechanical
waveguide modes or whispering gallery modes (WGMs). detection using microcantilever arrays and ultrasmall contain-
The sensing layer is modified with affinity reagents, and when ers.
the target protein binds there is a shift in the resonance 7.2.1. Microcantilever-Based Assays. Advances in
wavelength. Figure 18 shows a schematic of WGM for protein micro- and nanofabrication technologies have enabled develop-
detection. WGM can have high sensitivity capable of detecting ment of miniaturized mechanical-based assays for protein
detection in which binding of a protein analyte to an affinity
reagent leads to a detectable change in mass.292,293 Mechanical
assays have several advantages for protein detection. They can
achieve yoctogram resolution,294 with single-protein molecule
detection,295 are amenable to multiplexing, are low cost, and
can be used for point of care applications.296 Additionally, they
are available in a label-free assay format but can also be
integrated with secondary labeled affinity reagents to enhance
the signal.
Recently, microcanteliver assays have been used for sensitive
and multiplexed protein detection based on mechanical signal
transduction.297−300 In such assays, an affinity reagent
immobilized on the surface of the microcantilever specifically
binds to the target protein, resulting in a shift of the resonance
frequency or lateral sheer stress that leads to bending of the
microcantilever.301 The resonance frequency and bending can
be measured using piezoresistivity or optical deflection of a
light beam at the surface of the microcantilever. A scheme is
shown in Figure 20.302
Figure 18. Whispering gallery mode (WGM) sensor for protein Microcantilevers have several advantages for protein
detection. (a) Resonance shift is associated with protein binding. (b)
detection One advantage of microcantilever arrays is their
WGM in a dielectric sphere. Light circumnavigates the glass sphere.
Antibodies are immobilized onto the surface, and binding of the ability for multiplexed detection (Figure 20B).303−305 Addi-
protein results in a shift of the resonance wavelength. (c) Single- tionally, microcantilever assays have a fast response time and
protein molecule binding events are theorized to appear as steps in low cost. They are also amenable to microfabrication
the wavelength shift. Reprinted with permission from ref 275. technologies and integration into portable point of care
Copyright 2008 Nature Publishing Group. devices. Microcantilevers can be used for highly sensitive and
310 DOI: 10.1021/acs.chemrev.8b00257
Chem. Rev. 2019, 119, 293−321
Chemical Reviews Review

Figure 19. Schematic of multiplex protein sandwich assay on a porous hydrogel bead using SERS nanotags. Porous hydrogel beads are modified
with capture antibodies, which bind to the target protein. Target protein is then labeled with SERS nanotags which are functionalized with a
detection antibody. Raman dyes are embedded in the Au core and the Ag shell. Two different SERS nanotags have been used for multiplexed
detection. Reprinted with permission from ref 284. Copyright 2018 American Chemical Society.

Figure 20. Microcantilever for protein detection. (a) Affinity reagents are immobilized onto the surface of the microcantilever. Antifouling
molecules are coated onto the bottom surface of the microcantilevers to reduce nonspecific binding. Target protein then binds to the affinity
reagent. Secondary gold−nanoparticle functionalized with antibodies binds to the protein. (b) SEM of the microcantilevers. (c) Optical beam
deflection for measuring microcantilever vibration. (d) Mass loading onto the microcantilever results in a shift in resonance frequency. Reprinted
with permission from ref 302. Copyright 2014 Nature Publishing Group.

quantitative detection of proteins.306−308 Improvements in container. Protein molecules are a few nanometers in size and a
sensitivity using secondary antibodies tagged with nano- single cell about a few micrometers in size. Analysis performed
particles, which increase the mass response of the microcanti- in a test tube, in which a single protein molecule or a single cell
lever assays, have resulted in sensitivity in the low fg/mL is present in a large volume, will likely result in inability to
range.302 An important consideration is nonspecific binding at locate, capture, and detect the protein molecule or cell.
the bottom surface of the microcantilever. Surface passivation Confinement of molecules into ultrasmall containers enables
can be used to overcome this limitation.309 detection of proteins at the single-molecule and single-cell
7.2.2. Ultrasmall Containers. Miniaturized systems are level.
favorable for protein detection due to the compatibility Several approaches using ultrasmall volume containers for
between the target protein molecule and the size of the protein analysis have been developed. One example is the
311 DOI: 10.1021/acs.chemrev.8b00257
Chem. Rev. 2019, 119, 293−321
Chemical Reviews Review

Figure 21. Single-molecule enzyme analysis using a microwell array. (a) Enzyme F0F1 ATPase pumps protons from the outside to the inside of the
well by hydrolyzing ATP. Fluorescent pH indicator is present inside the well and monitors changes in pH. (b) Fluorescence images after addition of
ATP at 0 and 6000 s. Right panel (blue) shows the difference in intensity between the two time points. Reprinted with permission from ref 311.
Copyright 2014 Nature Publishing Group.

single-molecule array (Simoa) immunoassay, which was


discussed in section 6.2.2. In this technique, the fluorescent
product of the enzyme−substrate reaction is confined to a
small volume of approximately 50 fL. Another example is the
SlipChip immunoassay in which a bead-based immunoassay is
performed in nanoliter volumes.310 Finally, ultrasmall contain-
ers can also be used to measure the enzymatic activity of
proteins. For example, this approach was used to study the
transporter activity of F0F1 ATPase, which pumps protons
across a membrane by hydrolyzing ATP (Figure 21).311 In this
method, a fluorescent pH indicator is first added to the wells
on a microwell array. The microwell array is then coated with a
lipid bilayer, which mimics the lipid bilayer of a cell. Liposomes
embedded with zero, one, or two molecules of the enzyme
F0F1 are then introduced onto the array, and some fuse with
the lipid bilayer on the microwells. ATP is then added to
initiate the proton-pumping enzymatic activity of F0F1 ATPase
across the lipid bilayer. Fluorescent images are then obtained
to detect single-molecule activity. Another example is the
Nanopore technology, which can be used to detect single-
protein molecules.312 In this method, changes in ionic current
when the protein moves into the pore are monitored.
Nanopores have been used to distinguish between different Figure 22. Highly multiplexed detection of secreted proteins from
phosphorylation states. single cells. Single cells are first captured inside a microwell array.
Ultrasmall containers have also been used for single-cell Following incubation, secreted cytokines are captured onto capture
analysis. One application is detection of cytokine release from antibodies. Barcoded slide is then removed, and a sandwich assay is
single cells using an array of wells in which only one cell can fit performed. By using three optical encodings and 15 spatial encodings,
per well.313 Using this approach, highly multiplexed detection a 45-plex assay is possible. Modified and reprinted with permission
of secreted proteins from single cells was developed (Figure from ref 315. Copyright 2015 National Academy of Sciences.
22).314,315 In this method, single cells are first isolated inside
individual microchambers. Fifteen capture antibodies specific droplet digital PCR (ddPCR) for mRNA quantification
to different cytokines are coated onto the surface of the (Figure 23).164 Single cells are isolated and then lysed, and
chamber at known and predefined positions. Following a 12− the cell lysate is split into two separate samples. For digital
24 h incubation, the capture antibodies bind to the secreted PLA, the cell lysate sample is incubated with oligonucleotide-
cytokines. The barcoded chamber is then removed, and a bound antibodies. The antibodies bind to the target protein,
sandwich assay is performed by using three different optical bringing the oligonucleotides near each other. A connector
labels. Using a combination of spatial and spectral encoding, a oligonucleotide is added that hybridizes to the probes to form
42-plex assay was developed. A second approach based on a complex consisting of the target protein analyte, the probes,
isolating single cells inside microwells is the single-cell Western and connector oligonucleotide. Ligation is performed, forming
blot, which was previously described in section 6.1.4. double-stranded DNA (dsDNA) followed by proteolytic
Another method used to quantify proteins and mRNA levels digestion, resulting in a solution containing the dsDNA.
simultaneously in single mammalian cells is based on a digital Using a commercially available device for ddPCR (BioRad),
proximity ligation assay (PLA) for protein quantification and the solution containing the dsDNA is diluted and emulsified to
312 DOI: 10.1021/acs.chemrev.8b00257
Chem. Rev. 2019, 119, 293−321
Chemical Reviews Review

Figure 23. Digital Droplet PCR for quantification of proteins and mRNA in single cells. Single cells are isolated and then lysed, and the cell lysate is
split into two separate samples. For protein detection, PLA is performed followed by proteolytic digestion, resulting in a solution containing
dsDNA. Solution containing the dsDNA is diluted and emulsified to form about 20 000 nL droplets, such that each droplet contains either 1 or 0
dsDNA molecules. Each dsDNA molecule is then amplified by PCR, and digital counting is performed by measuring the fluorescence using a
droplet reader. For mRNA detection, digital droplet PCR is performed. In this manner, both protein and mRNA from a single cell can be measured.
Reprinted with permission from ref 164. Copyright 2016 Cell Press.

form about 20 000 nanoliter droplets, such that each droplet protein measurements. Ultrasensitive methods such as the
contains either one or zero dsDNA molecules. Each dsDNA Meso Scale Discovery assay (MSD) and single-molecule arrays
molecule is then amplified by PCR, and digital counting is (Simoa) are being used to detect previously unmeasurable
performed by measuring the fluorescence using a droplet levels of proteins. Recent advances from the literature, such as
reader. Digital PLA using ddPCR is combined with RT-ddPCR novel optical labels and assay miniaturization, enable protein
for simultaneous quantification of both protein and mRNA detection on the single-molecule and single-cell level. These
from a single cell. For protein quantification, femtomolar assays will undoubtedly lead to interesting new discoveries in
sensitivity corresponding to approximately 10 000 molecules biology as well as new clinical diagnostics applications.
was achieved with a dynamic range of about 3 orders of
magnitude. For mRNA quantification, a limit of detection of AUTHOR INFORMATION
approximately 52 mRNAs per cell was achieved.
Corresponding Author
8. CONCLUSION AND FUTURE DIRECTIONS *E-mail: dwalt@bwh.harvard.edu.
In this review, we discussed the wide variety of approaches for ORCID
protein detection. Several parameters, including sensitivity, Limor Cohen: 0000-0003-1448-0925
specificity, dynamic range, multiplexing capabilities, reprodu-
David R. Walt: 0000-0002-5524-7348
cibility, ease of use, and cost, are important in protein
detection techniques. A major analytical challenge with current Notes
protein detection techniques is measuring multiple proteins The authors declare the following competing financial
that are present in complex biological samples with high interest(s): David R. Walt is the scientific founder and a
sensitivity and a wide dynamic range. Strategies to overcome board member of Quanterix Corporation. All other authors
these challenges have been developed; however, there is still a declare no competing financial interest.
trade-off between high-sensitivity measurements and highly
multiplexed measurements of proteins. As a result, it is often Biographies
necessary to decide whether it is more advantageous to detect
David R. Walt is Professor of Pathology at Harvard Medical School
several specific proteins using a candidate-based, hypothesis-
and Brigham and Women’s Hospital, a Core Faculty Member of the
driven approach or to detect as many proteins as possible using
Wyss Institute at Harvard University, and a Howard Hughes Medical
an unbiased, high-throughput approach that is less sensitive.
Institute. He is the Scientific Founder of multiple life sciences
A major challenge in ultrasensitive and highly multiplexed
companies. He has received numerous national and international
protein detection is nonspecific binding between biomolecules
awards for his work in the field of optical microwell arrays and single
in the sample, assay reagents, and surfaces. Nonspecific binding
molecules. In addition to his lab’s fundamental work on single
leads to background signals that obscure target protein
molecules, his lab develops new diagnostics technologies and applies
binding. Surface passivation methods that reduce nonspecific
them to pressing clinical problems. He is a member of the National
binding as well as development of high-affinity reagents that do
Academy of Engineering, the National Academy of Medicine, and a
not cross-react will enhance the performance of protein
detection methods. In addition, low-cost protein detection Fellow of the American Academy of Arts and Sciences, the American
methods for applications in developing countries or point of Institute for Medical and Biological Engineering, and the National
care clinical applications are still necessary. Academy of Inventors.
Nevertheless, major advances in protein detection methods Limor Cohen is a Ph.D. candidate in the Department of Chemical
have been developed in recent years. One example is the Biology at Harvard University. She received her B.A. degree in
SOMAscan assay, which is capable of highly multiplexed Chemistry from New York University in 2012. Her research interests

313 DOI: 10.1021/acs.chemrev.8b00257


Chem. Rev. 2019, 119, 293−321
Chemical Reviews Review

lie in the development of analytical tools for ultrasensitive detection of (23) Köhler, G.; Milstein, C. Continuous Cultures of Fused Cells
proteins and nucleic acids along with clinical applications. Secreting Antibody of Predefined Specificity. Nature 1975, 256, 495−
497.
(24) Boder, E. T.; Raeeszadeh-Sarmazdeh, M.; Price, J. V.
REFERENCES Engineering Antibodies by Yeast Display. Arch. Biochem. Biophys.
(1) Uhlen, M.; Fagerberg, L.; Hallstrom, B. M.; Lindskog, C.; 2012, 526, 99−106.
Oksvold, P.; Mardinoglu, A.; Sivertsson, A.; Kampf, C.; Sjostedt, E.; (25) Hanes, J.; Schaffitzel, C.; Knappik, A.; Plückthun, A. Picomolar
Asplund, A.; et al. Tissue-Based Map of the Human Proteome. Science Affinity Antibodies from a Fully Synthetic Naive Library Selected and
2015, 347, 1260419. Evolved by Ribosome Display. Nat. Biotechnol. 2000, 18, 1287−1292.
(2) Aebersold, R.; et al. How Many Human Proteoforms Are There? (26) Hanes, J.; Plückthun, A. In Vitro Selection and Evolution of
Nat. Chem. Biol. 2018, 14, 206−214. Functional Proteins by Using Ribosome Display. Proc. Natl. Acad. Sci.
(3) Schmid, A.; Kortmann, H.; Dittrich, P. S.; Blank, L. M. Chemical U. S. A. 1997, 94, 4937−4942.
and Biological Single Cell Analysis. Curr. Opin. Biotechnol. 2010, 21, (27) Dufner, P.; Jermutus, L.; Minter, R. R. Harnessing Phage and
12−20. Ribosome Display for Antibody Optimisation. Trends Biotechnol.
(4) Walsh, C. T.; Garneau-Tsodikova, S.; Gatto, G. J. Protein 2006, 24, 523−529.
Posttranslational Modifications: The Chemistry of Proteome (28) Winter, G.; Griffiths, A. D.; Hawkins, R. E.; Hoogenboom, H.
Diversifications. Angew. Chem., Int. Ed. 2005, 44, 7342−7372. R. Making Antibodies by Phage Display Technology. Annu. Rev.
(5) Jiang, H.; Zhang, X.; Chen, X.; Aramsangtienchai, P.; Tong, Z.; Immunol. 1994, 12, 433−455.
Lin, H. Protein Lipidation: Occurrence, Mechanisms, Biological (29) Landry, J. P.; Fei, Y.; Zhu, X. Simultaneous Measurement of
Functions, and Enabling Technologies. Chem. Rev. 2018, 118, 919− 10,000 Protein-Ligand Affinity Constants Using Microarray-Based
988. Kinetic Constant Assays. Assay Drug Dev. Technol. 2012, 10, 250−
(6) Mann, M.; Jensen, O. N. Proteomic Analysis of Post- 259.
Translational Modifications. Nat. Biotechnol. 2003, 21, 255−261. (30) Boder, E. T.; Midelfort, K. S.; Wittrup, K. D. Directed
(7) Witze, E. S. S.; Old, W. M. M.; Resing, K. A. A.; Ahn, N. G. G. Evolution of Antibody Fragments with Monovalent Femtomolar
Mapping Protein Post-Translational Modifications with Mass Antigen-Binding Affinity. Proc. Natl. Acad. Sci. U. S. A. 2000, 97,
Spectrometry. Nat. Methods 2007, 4, 798−806. 10701−10705.
(8) Davies, M. J. Protein Oxidation and Peroxidation. Biochem. J. (31) Byrne, B.; Stack, E.; Gilmartin, N.; O’Kennedy, R. Antibody-
2016, 473, 805−825. Based Sensors: Principles, Problems and Potential for Detection of
(9) Halliwell, B.; Gutteridge, J. M. C. Free Radicals in Biology and Pathogens and Associated Toxins. Sensors 2009, 9, 4407−4445.
Medicine, 5th ed.; Oxford University Press: Oxford, UK, 2015. (32) Jayasena, S. D. Aptamers: An Emerging Class of Molecules
(10) Cohen, P. The Origins of Protein Phosphorylation. Nat. Cell That Rival Antibodies in Diagnostics. Clin. Chem. 1999, 45, 1628−
Biol. 2002, 4, E127−E130. 1650.
(11) Corthals, C. L.; Wasinger, V. C.; Hochstrasser, D. F.; Sanchez, (33) Hermann, T.; Patel, D. J. Adaptive Recognition by Nucleic Acid
J. C. The Dynamic Range of Protein Expression: A Challenge for Aptamers. Science 2000, 287, 820−825.
Proteomic Research. Electrophoresis 2000, 21, 1104−1115. (34) Ellington, A. D.; Szostak, J. W. In Vitro Selection of RNA
(12) Yates, J. R.; Ruse, C. I.; Nakorchevsky, A. Proteomics by Mass Molecules That Bind Specific Ligands. Nature 1990, 346, 818−822.
Spectrometry: Approaches, Advances, and Applications. Annu. Rev. (35) Tuerk, C.; Gold, L. Systematic Evolution of Ligands by
Biomed. Eng. 2009, 11, 49−79. Exponential Enrichment:RNA Ligands to Bacteriophage T4 DNA
(13) Mann, M.; Hendrickson, R. C.; Pandey, A. Analysis of Proteins Polymerase. Science 1990, 249, 505−510.
and Proteomes by Mass Spectrometry. Annu. Rev. Biochem. 2001, 70, (36) Toh, S. Y. i.; Citartan, M.; Gopinath, S. C. B.; Tang, T. H.
437−473.
Aptamers as a Replacement for Antibodies in Enzyme-Linked
(14) Hulanicki, A.; Glab, S.; Ingman, F. Chemical Sensors:
Immunosorbent Assay. Biosens. Bioelectron. 2015, 64, 392−403.
Definitions and Classification. Pure Appl. Chem. 1991, 63, 1247−
(37) Cho, E. J.; Lee, J.-W.; Ellington, A. D. Applications of Aptamers
1250.
as Sensors. Annu. Rev. Anal. Chem. 2009, 2, 241−264.
(15) Hermanson, G. T. Bioconjugate Techniques, 3rd ed.; Academic
(38) Darmostuk, M.; Rimpelova, S.; Gbelcova, H.; Ruml, T. Current
Press: San Diego, CA, 2013.
(16) Goldman, E. R.; Anderson, G. P.; Liu, J. L.; Delehanty, J. B.; Approaches in SELEX: An Update to Aptamer Selection Technology.
Sherwood, L. J.; Osborn, L. E.; Cummins, L. B.; Hayhurst, A. Facile Biotechnol. Adv. 2015, 33, 1141−1161.
Generation of Heat-Stable Antiviral and Antitoxin Single Domain (39) Whitcombe, M. J.; Chianella, I.; Larcombe, L.; Piletsky, S. A.;
Antibodies from a Semisynthetic Llama Library. Anal. Chem. 2006, Noble, J.; Porter, R.; Horgan, A. The Rational Development of
78, 8245−8255. Molecularly Imprinted Polymer-Based Sensors for Protein Detection.
(17) Hamers-Casterman, C.; Atarhouch, T.; Muyldermans, S.; Chem. Soc. Rev. 2011, 40, 1547−1571.
Robinson, G.; Hammers, C.; Songa, E. B.; Bendahman, N.; Hammers, (40) Haupt, K.; Mosbach, K. Molecularly Imprinted Polymers and
R. Naturally Occurring Antibodies Devoid of Light Chains. Nature Their Use in Biomimetic Sensors. Chem. Rev. 2000, 100, 2495−2504.
1993, 363, 446−448. (41) Lee, Y.-C.; Block, G.; Chen, H.; Folch-Puy, E.; Foronjy, R.;
(18) Harmsen, M. M.; De Haard, H. J. Properties, Production, and Jalili, R.; Jendresen, C. B.; Kimura, M.; Kraft, E.; Lindemose, S.; et al.
Applications of Camelid Single-Domain Antibody Fragments. Appl. One-Step Isolation of Plasma Membrane Proteins Using Magnetic
Microbiol. Biotechnol. 2007, 77, 13−22. Beads with Immobilized Concanavalin A. Protein Expression Purif.
(19) Muyldermans, S. Nanobodies: Natural Single-Domain Anti- 2008, 62, 223−229.
bodies. Annu. Rev. Biochem. 2013, 82, 775−797. (42) Squires, T. M.; Messinger, R. J.; Manalis, S. R. Making It Stick:
(20) Yu, X.; Yang, Y.-P.; Dikici, E.; Deo, S. K.; Daunert, S. Beyond Convection, Reaction and Diffusion in Surface-Based Biosensors. Nat.
Antibodies as Binding Partners: The Role of Antibody Mimetics in Biotechnol. 2008, 26, 417−426.
Bioanalysis. Annu. Rev. Anal. Chem. 2017, 10, 293−320. (43) Love, J. C.; Estroff, L. A.; Kriebel, J. K.; Nuzzo, R. G.;
(21) Doshi, R.; Chen, B. R.; Vibat, C. R. T.; Huang, N.; Lee, C. W.; Whitesides, G. M. Self-Assembled Monolayers of Thiolates on Metals
Chang, G. In Vitro Nanobody Discovery for Integral Membrane as a Form of Nanotechnology. Chem. Rev. 2005, 105, 1103−1169.
Protein Targets. Sci. Rep. 2015, 4, 6760. (44) Aubin-Tam, M. E.; Hamad-Schifferli, K. Structure and Function
(22) Schatz, D. G.; Ji, Y. Recombination Centres and the of Nanoparticle-Protein Conjugates. Biomed. Mater. 2008, 3, 034001.
Orchestration of V(D)J Recombination. Nat. Rev. Immunol. 2011, (45) Sapsford, K. E.; Algar, W. R.; Berti, L.; Gemmill, K. B.; Casey,
11, 251−263. B. J.; Oh, E.; Stewart, M. H.; Medintz, I. L. Functionalizing

314 DOI: 10.1021/acs.chemrev.8b00257


Chem. Rev. 2019, 119, 293−321
Chemical Reviews Review

Nanoparticles with Biological Molecules: Developing Chemistries (68) Lakowicz, J. R. Principles of Fluorescence Spectroscopy, 3rd ed.;
That Facilitate Nanotechnology. Chem. Rev. 2013, 113, 1904−2074. Springer: Baltimore, MD, 2006.
(46) Trilling, A. K.; Beekwilder, J.; Zuilhof, H. Antibody Orientation (69) Panchuk-Voloshina, N.; Haugland, R. P.; Bishop-Stewart, J.;
on Biosensor Surfaces: A Minireview. Analyst 2013, 138, 1619−1627. Bhalgat, M. K.; Millard, P. J.; Mao, F.; Leung, W. Y.; Haugland, R. P.
(47) Bonanno, L. M.; DeLouise, L. A. Steric Crowding Effects on Alexa Dyes, a Series of New Fluorescent Dyes That Yield
Target Detection in an Affinity Biosensor. Langmuir 2007, 23, 5817− Exceptionally Bright, Photostable Conjugates. J. Histochem. Cytochem.
5823. 1999, 47, 1179−1188.
(48) Saha, B.; Evers, T. H.; Prins, W. J.; et al. How Antibody Surface (70) Gao, N.; Gao, T.; Yang, X.; Dai, X.; Zhou, W.; Zhang, A.;
Coverage on Nanoparticles Determines the Activity and Kinetics of Lieber, C. M. Specific Detection of Biomolecules in Physiological
Antigen Capturing for Biosensing. Anal. Chem. 2014, 86, 8158−8166. Solutions Using Graphene Transistor Biosensors. Proc. Natl. Acad. Sci.
(49) Byzova, N. A.; Safenkova, I. V.; Slutskaya, E. S.; Zherdev, A. V.; U. S. A. 2016, 113, 14633−14638.
Dzantiev, B. B. Less Is More: A Comparison of Antibody-Gold (71) Shen, J.; Li, Y.; Gu, H.; Xia, F.; Zuo, X. Recent Development of
Nanoparticle Conjugates of Different Ratios. Bioconjugate Chem. Sandwich Assay Based on the Nanobiotechnologies for Proteins,
2017, 28, 2737−2746. Nucleic Acids, Small Molecules, and Ions. Chem. Rev. 2014, 114,
(50) Š pringer, T.; Chadtová Song, X.; Ermini, M. L.; Lamačová, J.; 7631−7677.
Homola, J. Functional Gold Nanoparticles for Optical Affinity (72) Farka, Z.; Juřík, T.; Kovár,̌ D.; Trnková, L.; Skládal, P.
Biosensing. Anal. Bioanal. Chem. 2017, 409, 4087−4097. Nanoparticle-Based Immunochemical Biosensors and Assays: Recent
(51) Malaspina, D. C.; Longo, G.; Szleifer, I. Behavior of Ligand Advances and Challenges. Chem. Rev. 2017, 117, 9973−10042.
Binding Assays with Crowded Surfaces: Molecular Model of Antigen (73) Xu, X.; Ray, R.; Gu, Y.; Ploehn, H. J.; Gearheart, L.; Raker, K.;
Capture by Antibody-Conjugated Nanoparticles. PLoS One 2017, 12, Scrivens, W. A. Electrophoretic Analysis and Purification of
e0185518. Fluorescent Single-Walled Carbon Nanotube Fragments. J. Am.
(52) Katz, E.; Willner, I. Biomolecule Functionalized Carbon Chem. Soc. 2004, 126, 12736−12737.
Nanotubes: Applications in Nanobioelectronics. ChemPhysChem (74) Sun, Y. P.; Zhou, B.; Lin, Y.; Wang, W.; Fernando, K. A. S.;
2004, 5, 1084−1104. Pathak, P.; Meziani, M. J.; Harruff, B. A.; Wang, X.; Wang, H.; et al.
(53) Balasubramanian, K.; Burghard, M. Chemically Functionalized Quantum-Sized Carbon Dots for Bright and Colorful Photo-
Carbon Nanotubes. Small 2005, 1, 180−192. luminescence. J. Am. Chem. Soc. 2006, 128, 7756−7757.
(54) Yang, W.; Ratinac, K. R.; Ringer, S. R.; Thordarson, P.; (75) Zhu, S.; Song, Y.; Zhao, X.; Shao, J.; Zhang, J.; Yang, B. The
Gooding, J. J.; Braet, F. Carbon Nanomaterials in Biosensors: Should Photoluminescence Mechanism in Carbon Dots (Graphene Quantum
You Use Nanotubes or Graphene. Angew. Chem., Int. Ed. 2010, 49, Dots, Carbon Nanodots, and Polymer Dots): Current State and
2114−2138. Future Perspective. Nano Res. 2015, 8, 355−381.
(55) Kumar, M. A.; Jung, S.; Ji, T. Protein Biosensors Based on (76) Pecher, J.; Mecking, S. Nanoparticles of Conjugated Polymers.
Chem. Rev. 2010, 110, 6260−6279.
Polymer Nanowires, Carbon Nanotubes and Zinc Oxide Nanorods.
(77) Wu, C.; Chiu, D. T. Highly Fluorescent Semiconducting
Sensors 2011, 11, 5087−5111.
Polymer Dots for Biology and Medicine. Angew. Chem., Int. Ed. 2013,
(56) Jeyachandran, Y. L.; Mielczarski, J. A.; Mielczarski, E.; Rai, B.
52, 3086−3109.
Efficiency of Blocking of Non-Specific Interaction of Different
(78) Wang, Y.; Hu, A. Carbon Quantum Dots: Synthesis, Properties
Proteins by BSA Adsorbed on Hydrophobic and Hydrophilic
and Applications. J. Mater. Chem. C 2014, 2, 6921−6939.
Surfaces. J. Colloid Interface Sci. 2010, 341, 136−142. (79) Zhao, X.; Bagwe, R. P.; Tan, W. Development of Organic-Dye-
(57) Chen, S.; Li, L.; Zhao, C.; Zheng, J. Surface Hydration:
Doped Silica Nanoparticles in a Reverse Microemulsion. Adv. Mater.
Principles and Applications toward Low-Fouling/Nonfouling Bio- 2004, 16, 173−176.
materials. Polymer 2010, 51, 5283−5293. (80) Ow, H.; Larson, D. R.; Srivastava, M.; Baird, B. A.; Webb, W.
(58) Blaszykowski, C.; Sheikh, S.; Thompson, M. Surface Chemistry W.; Wiesner, U. Bright and Stable Core-Shell Fluorescent Silica
to Minimize Fouling from Blood-Based Fluids. Chem. Soc. Rev. 2012, Nanoparticles. Nano Lett. 2005, 5, 113−117.
41, 5599−5612. (81) Warner, J. H.; Hoshino, A.; Yamamoto, K.; Tilley, R. D. Water-
(59) Alcantar, N. A.; Aydil, E. S.; Israelachvili, J. N. Polyethylene Soluble Photoluminescent Silicon Quantum Dots. Angew. Chem., Int.
Glycol-Coated Biocompatible Surfaces. J. Biomed. Mater. Res. 2000, Ed. 2005, 44, 4550−4554.
51, 343−351. (82) Zanetti-Domingues, L. C.; Tynan, C. J.; Rolfe, D. J.; Clarke, D.
(60) Sharma, S.; Johnson, R. W.; Desai, T. A. Evaluation of the T.; Martin-Fernandez, M. Hydrophobic Fluorescent Probes Introduce
Stability of Nonfouling Ultrathin Poly(Ethylene Glycol) Films for Artifacts into Single Molecule Tracking Experiments Due to Non-
Silicon-Based Microdevices. Langmuir 2004, 20, 348−356. Specific Binding. PLoS One 2013, 8, e74200.
(61) Goode, J. A.; Rushworth, J. V. H.; Millner, P. A. Biosensor (83) Roda, A.; Mirasoli, M.; Michelini, E.; Di Fusco, M.; Zangheri,
Regeneration: A Review of Common Techniques and Outcomes. M.; Cevenini, L.; Roda, B.; Simoni, P. Progress in Chemical
Langmuir 2015, 31, 6267−6276. Luminescence-Based Biosensors: A Critical Review. Biosens. Bioelec-
(62) Zhou, Q.; Kwa, T.; Gao, Y.; Liu, Y.; Rahimian, A.; Revzin, A. tron. 2016, 76, 164−179.
On-Chip Regeneration of Aptasensors for Monitoring Cell Secretion. (84) Chen, H.; Lin, L.; Li, H.; Lin, J. M. Quantum Dots-Enhanced
Lab Chip 2014, 14, 276−279. Chemiluminescence: Mechanism and Application. Coord. Chem. Rev.
(63) Fan, X.; White, I. M.; Shopova, S. I.; Zhu, H.; Suter, J. D.; Sun, 2014, 263-264, 86−100.
Y. Sensitive Optical Biosensors for Unlabeled Targets: A Review. (85) Aslan, K.; Geddes, C. D. Metal-Enhanced Chemiluminescence:
Anal. Chim. Acta 2008, 620, 8−26. Advanced Chemiluminescence Concepts for the 21st Century. Chem.
(64) Hunt, H. K.; Armani, A. M. Label-Free Biological and Chemical Soc. Rev. 2009, 38, 2556−2564.
Sensors. Nanoscale 2010, 2, 1544−1559. (86) Bi, S.; Yan, Y.; Yang, X.; Zhang, S. Gold Nanolabels for New
(65) Tokel, O.; Inci, F.; Demirci, U. Advances in Plasmonic Enhanced Chemiluminescence Imniunoassay of Alpha-Fetoprotein
Technologies for Point of Care Applications. Chem. Rev. 2014, 114, Based on Magnetic Beads. Chem. - Eur. J. 2009, 15, 4704−4709.
5728−5752. (87) Zhang, Z. F.; Cui, H.; Lai, C. Z.; Liu, L. J. Gold Nanoparticle-
(66) Caucheteur, C.; Guo, T.; Albert, J. Review of Plasmonic Fiber Catalyzed Luminol Chemiluminescence and Its Analytical Applica-
Optic Biochemical Sensors: Improving the Limit of Detection. Anal. tions. Anal. Chem. 2005, 77, 3324−3329.
Bioanal. Chem. 2015, 407, 3883−3897. (88) Lin, Z.; Xue, W.; Chen, H.; Lin, J.-M. Classical Oxidant
(67) Bănică, F.-G. Chemical Sensors and Biosensors; 1st ed.; John Induced Chemiluminescence of Fluorescent Carbon Dots. Chem.
Wiley & Sons: West Sussex, UK, 2012. Commun. 2012, 48, 1051−1053.

315 DOI: 10.1021/acs.chemrev.8b00257


Chem. Rev. 2019, 119, 293−321
Chemical Reviews Review

(89) Martínez-Máñez, R.; Sancenón, F. Fluorogenic and Chromo- (111) Stoeva, S. I.; Lee, J. S.; Smith, J. E.; Rosen, S. T.; Mirkin, C. A.
genic Chemosensors and Reagents for Anions. Chem. Rev. 2003, 103, Multiplexed Detection of Protein Cancer Markers with Biobarcoded
4419−4476. Nanoparticle Probes. J. Am. Chem. Soc. 2006, 128, 8378−8379.
(90) Zhao, Y.; Zheng, Y.; Zhao, C.; You, J.; Qu, F. Hollow PDA-Au (112) Juncker, D.; Bergeron, S.; Laforte, V.; Li, H. Cross-Reactivity
Nanoparticles-Enabled Signal Amplification for Sensitive Non- in Antibody Microarrays and Multiplexed Sandwich Assays: Shedding
enzymatic Colorimetric Immunodetection of Carbohydrate Antigen Light on the Dark Side of Multiplexing. Curr. Opin. Chem. Biol. 2014,
125. Biosens. Bioelectron. 2015, 71, 200−206. 18, 29−37.
(91) Yu, R. J.; Ma, W.; Liu, X. Y.; Jin, H. Y.; Han, H. X.; Wang, H. (113) Gubala, V.; Klein, R.; Templeton, D. M.; Schwenk, M.
Y.; Tian, H.; Long, Y. T. Metal-Linked Immunosorbent Assay Immunodiagnostics and Immunosensor Design. Pure Appl. Chem.
(MeLISA): The Enzyme-Free Alternative to Elisa for Biomarker 2014, 86, 1539−1571.
Detection in Serum. Theranostics 2016, 6, 1732−1739. (114) Stenken, J. A.; Poschenrieder, A. J. Bioanalytical Chemistry of
(92) Gao, Z.; Xu, M.; Hou, L.; Chen, G.; Tang, D. Magnetic Bead- Cytokines - A Review. Anal. Chim. Acta 2015, 853, 95−115.
Based Reverse Colorimetric Immunoassay Strategy for Sensing (115) Fischer, S. K.; Joyce, A.; Spengler, M.; Yang, T.-Y.; Zhuang, Y.;
Biomolecules. Anal. Chem. 2013, 85, 6945−6952. Fjording, M. S.; Mikulskis, A. Emerging Technologies to Increase
(93) Gao, Z.; Hou, L.; Xu, M.; Tang, D. Enhanced Colorimetric Ligand Binding Assay Sensitivity. AAPS J. 2015, 17, 93−101.
Immunoassay Accompanying with Enzyme Cascade Amplification (116) Tighe, P. J.; Ryder, R. R.; Todd, I.; Fairclough, L. C. ELISA in
Strategy for Ultrasensitive Detection of Low-Abundance Protein. Sci. the Multiplex Era: Potentials and Pitfalls. Proteomics: Clin. Appl. 2015,
Rep. 2015, 4, 3996. 9, 406−422.
(94) Chen, C.; Liu, Y.; Zheng, Z.; Zhou, G.; Ji, X.; Wang, H.; He, Z. (117) Pierobon, M.; Wulfkuhle, J.; Liotta, L.; Petricoin, E.
A New Colorimetric Platform for Ultrasensitive Detection of Protein Application of Molecular Technologies for Phosphoproteomic
and Cancer Cells Based on the Assembly of Nucleic Acids and Analysis of Clinical Samples. Oncogene 2015, 34, 805−814.
Proteins. Anal. Chim. Acta 2015, 880, 1−7. (118) Kupcova Skalnikova, H.; Cizkova, J.; Cervenka, J.; Vodicka, P.
(95) Chen, S.; Svedendahl, M.; Van Duyne, R. P.; Käll, M. Plasmon- Advances in Proteomic Techniques for Cytokine Analysis: Focus on
Enhanced Colorimetric ELISA with Single Molecule Sensitivity. Nano Melanoma Research. Int. J. Mol. Sci. 2017, 18, 2697.
Lett. 2011, 11, 1826−1830. (119) Basu, A. S. Digital Assays Part II: Digital Protein and Cell
(96) Liu, D.; Yang, J.; Wang, H. F.; Wang, Z.; Huang, X.; Wang, Z.; Assays. SLAS Technol. 2017, 22, 387−405.
Niu, G.; Hight Walker, A. R.; Chen, X. Glucose Oxidase-Catalyzed (120) Zhang, Y.; Noji, H. Digital Bioassays: Theory, Applications,
Growth of Gold Nanoparticles Enables Quantitative Detection of and Perspectives. Anal. Chem. 2017, 89, 92−101.
Attomolar Cancer Biomarkers. Anal. Chem. 2014, 86, 5800−5806. (121) Smith, P. K.; Krohn, R. I.; Hermanson, G. T.; Mallia, A. K.;
(97) De La Rica, R.; Stevens, M. M. Plasmonic ELISA for the Gartner, F. H.; Provenzano, M. D.; Fujimoto, E. K.; Goeke, N. M.;
Ultrasensitive Detection of Disease Biomarkers with the Naked Eye. Olson, B. J.; Klenk, D. C. Measurement of Protein Using
Nat. Nanotechnol. 2012, 7, 821−824. Bicinchoninic Acid. Anal. Biochem. 1985, 150, 76−85.
(98) Labib, M.; Sargent, E. H.; Kelley, S. O. Electrochemical (122) Lowry, O. H.; Rosebrough, N. J.; Farr, A. L.; Randall, R. J.
Methods for the Analysis of Clinically Relevant Biomolecules. Chem. Protein Measurement with the Folin-Phenol Reagent. J. Biol. Chem.
Rev. 2016, 116, 9001−9090. 1951, 193, 265−275.
(99) Wen, W.; Yan, X.; Zhu, C.; Du, D.; Lin, Y. Recent Advances in (123) Bradford, M. M. A Rapid and Sensitive Method for the for the
Electrochemical Immunosensors. Anal. Chem. 2017, 89, 138−156. Quantitation of Microgram Quantities of Protein Utilizing the
(100) Munge, B. S.; Stracensky, T.; Gamez, K.; DiBiase, D.; Rusling, Principle of Protein Dye-Binding. Anal. Biochem. 1976, 72, 248−254.
J. F. Multiplex Immunosensor Arrays for Electrochemical Detection of (124) Jones, L. J.; Haugland, R. P.; Singer, V. L. Development and
Cancer Biomarker Proteins. Electroanalysis 2016, 28, 2644−2658. Characterization of the NanoOrange Protein Quantitation Assay: A
(101) Liotta, L. A.; Espina, V.; Mehta, A. I.; Calvert, V.; Rosenblatt, Fluorescence-Based Assay of Proteins in Solution. Biotechniques 2003,
K.; Geho, D.; Munson, P. J.; Young, L.; Wulfkuhle, J.; Petricoin, E. F. 34, 850−861.
Protein Microarrays: Meeting Analytical Challenges for Clinical (125) Noble, J. E.; Bailey, M. J. A. Chapter 8 Quantitation of
Applications. Cancer Cell 2003, 3, 317−325. Protein. Methods Enzymol. 2009, 463, 73−95.
(102) Kodadek, T. Protein Microarrays: Prospects and Problems. (126) Yalow, R. S.; Berson, S. A. Immunoassay of Endogenous
Chem. Biol. 2001, 8, 105−115. Plasma Insulin in Man. J. Clin. Invest. 1960, 39, 1157−1175.
(103) Leng, Y.; Sun, K.; Chen, X.; Li, W. Suspension Arrays Based (127) Lequin, R. M. Enzyme Immunoassay (EIA)/Enzyme-Linked
on Nanoparticle-Encoded Microspheres for High-Throughput Multi- Immunosorbent Assay (ELISA). Clin. Chem. 2005, 51, 2415−2418.
plexed Detection. Chem. Soc. Rev. 2015, 44, 5552−5595. (128) Engvall, E.; Perlmann, P. Enzyme-Linked Immunosorbent
(104) Su, X.; Zhang, J.; Sun, L.; Koo, T. W.; Chan, S.; Sundararajan, Assay (ELISA) Quantitative Assay of Immunoglobulin G. Immuno-
N.; Yamakawa, M.; Berlin, A. A. Composite Organic-Inorganic chemistry 1971, 8, 871−874.
Nanoparticles (COINs) with Chemically Encoded Optical Signatures. (129) Van Weemen, B. K.; Schuurs, A. H. W. M. Immunoassay
Nano Lett. 2005, 5, 49−54. Using Antigen-Enzyme Conjugates. FEBS Lett. 1971, 15, 232−236.
(105) Han, M.; Gao, X.; Su, J. Z.; Nie, S. Quantum-Dot-Tagged (130) Black, S.; Kushner, I.; Samols, D. C-Reactive Protein. J. Biol.
Microbeads for Multiplexed Optical Coding of Biomolecules. Nat. Chem. 2004, 279, 48487−48490.
Biotechnol. 2001, 19, 631−635. (131) Kim, E. Y.; Stanton, J.; Korber, B. T. M.; Krebs, K.; Bogdan,
(106) Vignali, D. A. A. Multiplexed Particle-Based Flow Cytometric D.; Kunstman, K.; Wu, S.; Phair, J. P.; Mirkin, C. A.; Wolinsky, S. M.
Assays. J. Immunol. Methods 2000, 243, 243−255. Detection of HIV-1 P24 Gag in Plasma by a Nanoparticle-Based Bio-
(107) Kellar, K. L.; Iannone, M. A. Multiplexed Microsphere-Based Barcode-Amplification Method. Nanomedicine 2008, 3, 293−303.
Flow Cytometric Assays. Exp. Hematol. 2002, 30, 1227−1237. (132) Czerkinsky, C. C.; Nilsson, L. A.; Nygren, H.; Ouchterlony,
(108) Wilson, R.; Cossins, A. R.; Spiller, D. G. Encoded O.; Tarkowski, A. A Solid-Phase Enzyme-Linked Immunospot
Microcarriers for High-Throughput Multiplexed Detection. Angew. (ELISPOT) Assay for Enumeration of Specific Antibody-Secreting
Chem., Int. Ed. 2006, 45, 6104−6117. Cells. J. Immunol. Methods 1983, 65, 109−121.
(109) Braeckmans, K.; De Smedt, S. C.; Leblans, M.; Pauwels, R.; (133) Rutledge, T. F.; Boyd, M. F.; Mazurek, M.; Jereb, J.; Vernon,
Demeester, J. Encoding Microcarriers: Present and Future Tech- A.; LoBue, P.; Goldberg, S.; Castro, K. Updated Guidelines for Using
nologies. Nat. Rev. Drug Discovery 2002, 1, 447−456. Interferon Gamma Release Assays to Detect Mycobacterium
(110) Shikha, S.; Salafi, T.; Cheng, J.; Zhang, Y. Versatile Design and Tuberculosis Infection-United States, 2010. MMWR Recomm. Rep.
Synthesis of Nano-Barcodes. Chem. Soc. Rev. 2017, 46, 7054−7093. 2010, 59 (RR05), 1−25.

316 DOI: 10.1021/acs.chemrev.8b00257


Chem. Rev. 2019, 119, 293−321
Chemical Reviews Review

(134) Renart, J.; Reiser, J.; Stark, G. R. Transfer of Proteins from (152) Greenwood, C.; Ruff, D.; Kirvell, S.; Johnson, G.; Dhillon, H.
Gels to Diazobenzyloxymethyl-Paper and Detection with Antisera: A S.; Bustin, S. A. Proximity Assays for Sensitive Quantification of
Method for Studying Antibody Specificity and Antigen Structure. Proteins. Biomol. Detect. Quantif. 2015, 4, 10−16.
Proc. Natl. Acad. Sci. U. S. A. 1979, 76, 3116−3120. (153) Sano, T.; Smith, C. L.; Cantor, C. R. Immuno-PCR: Very
(135) Towbin, H.; Staehelin, T.; Gordon, J. Electrophoretic Transfer Sensitive Antigen Detection by Means of Specific Antibody-DNA
of Proteins from Polyacrylamide Gels to Nitrocellulose Sheets: Conjugates. Science 1992, 258, 120−122.
Procedure and Some Applications. Proc. Natl. Acad. Sci. U. S. A. 1979, (154) Lind, K.; Kubista, M. Development and Evaluation of Three
76, 4350−4354. Real-Time Immuno-PCR Assemblages for Quantification of PSA. J.
(136) Burnette, W. N. Western Blotting”: Electrophoretic Transfer Immunol. Methods 2005, 304, 107−116.
of Proteins from Sodium Dodecyl Sulfate-Polyacrylamide Gels to (155) Fredriksson, S.; Gullberg, M.; Jarvius, J.; Olsson, C.; Pietras,
Unmodified Nitrocellulose and Radiographic Detection with Anti- K.; Gústafsdóttir, S. M.; Ö stman, A.; Landegren, U. Protein Detection
body and Radioiodinated Protein A. Anal. Biochem. 1981, 112, 195− Using Proximity-Dependent DNA Ligation Assays. Nat. Biotechnol.
203. 2002, 20, 473−477.
(137) Mahmood, T.; Yang, P. C. Western Blot: Technique, Theory, (156) Gullberg, M.; Gustafsdottir, S. M.; Schallmeiner, E.; Jarvius, J.;
and Trouble Shooting. N. Am. J. Med. Sci. 2012, 4, 429−434. Bjarnegard, M.; Betsholtz, C.; Landegren, U.; Fredriksson, S.
(138) Hecht, F. M.; Busch, M. P.; Rawal, B.; Webb, M.; Rosenberg, Cytokine Detection by Antibody-Based Proximity Ligation. Proc.
E.; Swanson, M.; Chesney, M.; Anderson, J.; Levy, J.; Kahn, J. O. Use Natl. Acad. Sci. U. S. A. 2004, 101, 8420−8424.
of Laboratory Tests and Clinical Symptoms for Identification of (157) Ke, R.; Nong, R. Y.; Fredriksson, S.; Landegren, U.; Nilsson,
Primary HIV Infection. AIDS 2002, 16, 1119−1129. M. Improving Precision of Proximity Ligation Assay by Amplified
(139) Iweala, O. I. HIV Diagnostic Tests: An Overview. Contra- Single Molecule Detection. PLoS One 2013, 8, e69813.
ception 2004, 70, 141−147. (158) Lundberg, M.; Eriksson, A.; Tran, B.; Assarsson, E.;
(140) Romanov, V.; Davidoff, S. N.; Miles, A. R.; Grainger, D. W.; Fredriksson, S. Homogeneous Antibody-Based Proximity Extension
Gale, B. K.; Brooks, B. D. A Critical Comparison of Protein Assays Provide Sensitive and Specific Detection of Low-Abundant
Microarray Fabrication Technologies. Analyst 2014, 139, 1303−1326. Proteins in Human Blood. Nucleic Acids Res. 2011, 39, e102.
(141) Zhu, H.; Qian, J. Applications of Functional Protein (159) Lundberg, M.; Thorsen, S. B.; Assarsson, E.; Villablanca, A.;
Microarrays in Basic and Clinical Research. Adv. Genet. 2012, 79, Tran, B.; Gee, N.; Knowles, M.; Nielsen, B. S.; González Couto, E.;
123−155. Martin, R.; et al. Multiplexed Homogeneous Proximity Ligation
(142) Zhu, H.; Klemic, J. F.; Chang, S.; Bertone, P.; Casamayor, A.; Assays for High-Throughput Protein Biomarker Research in
Klemic, K. G.; Smith, D.; Gerstein, M.; Reed, M. A.; Snyder, M.; et al. Serological Material. Mol. Cell. Proteomics 2011, 10, M110.004978.
Analysis of Yeast Protein Kinases Using Protein Chips. Nat. Genet. (160) Nam, J.-M.; Thaxton, C. S.; Mirkin, C. A. Nanoparticle-Based
Bio-Bar Codes for the Ultrasensitive Detection of Proteins. Science
2000, 26, 283−289.
(143) Zhu, H.; Bilgin, M.; Bangham, R.; Hall, D.; Casamayor, A.; 2003, 301, 1884−1886.
(161) Goluch, E. D.; Nam, J.-M.; Georganopoulou, D. G.; Chiesl, T.
Bertone, P.; Lan, N.; Jansen, R.; Bidlingmaier, S.; Houfek, T.; et al.
N.; Shaikh, K. A.; Ryu, K. S.; Barron, A. E.; Mirkin, C. A.; Liu, C. A
Global Analysis of Protein Activities Using Proteome Chips. Science
Bio-Barcode Assay for on-Chip Attomolar-Sensitivity Protein
2001, 293, 2101−2105.
Detection. Lab Chip 2006, 6, 1293−1299.
(144) Crompton, P. D.; Kayala, M. A.; Traore, B.; Kayentao, K.;
(162) Hansen, C. H.; Yang, D.; Koussa, M. A.; Wong, W. P.
Ongoiba, A.; Weiss, G. E.; Molina, D. M.; Burk, C. R.; Waisberg, M.;
Nanoswitch-Linked Immunosorbent Assay (NLISA) for Fast,
Jasinskas, A.; et al. A Prospective Analysis of the Ab Response to
Sensitive, and Specific Protein Detection. Proc. Natl. Acad. Sci. U. S.
Plasmodium Falciparum before and after a Malaria Season by Protein A. 2017, 114, 10367−10372.
Microarray. Proc. Natl. Acad. Sci. U. S. A. 2010, 107, 6958−6963. (163) Hindson, B. J.; Ness, K. D.; Masquelier, D. A.; Belgrader, P.;
(145) Hu, S.; Xie, Z.; Onishi, A.; Yu, X.; Jiang, L.; Lin, J.; Rho, H.-s.; Heredia, N. J.; Makarewicz, A. J.; Bright, I. J.; Lucero, M. Y.;
Woodard, C.; Wang, H.; Jeong, J. S.; et al. Profiling the Human Hiddessen, A. L.; Legler, T. C.; et al. High-Throughput Droplet
Protein-DNA Interactome Reveals ERK2 as a Transcriptional Digital PCR System for Absolute Quantitation of DNA Copy
Repressor of Interferon Signaling. Cell 2009, 139, 610−622. Number. Anal. Chem. 2011, 83, 8604−8610.
(146) Huang, J.; Zhu, H.; Haggarty, S. J.; Spring, D. R.; Hwang, H.; (164) Albayrak, C.; Jordi, C. A.; Zechner, C.; Lin, J.; Bichsel, C. A.;
Jin, F.; Snyder, M.; Schreiber, S. L. Finding New Components of the Khammash, M.; Tay, S. Digital Quantification of Proteins and MRNA
Target of Rapamycin (TOR) Signaling Network through Chemical in Single Mammalian Cells. Mol. Cell 2016, 61, 914−924.
Genetics and Proteome Chips. Proc. Natl. Acad. Sci. U. S. A. 2004, (165) Russell, C.; Roy, S.; Ganguly, S.; Qian, X.; Caruthers, M. H.;
101, 16594−16599. Nilsson, M. Formation of Silver Nanostructures by Rolling Circle
(147) Cretich, M.; Damin, F.; Chiari, M. Protein Microarray Amplification Using Boranephosphonate-Modified Nucleotides. Anal.
Technology: How Far off Is Routine Diagnostics? Analyst 2014, 139, Chem. 2015, 87, 6660−6666.
528−542. (166) Hill, H. D.; Mirkin, C. A. The Bio-Barcode Assay for the
(148) Hardy, R. R.; Carmack, C. E.; Shinton, S. A.; Kemp, J. D.; Detection of Protein and Nucleic Acid Targets Using DTT-Induced
Hayakawa, K. Resolution and Characterization of Pro-B and Pre-Pro- Ligand Exchange. Nat. Protoc. 2006, 1, 324−336.
B Cell Stages in Normal Mouse Bone Marrow. J. Exp. Med. 1991, 173, (167) Thaxton, C. S.; Georganopoulou, D. G.; Mirkin, C. A. Gold
1213−1225. Nanoparticle Probes for the Detection of Nucleic Acid Targets. Clin.
(149) Abraham, R. S.; Aubert, G. Flow Cytometry, a Versatile Tool Chim. Acta 2006, 363, 120−126.
for Diagnosis and Monitoring of Primary Immunodeficiencies. Clin. (168) Thaxton, C. S.; Elghanian, R.; Thomas, A. D.; Stoeva, S. I.;
Vaccine Immunol. 2016, 23, 254−271. Lee, J.-S.; Smith, N. D.; Schaeffer, A. J.; Klocker, H.; Horninger, W.;
(150) Hedley, B. D.; Cheng, G.; Luider, J.; Kern, W.; Lozanski, G.; Bartsch, G.; et al. Nanoparticle-Based Bio-Barcode Assay Redefines
Chin-Yee, I.; Lowes, L. E.; Keeney, M.; Careaga, D.; Magari, R.; et al. “Undetectable” PSA and Biochemical Recurrence after Radical
Initial Flow Cytometric Evaluation of the ClearLLab Lymphoid Prostatectomy. Proc. Natl. Acad. Sci. U. S. A. 2009, 106, 18437−
Screen. Cytometry, Part B 2017, DOI: 10.1002/cyto.b.21603. 18442.
(151) Tan, Y.; Guo, Q.; Zhao, X.; Yang, X.; Wang, K.; Huang, J.; (169) Eng, J. K.; McCormack, A. L.; Yates, J. R. An Approach to
Zhou, Y. Proximity-Dependent Protein Detection Based on Enzyme- Correlate Tandem Mass Spectral Data of Peptides with Amino Acid
Assisted Fluorescence Signal Amplification. Biosens. Bioelectron. 2014, Sequences in a Protein Database. J. Am. Soc. Mass Spectrom. 1994, 5,
51, 255−260. 976−989.

317 DOI: 10.1021/acs.chemrev.8b00257


Chem. Rev. 2019, 119, 293−321
Chemical Reviews Review

(170) Gillet, L. C.; Leitner, A.; Aebersold, R. Mass Spectrometry M.; et al. A Subcellular Map of the Human Proteome. Science 2017,
Applied to Bottom-Up Proteomics: Entering the High-Throughput 356, eaal3321.
Era for Hypothesis Testing. Annu. Rev. Anal. Chem. 2016, 9, 449−472. (192) Uhlen, M.; Oksvold, P.; Fagerberg, L.; Lundberg, E.; Jonasson,
(171) Catherman, A. D.; Skinner, O. S.; Kelleher, N. L. Top Down K.; Forsberg, M.; Zwahlen, M.; Kampf, C.; Wester, K.; Hober, S.;
Proteomics: Facts and Perspectives. Biochem. Biophys. Res. Commun. et al. Towards a Knowledge-Based Human Protein Atlas. Nat.
2014, 445, 683−693. Biotechnol. 2010, 28, 1248−1250.
(172) Aebersold, R.; Mann, M. Mass-Spectrometric Exploration of (193) Uhlen, M. A Human Protein Atlas for Normal and Cancer
Proteome Structure and Function. Nature 2016, 537, 347−355. Tissues Based on Antibody Proteomics. Mol. Cell. Proteomics 2005, 4,
(173) Bantscheff, M.; Lemeer, S.; Savitski, M. M.; Kuster, B. 1920−1932.
Quantitative Mass Spectrometry in Proteomics: Critical Review (194) Ettinger, A.; Wittmann, T. Fluorescence Live Cell Imaging.
Update from 2007 to the Present. Anal. Bioanal. Chem. 2012, 404, Methods Cell Biol. 2014, 123, 77−94.
939−965. (195) Huang, B.; Bates, M.; Zhuang, X. Super-Resolution
(174) Koczula, K. M.; Gallotta, A. Lateral Flow Assays. Essays Fluorescence Microscopy. Annu. Rev. Biochem. 2009, 78, 993−1016.
Biochem. 2016, 60, 111−120. (196) Huang, B.; Babcock, H.; Zhuang, X. Breaking the Diffraction
(175) Bahadır, E. B.; Sezgintürk, M. K. Lateral Flow Assays: Barrier: Super-Resolution Imaging of Cells. Cell 2010, 143, 1047−
Principles, Designs and Labels. TrAC, Trends Anal. Chem. 2016, 82, 1058.
286−306. (197) Patterson, G.; Davidson, M.; Manley, S.; Lippincott-Schwartz,
(176) Posthuma-Trumpie, G. A.; Korf, J.; Van Amerongen, A. J. Superresolution Imaging Using Single-Molecule Localization. Annu.
Lateral Flow (Immuno)Assay: Its Strengths, Weaknesses, Oppor- Rev. Phys. Chem. 2010, 61, 345−367.
tunities and Threats. A Literature Survey. Anal. Bioanal. Chem. 2009, (198) Hauser, M.; Wojcik, M.; Kim, D.; Mahmoudi, M.; Li, W.; Xu,
393, 569−582. K. Correlative Super-Resolution Microscopy: New Dimensions and
(177) Assadollahi, S.; Reininger, C.; Palkovits, R.; Pointl, P.; New Opportunities. Chem. Rev. 2017, 117, 7428−7456.
Schalkhammer, T. From Lateral Flow Devices to a Novel Nano-Color (199) Taniguchi, Y.; Choi, P. J.; Li, G. W.; Chen, H.; Babu, M.;
Microfluidic Assay. Sensors 2009, 9, 6084−6100. Hearn, J.; Emili, A.; Xie, X. S. Quantifying E. Coli Proteome and
(178) Masson, J. F. Surface Plasmon Resonance Clinical Biosensors Transcriptome with Single-Molecule Sensitivity in Single Cells. Science
for Medical Diagnostics. ACS Sensors 2017, 2, 16−30. 2010, 329, 533−538.
(179) Brolo, A. G. Plasmonics for Future Biosensors. Nat. Photonics (200) Frei, A. P.; Bava, F. A.; Zunder, E. R.; Hsieh, E. W. Y.; Chen,
2012, 6, 709−713. S. Y.; Nolan, G. P.; Gherardini, P. F. Highly Multiplexed Simultaneous
(180) Li, M.; Cushing, S. K.; Wu, N. Plasmon-Enhanced Optical Detection of RNAs and Proteins in Single Cells. Nat. Methods 2016,
Sensors: A Review. Analyst 2015, 140, 386−406. 13, 269−275.
(181) Homola, J. Surface Plasmon Resonance Sensors for Detection (201) Darmanis, S.; Gallant, C. J.; Marinescu, V. D.; Niklasson, M.;
of Chemical and Biological Species. Chem. Rev. 2008, 108, 462−493. Segerman, A.; Flamourakis, G.; Fredriksson, S.; Assarsson, E.;
(182) El-Sayed, I. H.; Huang, X.; El-Sayed, M. A. Surface Plasmon Lundberg, M.; Nelander, S.; et al. Simultaneous Multiplexed
Resonance Scattering and Absorption of Anti-EGFR Antibody Measurement of RNA and Proteins in Single Cells. Cell Rep. 2016,
Conjugated Gold Nanoparticles in Cancer Diagnostics: Applications 14, 380−389.
in Oral Cancer. Nano Lett. 2005, 5, 829−834. (202) Anderson, N. L.; Anderson, N. G. The Human Plasma
(183) Im, H.; Shao, H.; Park, Y. I.; Peterson, V. M.; Castro, C. M.; Proteome: History, Character, and Diagnostic Prospects. Mol. Cell.
Weissleder, R.; Lee, H. Label-Free Detection and Molecular Profiling Proteomics 2002, 1, 845−867.
of Exosomes with a Nano-Plasmonic Sensor. Nat. Biotechnol. 2014, (203) Ponomarenko, E. A.; Poverennaya, E. V.; Ilgisonis, E. V.;
32, 490−495. Pyatnitskiy, M. A.; Kopylov, A. T.; Zgoda, V. G.; Lisitsa, A. V.;
(184) Wark, A. W.; Lee, H. J.; Qavi, A. J.; Corn, R. M. Nanoparticle- Archakov, A. I. The Size of the Human Proteome: The Width and
Enhanced Diffraction Gratings for Ultrasensitive Surface Plasmon Depth. Int. J. Anal. Chem. 2016, 2016, 1−6.
Biosensing. Anal. Chem. 2007, 79, 6697−6701. (204) Armbruster, D. A.; Pry, T. Limit of Blank, Limit of Detection
(185) Sepúlveda, B.; Angelomé, P. C.; Lechuga, L. M.; Liz-Marzán, and Limit of Quantitation. Clin. Biochem. Rev. 2008, 29, S49−S52.
L. M. LSPR-Based Nanobiosensors. Nano Today 2009, 4, 244−251. (205) Holstein, C. A.; Griffin, M.; Hong, J.; Sampson, P. D.
(186) Cao, C.; Sim, S. J. Signal Enhancement of Surface Plasmon Statistical Method for Determining and Comparing Limits of
Resonance Immunoassay Using Enzyme Precipitation-Functionalized Detection of Bioassays. Anal. Chem. 2015, 87, 9795−9801.
Gold Nanoparticles: A Femto Molar Level Measurement of Anti- (206) Wild, D. The Immunoassay Handbook: Theory and Applications
Glutamic Acid Decarboxylase Antibody. Biosens. Bioelectron. 2007, 22, of Ligand Binding, ELISA and Related Techniques, 4th ed.; Elsevier:
1874−1880. Amsterdam, The Netherlands, 2013.
(187) Uludag, Y.; Tothill, I. E. Cancer Biomarker Detection in (207) Guglielmo-Viret, V.; Thullier, P. Comparison of an Electro-
Serum Samples Using Surface Plasmon Resonance and Quartz Crystal chemiluminescence Assay in Plate Format over a Colorimetric ELISA,
Microbalance Sensors with Nanoparticle Signal Amplification. Anal. for the Detection of Ricin B Chain (RCA-B). J. Immunol. Methods
Chem. 2012, 84, 5898−5904. 2007, 328, 70−78.
(188) Coons, A. H.; Creech, H. J.; Jones, R. N. Immunological (208) Richter, M. M. Electrochemiluminescence (ECL). Chem. Rev.
Properties of an Antibody Containing a Fluorescent Group. Exp. Biol. 2004, 104, 3003−3036.
Med. 1941, 47, 200−202. (209) Miao, W. Electrogenerated Chemiluminescence and Its
(189) Ramos-Vara, J. A.; Miller, M. A. When Tissue Antigens and Biorelated Applications. Chem. Rev. 2008, 108, 2506−2553.
Antibodies Get Along: Revisiting the Technical Aspects of (210) Forster, R. J.; Bertoncello, P.; Keyes, T. E. Electrogenerated
Immunohistochemistry-The Red, Brown, and Blue Technique. Vet. Chemiluminescence. Annu. Rev. Anal. Chem. 2009, 2, 359−385.
Pathol. 2014, 51, 42−87. (211) Hu, L.; Xu, G. Applications and Trends in Electro-
(190) Shi, S. R.; Key, M. E.; Kalra, K. L. Antigen Retrieval in chemiluminescence. Chem. Soc. Rev. 2010, 39, 3275−3304.
Formalin-Fixed, Paraffin-Embedded Tissues: An Enhancement (212) Huang, H.; Li, J.; Zhu, J.-J. Electrochemiluminescence Based
Method for Immunohistochemical Staining Based on Microwave on Quantum Dots and Their Analytical Application. Anal. Methods
Oven Heating of Tissue Sections. J. Histochem. Cytochem. 1991, 39, 2011, 3, 33−42.
741−748. (213) Xu, Y.; Liu, J.; Gao, C.; Wang, E. Applications of Carbon
(191) Thul, P. J.; Åkesson, L.; Wiking, M.; Mahdessian, D.; Quantum Dots in Electrochemiluminescence: A Mini Review.
Geladaki, A.; Ait Blal, H.; Alm, T.; Asplund, A.; Björk, L.; Breckels, L. Electrochem. Commun. 2014, 48, 151−154.

318 DOI: 10.1021/acs.chemrev.8b00257


Chem. Rev. 2019, 119, 293−321
Chemical Reviews Review

(214) Muzyka, K. Current Trends in the Development of the Tumour Cells Using Microfluidic Western Blotting. Nat. Commun.
Electrochemiluminescent Immunosensors. Biosens. Bioelectron. 2014, 2017, 8, 14622.
54, 393−407. (233) Hughes, A. J.; Spelke, D. P.; Xu, Z.; Kang, C.-C.; Schaffer, D.
(215) Liu, Z.; Qi, W.; Xu, G. Recent Advances in Electro- V.; Herr, A. E. Single-Cell Western Blotting. Nat. Methods 2014, 11,
chemiluminescence. Chem. Soc. Rev. 2015, 44, 3117−3142. 749−755.
(216) Li, L.; Chen, Y.; Zhu, J. J. Recent Advances in Electro- (234) Vaught, J. D.; Bock, C.; Carter, J.; Fitzwater, T.; Otis, M.;
chemiluminescence Analysis. Anal. Chem. 2017, 89, 358−371. Schneider, D.; Rolando, J.; Waugh, S.; Wilcox, S. K.; Eaton, B. E.
(217) Dabitao, D.; Margolick, J. B.; Lopez, J.; Bream, J. H. Multiplex Expanding the Chemistry of DNA for in Vitro Selection. J. Am. Chem.
Measurement of Proinflammatory Cytokines in Human Serum: Soc. 2010, 132, 4141−4151.
Comparison of the Meso Scale Discovery Electrochemiluminescence (235) Davies, D. R.; Gelinas, A. D.; Zhang, C.; Rohloff, J. C.; Carter,
Assay and the Cytometric Bead Array. J. Immunol. Methods 2011, 372, J. D.; O’Connell, D.; Waugh, S. M.; Wolk, S. K.; Mayfield, W. S.;
71−77. Burgin, A. B.; et al. Unique Motifs and Hydrophobic Interactions
(218) Cohen, L.; Walt, D. R. Single-Molecule Arrays for Protein and Shape the Binding of Modified DNA Ligands to Protein Targets. Proc.
Nucleic Acid Analysis. Annu. Rev. Anal. Chem. 2017, 10, 345−363. Natl. Acad. Sci. U. S. A. 2012, 109, 19971−19976.
(219) Rissin, D. M.; Kan, C. W.; Campbell, T. G.; Howes, S. C.; (236) Rohloff, J. C.; Gelinas, A. D.; Jarvis, T. C.; Ochsner, U. A.;
Fournier, D. R.; Song, L.; Piech, T.; Patel, P. P.; Chang, L.; Rivnak, A. Schneider, D. J.; Gold, L.; Janjic, N. Nucleic Acid Ligands with
J.; et al. Single-Molecule Enzyme-Linked Immunosorbent Assay Protein-like Side Chains: Modified Aptamers and Their Use as
Detects Serum Proteins at Subfemtomolar Concentrations. Nat. Diagnostic and Therapeutic Agents. Mol. Ther.–Nucleic Acids 2014, 3,
Biotechnol. 2010, 28, 595−599. e201.
(220) Rissin, D. M.; Walt, D. R. Digital Concentration Readout of (237) Gold, L.; Ayers, D.; Bertino, J.; Bock, C.; Bock, A.; Brody, E.
Single Enzyme Molecules Using Femtoliter Arrays and Poisson N.; Carter, J.; Dalby, A. B.; Eaton, B. E.; Fitzwater, T.; et al. Aptamer-
Statistics. Nano Lett. 2006, 6, 520−523. Based Multiplexed Proteomic Technology for Biomarker Discovery.
(221) Rissin, D. M.; Fournier, D. R.; Piech, T.; Kan, C. W.; PLoS One 2010, 5, e15004.
Campbell, T. G.; Song, L.; Chang, L.; Rivnak, A. J.; Patel, P. P.; (238) Gold, L.; Walker, J. J.; Wilcox, S. K.; Williams, S. Advances in
Provuncher, G. K.; et al. Simultaneous Detection of Single Molecules Human Proteomics at High Scale with the SOMAscan Proteomics
and Singulated Ensembles of Molecules Enables Immunoassays with Platform. New Biotechnol. 2012, 29, 543−549.
Broad Dynamic Range. Anal. Chem. 2011, 83, 2279−2285. (239) Candia, J.; Cheung, F.; Kotliarov, Y.; Fantoni, G.; Sellers, B.;
(222) Rivnak, A. J.; Rissin, D. M.; Kan, C. W.; Song, L.; Fishburn, M. Griesman, T.; Huang, J.; Stuccio, S.; Zingone, A.; Ryan, B. M.; et al.
W.; Piech, T.; Campbell, T. G.; DuPont, D. R.; Gardel, M.; Sullivan, Assessment of Variability in the SOMAscan Assay. Sci. Rep. 2017, 7,
S.; et al. A Fully-Automated, Six-Plex Single Molecule Immunoassay
14248.
for Measuring Cytokines in Blood. J. Immunol. Methods 2015, 424, (240) Dunbar, S. A.; Hoffmeyer, M. R. Microsphere-Based Multiplex
20−27.
Immunoassays: Development and Applications Using Luminex®
(223) Shahim, P.; Tegner, Y.; Wilson, D. H.; Randall, J.; Skillbäck,
XMAP® Technology. In The Immunoassay Handbook; Elsevier:
T.; Pazooki, D.; Kallberg, B.; Blennow, K.; Zetterberg, H. Blood
Amsterdam, The Netherlands, 2013; Chapter 2.9, pp 157−174.
Biomarkers for Brain Injury in Concussed Professional Ice Hockey
(241) Morgan, E.; Varro, R.; Sepulveda, H.; Ember, J. A.; Apgar, J.;
Players. JAMA Neurol. 2014, 71, 684−692.
Wilson, J.; Lowe, L.; Chen, R.; Shivraj, L.; Agadir, A. Cytometric Bead
(224) Jarolim, P.; Patel, P. P.; Conrad, M. J.; Chang, L.; Melenovsky,
Array: A Multiplexed Assay Platform with Applications in Various
V.; Wilson, D. H. Fully Automated Ultrasensitive Digital Immuno-
assay for Cardiac Troponin i Based on Single Molecule Array Areas of Biology. Clin. Immunol. 2004, 110, 252−266.
(242) Elshal, M. F.; McCoy, J. P. Multiplex Bead Array Assays:
Technology. Clin. Chem. 2015, 61, 1283−1291.
(225) Schubert, S. M.; Walter, S. R.; Manesse, M.; Walt, D. R. Performance Evaluation and Comparison of Sensitivity to ELISA.
Protein Counting in Single Cancer Cells. Anal. Chem. 2016, 88, Methods 2006, 38, 317−323.
2952−2957. (243) Bandura, D. R.; Baranov, V. I.; Ornatsky, O. I.; Antonov, A.;
(226) Schubert, S. M.; Arendt, L. M.; Zhou, W.; Baig, S.; Walter, S. Kinach, R.; Lou, X.; Pavlov, S.; Vorobiev, S.; Dick, J. E.; Tanner, S. D.
R.; Buchsbaum, R. J.; Kuperwasser, C.; Walt, D. R. Ultra-Sensitive Mass Cytometry: Technique for Real Time Single Cell Multitarget
Protein Detection via Single Molecule Arrays towards Early Stage Immunoassay Based on Inductively Coupled Plasma Time-of-Flight
Cancer Monitoring. Sci. Rep. 2015, 5, 11034. Mass Spectrometry. Anal. Chem. 2009, 81, 6813−6822.
(227) Song, L.; Hanlon, D. W.; Chang, L.; Provuncher, G. K.; Kan, (244) Bendall, S. C.; Nolan, G. P.; Roederer, M.; Chattopadhyay, P.
C. W.; Campbell, T. G.; Fournier, D. R.; Ferrell, E. P.; Rivnak, A. J.; K. A Deep Profiler’s Guide to Cytometry. Trends Immunol. 2012, 33,
Pink, B. A.; et al. Single Molecule Measurements of Tumor Necrosis 323−332.
Factor Alpha and Interleukin-6 in the Plasma of Patients with Crohn’s (245) Spitzer, M. H.; Nolan, G. P. Mass Cytometry: Single Cells,
Disease. J. Immunol. Methods 2011, 372, 177−186. Many Features. Cell 2016, 165, 780−791.
(228) Wu, A. H. B.; Fukushima, N.; Puskas, R.; Todd, J.; Goix, P. (246) Bendall, S. C.; Simonds, E. F.; Qiu, P.; Amir, E. D.; Krutzik, P.
Development and Preliminary Clinical Validation of a High Sensitivity O.; Finck, R.; Bruggner, R. V.; Melamed, R.; Trejo, A.; Ornatsky, O.
Assay for Cardiac Troponin Using a Capillary Flow (Single Molecule) I.; et al. Single-Cell Mass Cytometry of Differential Immune and Drug
Fluorescence Detector. Clin. Chem. 2006, 52, 2157−2159. Responses across a Human Hematopoietic Continuum. Science 2011,
(229) Todd, J.; Freese, B.; Lu, A.; Held, D.; Morey, J.; Livingston, 332, 687−696.
R.; Goix, P. Ultrasensitive Flow-Based Immunoassays Using Single- (247) Corneau, A.; Cosma, A.; Even, S.; Katlama, C.; Le Grand, R.;
Molecule Counting. Clin. Chem. 2007, 53, 1990−1995. Frachet, V.; Blanc, C.; Autran, B. Comprehensive Mass Cytometry
(230) Gilbert, M.; Livingston, R.; Felberg, J.; Bishop, J. J. Multiplex Analysis of Cell Cycle, Activation, and Coinhibitory Receptors
Single Molecule Counting Technology Used to Generate Interleukin Expression in CD4 T Cells from Healthy and HIV-Infected
4, Interleukin 6, and Interleukin 10 Reference Limits. Anal. Biochem. Individuals. Cytometry, Part B 2017, 92, 21−32.
2016, 503, 11−20. (248) Giesen, C.; Wang, H. A. O.; Schapiro, D.; Zivanovic, N.;
(231) Hughes, A. J.; Spelke, D. P.; Xu, Z.; Kang, C.-C.; Schaffer, D. Jacobs, A.; Hattendorf, B.; Schüffler, P. J.; Grolimund, D.; Buhmann,
V.; Herr, A. E. Single-Cell Western Blotting. Nat. Methods 2014, 11, J. M.; Brandt, S.; et al. Highly Multiplexed Imaging of Tumor Tissues
749−755. with Subcellular Resolution by Mass Cytometry. Nat. Methods 2014,
(232) Sinkala, E.; Sollier-Christen, E.; Renier, C.; Rosàs-Canyelles, 11, 417−422.
E.; Che, J.; Heirich, K.; Duncombe, T. A.; Vlassakis, J.; Yamauchi, K. (249) Angelo, M.; Bendall, S. C.; Finck, R.; Hale, M. B.; Hitzman,
A.; Huang, H.; et al. Profiling Protein Expression in Circulating C.; Borowsky, A. D.; Levenson, R. M.; Lowe, J. B.; Liu, S. D.; Zhao,

319 DOI: 10.1021/acs.chemrev.8b00257


Chem. Rev. 2019, 119, 293−321
Chemical Reviews Review

S.; et al. Multiplexed Ion Beam Imaging of Human Breast Tumors. Enhanced Amplification Using Horseradish Peroxidase Induced
Nat. Med. 2014, 20, 436−442. Biocatalytic Precipitation on a CdS Quantum Dots Multilayer
(250) Auzel, F. Upconversion and Anti-Stokes Processes with f and Electrode. Anal. Chem. 2012, 84, 917−923.
d Ions in Solids. Chem. Rev. 2004, 104, 139−173. (272) Zhang, Y.; Ge, L.; Li, M.; Yan, M.; Ge, S.; Yu, J.; Song, X.;
(251) Vetrone, F.; Naccache, R.; Mahalingam, V.; Morgan, C. G.; Cao, B. Flexible Paper-Based ZnO Nanorod Light-Emitting Diodes
Capobianco, J. A. The Active-Core/Active-Shell Approach: A Strategy Induced Multiplexed Photoelectrochemical Immunoassay. Chem.
to Enhance the Upconversion Luminescence in Lanthanide-Doped Commun. 2014, 50, 1417−1419.
Nanoparticles. Adv. Funct. Mater. 2009, 19, 2924−2929. (273) Zhang, N.; Ma, Z. Y.; Ruan, Y. F.; Zhao, W. W.; Xu, J. J.;
(252) Zhang, F.; Shi, Q.; Zhang, Y.; Shi, Y.; Ding, K.; Zhao, D.; Chen, H. Y. Simultaneous Photoelectrochemical Immunoassay of
Stucky, G. D. Fluorescence Upconversion Microbarcodes for Dual Cardiac Markers Using Specific Enzyme Tags: A Proof of
Multiplexed Biological Detection: Nucleic Acid Encoding. Adv. Principle for Multiplexed Bioanalysis. Anal. Chem. 2016, 88, 1990−
Mater. 2011, 23, 3775−3779. 1994.
(253) Gorris, H. H.; Wolfbeis, O. S. Photon-Upconverting (274) Wang, P.; Ge, L.; Ge, S.; Yu, J.; Yan, M.; Huang, J. A Paper-
Nanoparticles for Optical Encoding and Multiplexing of Cells, Based Photoelectrochemical Immunoassay for Low-Cost and Multi-
Biomolecules, and Microspheres. Angew. Chem., Int. Ed. 2013, 52, plexed Point-of-Care Testing. Chem. Commun. 2013, 49, 3294−3296.
3584−3600. (275) Vollmer, F.; Arnold, S. Whispering-Gallery-Mode Biosensing:
(254) Huang, K.; Idris, N. M.; Zhang, Y. Engineering of Lanthanide- Label-Free Detection down to Single Molecules. Nat. Methods 2008,
Doped Upconversion Nanoparticles for Optical Encoding. Small 5, 591−596.
2016, 12, 836−852. (276) Baaske, M. D.; Foreman, M. R.; Vollmer, F. Single-Molecule
(255) Wang, F.; Liu, X. Recent Advances in the Chemistry of Nucleic Acid Interactions Monitored on a Label-Free Microcavity
Lanthanide-Doped Upconversion Nanocrystals. Chem. Soc. Rev. 2009, Biosensor Platform. Nat. Nanotechnol. 2014, 9, 933−939.
38, 976−989. (277) Armani, A. M.; Kulkarni, R. P.; Fraser, S. E.; Flagan, R. C.;
(256) Haase, M.; Schäfer, H. Upconverting Nanoparticles. Angew. Vahala, K. J. Label-Free, Single-Molecule Detection with Optical
Chem., Int. Ed. 2011, 50, 5808−5829. Microcavities. Science 2007, 317, 783−787.
(257) Bouzigues, C.; Gacoin, T.; Alexandrou, A. Biological (278) Kindt, J. T.; Luchansky, M. S.; Qavi, A. J.; Lee, S. H.; Bailey,
Applications of Rare-Earth Based Nanoparticles. ACS Nano 2011, 5, R. C. Subpicogram per Milliliter Detection of Interleukins Using
8488−8505. Silicon Photonic Microring Resonators and an Enzymatic Signal
(258) Gai, S.; Li, C.; Yang, P.; Lin, J. Recent Progress in Rare Earth Enhancement Strategy. Anal. Chem. 2013, 85, 10653−10657.
Micro/Nanocrystals: Soft Chemical Synthesis, Luminescent Proper- (279) Washburn, A. L.; Luchansky, M. S.; Bowman, A. L.; Bailey, R.
ties, and Biomedical Applications. Chem. Rev. 2014, 114, 2343−2389. C. Quantitative, Label-Free Detection of Five Protein Biomarkers
(259) Li, X.; Zhang, F.; Zhao, D. Lab on Upconversion
Using Multiplexed Arrays of Silicon Photonic Microring Resonators.
Nanoparticles: Optical Properties and Applications Engineering via
Anal. Chem. 2010, 82, 69−72.
Designed Nanostructure. Chem. Soc. Rev. 2015, 44, 1346−1378.
(280) Foreman, M. R.; Swaim, J. D.; Vollmer, F. Whispering Gallery
(260) Sedlmeier, A.; Gorris, H. H. Surface Modification and
Mode Sensors. Adv. Opt. Photonics 2015, 7, 168−240.
Characterization of Photon-Upconverting Nanoparticles for Bio-
(281) Kim, E.; Baaske, M. D.; Vollmer, F. Towards Next-Generation
analytical Applications. Chem. Soc. Rev. 2015, 44, 1526−1560.
Label-Free Biosensors: Recent Advances in Whispering Gallery Mode
(261) Wu, X.; Chen, G.; Shen, J.; Li, Z.; Zhang, Y.; Han, G.
Sensors. Lab Chip 2017, 17, 1190−1205.
Upconversion Nanoparticles: A Versatile Solution to Multiscale
(282) Yang, S.; Dai, X.; Stogin, B. B.; Wong, T.-S. Ultrasensitive
Biological Imaging. Bioconjugate Chem. 2015, 26, 166−175.
(262) Chen, G.; Qiu, H.; Prasad, P. N.; Chen, X. Upconversion Surface-Enhanced Raman Scattering Detection in Common Fluids.
Nanoparticles: Design, Nanochemistry, and Applications in Thera- Proc. Natl. Acad. Sci. U. S. A. 2016, 113, 268−273.
nostics. Chem. Rev. 2014, 114, 5161−5214. (283) Qian, X.; Peng, X. H.; Ansari, D. O.; Yin-Goen, Q.; Chen, G.
(263) Grätzel, M. Photoelectrochemical Cells. Nature 2001, 414, Z.; Shin, D. M.; Yang, L.; Young, A. N.; Wang, M. D.; Nie, S. In Vivo
338−344. Tumor Targeting and Spectroscopic Detection with Surface-
(264) Zhao, W. W.; Xu, J. J.; Chen, H. Y. Photoelectrochemical Enhanced Raman Nanoparticle Tags. Nat. Biotechnol. 2008, 26, 83−
DNA Biosensors. Chem. Rev. 2014, 114, 7421−7441. 90.
(265) Zhao, W.-W.; Xu, J.-J.; Chen, H.-Y. Photoelectrochemical (284) Liu, B.; Zhang, D.; Ni, H.; Wang, D.; Jiang, L.; Fu, D.; Han,
Bioanalysis: The State of the Art. Chem. Soc. Rev. 2015, 44, 729−741. X.; Zhang, C.; Chen, H.; Gu, Z.; et al. Multiplex Analysis on a Single
(266) Zhao, W. W.; Xu, J. J.; Chen, H. Y. Photoelectrochemical Porous Hydrogel Bead with Encoded SERS Nanotags. ACS Appl.
Immunoassays. Anal. Chem. 2018, 90, 615−627. Mater. Interfaces 2018, 10, 21−26.
(267) Yue, Z.; Lisdat, F.; Parak, W. J.; Hickey, S. G.; Tu, L.; Sabir, (285) Matteini, P.; Cottat, M.; Tavanti, F.; Panfilova, E.; Scuderi,
N.; Dorfs, D.; Bigall, N. C. Quantum-Dot-Based Photoelectrochem- M.; Nicotra, G.; Menziani, M. C.; Khlebtsov, N.; De Angelis, M.; Pini,
ical Sensors for Chemical and Biological Detection. ACS Appl. Mater. R. Site-Selective Surface-Enhanced Raman Detection of Proteins. ACS
Interfaces 2013, 5, 2800−2814. Nano 2017, 11, 918−926.
(268) Xiao, F. X.; Miao, J.; Liu, B. Layer-by-Layer Self-Assembly of (286) Liu, B.; Ni, H.; Zhang, D.; Wang, D.; Fu, D.; Chen, H.; Gu,
CdS Quantum Dots/Graphene Nanosheets Hybrid Films for Z.; Zhao, X. Ultrasensitive Detection of Protein with Wide Linear
Photoelectrochemical and Photocatalytic Applications. J. Am. Chem. Dynamic Range Based on Core-Shell SERS Nanotags and Photonic
Soc. 2014, 136, 1559−1569. Crystal Beads. ACS Sensors 2017, 2, 1035−1043.
(269) Haddour, N.; Chauvin, J.; Gondran, C.; Cosnier, S. (287) Zhang, D.; Huang, L.; Liu, B.; Ni, H.; Sun, L.; Su, E.; Chen,
Photoelectrochemical Immunosensor for Label-Free Detection and H.; Gu, Z.; Zhao, X. Quantitative and Ultrasensitive Detection of
Quantification of Anti-Cholera Toxin Antibody. J. Am. Chem. Soc. Multiplex Cardiac Biomarkers in Lateral Flow Assay with Core-Shell
2006, 128, 9693−9698. SERS Nanotags. Biosens. Bioelectron. 2018, 106, 204−211.
(270) Fan, G. C.; Zhu, H.; Du, D.; Zhang, J. R.; Zhu, J. J.; Lin, Y. (288) Wang, Z.; Zong, S.; Wu, L.; Zhu, D.; Cui, Y. SERS-Activated
Enhanced Photoelectrochemical Immunosensing Platform Based on Platforms for Immunoassay: Probes, Encoding Methods, and
CdSeTe@CdS:Mn Core-Shell Quantum Dots-Sensitized TiO2Am- Applications. Chem. Rev. 2017, 117, 7910−7963.
plified by CuS Nanocrystals Conjugated Signal Antibodies. Anal. (289) Wang, Y.; Yan, B.; Chen, L. SERS Tags: Novel Optical
Chem. 2016, 88, 3392−3399. Nanoprobes for Bioanalysis. Chem. Rev. 2013, 113, 1391−1428.
(271) Zhao, W. W.; Ma, Z. Y.; Yu, P. P.; Dong, X. Y.; Xu, J. J.; Chen, (290) Bantz, K. C.; Meyer, A. F.; Wittenberg, N. J.; Im, H.; Kurtuluş,
H. Y. Highly Sensitive Photoelectrochemical Immunoassay with Ö .; Lee, S. H.; Lindquist, N. C.; Oh, S.-H.; Haynes, C. L. Recent

320 DOI: 10.1021/acs.chemrev.8b00257


Chem. Rev. 2019, 119, 293−321
Chemical Reviews Review

Progress in SERS Biosensing. Phys. Chem. Chem. Phys. 2011, 13, (311) Watanabe, R.; Soga, N.; Fujita, D.; Tabata, K. V.; Yamauchi,
11551. L.; Hyeon Kim, S.; Asanuma, D.; Kamiya, M.; Urano, Y.; Suga, H.;
(291) Chao, J.; Cao, W.; Su, S.; Weng, L.; Song, S.; Fan, C.; Wang, et al. Arrayed Lipid Bilayer Chambers Allow Single-Molecule Analysis
L. Nanostructure-Based Surface-Enhanced Raman Scattering Bio- of Membrane Transporter Activity. Nat. Commun. 2014, 5, 4519.
sensors for Nucleic Acids and Proteins. J. Mater. Chem. B 2016, 4, (312) Rosen, C. B.; Rodriguez-Larrea, D.; Bayley, H. Single-
1757−1769. Molecule Site-Specific Detection of Protein Phosphorylation with a
(292) Tamayo, J.; Kosaka, P. M.; Ruz, J. J.; San Paulo, Á .; Calleja, M. Nanopore. Nat. Biotechnol. 2014, 32, 179−181.
Biosensors Based on Nanomechanical Systems. Chem. Soc. Rev. 2013, (313) Han, Q.; Bagheri, N.; Bradshaw, E. M.; Hafler, D. A.;
42, 1287−1311. Lauffenburger, D. A.; Love, J. C. Polyfunctional Responses by Human
(293) Arlett, J. L.; Myers, E. B.; Roukes, M. L. Comparative T Cells Result from Sequential Release of Cytokines. Proc. Natl. Acad.
Advantages of Mechanical Biosensors. Nat. Nanotechnol. 2011, 6, Sci. U. S. A. 2012, 109, 1607−1612.
203−215. (314) Shi, Q.; Qin, L.; Wei, W.; Geng, F.; Fan, R.; Shik Shin, Y.;
(294) Chaste, J.; Eichler, A.; Moser, J.; Ceballos, G.; Rurali, R.; Guo, D.; Hood, L.; Mischel, P. S.; Heath, J. R. Single-Cell Proteomic
Bachtold, A. A Nanomechanical Mass Sensor with Yoctogram Chip for Profiling Intracellular Signaling Pathways in Single Tumor
Resolution. Nat. Nanotechnol. 2012, 7, 301−304. Cells. Proc. Natl. Acad. Sci. U. S. A. 2012, 109, 419−424.
(295) Hanay, M. S.; Kelber, S.; Naik, A. K.; Chi, D.; Hentz, S.; (315) Lu, Y.; Xue, Q.; Eisele, M. R.; Sulistijo, E. S.; Brower, K.; Han,
Bullard, E. C.; Colinet, E.; Duraffourg, L.; Roukes, M. L. Single- L.; Amir, E. D.; Pe’er, D.; Miller-Jensen, K.; Fan, R. Highly
Protein Nanomechanical Mass Spectrometry in Real Time. Nat. Multiplexed Profiling of Single-Cell Effector Functions Reveals
Nanotechnol. 2012, 7, 602−608. Deep Functional Heterogeneity in Response to Pathogenic Ligands.
(296) Lafleur, J. P.; Jönsson, A.; Senkbeil, S.; Kutter, J. P. Recent Proc. Natl. Acad. Sci. U. S. A. 2015, 112, E607−E615.
Advances in Lab-on-a-Chip for Biosensing Applications. Biosens.
Bioelectron. 2016, 76, 213−233.
(297) Raiteri, R.; Nelles, G.; Butt, H. J.; Knoll, W.; Skládal, P.
Sensing of Biological Substances Based on the Bending of
Microfabricated Cantilevers. Sens. Actuators, B 1999, 61, 213−217.
(298) Fritz, J.; Baller, M. K.; Lang, H. P.; Rothuizen, H.; Vettiger, P.;
Meyer, E.; Güntherodt, H. J.; Gerber, C.; Gimzewski, J. K. Translating
Biomolecular Recognition into Nanomechanics. Science 2000, 288,
316−318.
(299) Wu, G.; Ji, H.; Hansen, K.; Thundat, T.; Datar, R.; Cote, R.;
Hagan, M. F.; Chakraborty, A. K.; Majumdar, A. Origin of
Nanomechanical Cantilever Motion Generated from Biomolecular
Interactions. Proc. Natl. Acad. Sci. U. S. A. 2001, 98, 1560−1564.
(300) Wu, G.; Datar, R. H.; Hansen, K. M.; Thundat, T.; Cote, R. J.;
Majumdar, A. Bioassay of Prostate-Specific Antigen (PSA) Using
Microcantilevers. Nat. Biotechnol. 2001, 19, 856−860.
(301) Swierczewska, M.; Liu, G.; Lee, S.; Chen, X. High-Sensitivity
Nanosensors for Biomarker Detection. Chem. Soc. Rev. 2012, 41,
2641−2655.
(302) Kosaka, P. M.; Pini, V.; Ruz, J. J.; Da Silva, R. A.; González, M.
U.; Ramos, D.; Calleja, M.; Tamayo, J. Detection of Cancer
Biomarkers in Serum Using a Hybrid Mechanical and Optoplasmonic
Nanosensor. Nat. Nanotechnol. 2014, 9, 1047−1053.
(303) Shekhawat, G. S.; Dravid, V. P. Biosensors: Microcantilevers
to Lift Biomolecules. Nat. Nanotechnol. 2015, 10, 830−831.
(304) Bietsch, A.; Zhang, J.; Hegner, M.; Lang, H. P.; Gerber, C.
Rapid Functionalization of Cantilever Array Sensors by Inkjet
Printing. Nanotechnology 2004, 15, 873−880.
(305) Yue, M.; Stachowiak, J. C.; Lin, H.; Datar, R.; Cote, R.;
Majumdar, A. Label-Free Protein Recognition Two-Dimensional
Array Using Nanomechanical Sensors. Nano Lett. 2008, 8, 520−524.
(306) Burg, T. P.; Godin, M.; Knudsen, S. M.; Shen, W.; Carlson,
G.; Foster, J. S.; Babcock, K.; Manalis, S. R. Weighing of
Biomolecules, Single Cells and Single Nanoparticles in Fluid. Nature
2007, 446, 1066−1069.
(307) Tao, Y.; Boss, J. M.; Moores, B. A.; Degen, C. L. Single-
Crystal Diamond Nanomechanical Resonators with Quality Factors
Exceeding One Million. Nat. Commun. 2014, 5, 3638.
(308) Waggoner, P. S.; Varshney, M.; Craighead, H. G. Detection of
Prostate Specific Antigen with Nanomechanical Resonators. Lab Chip
2009, 9, 3095−3099.
(309) Patil, S. B.; Vögtli, M.; Webb, B.; Mazza, G.; Pinzani, M.; Soh,
Y. A.; McKendry, R. A.; Ndieyira, J. W. Decoupling Competing
Surface Binding Kinetics and Reconfiguration of Receptor Footprint
for Ultrasensitive Stress Assays. Nat. Nanotechnol. 2015, 10, 899−907.
(310) Liu, W.; Chen, D.; Du, W.; Nichols, K. P.; Ismagilov, R. F.
SlipChip for Immunoassays in Nanoliter Volumes. Anal. Chem. 2010,
82, 3276−3282.

321 DOI: 10.1021/acs.chemrev.8b00257


Chem. Rev. 2019, 119, 293−321

You might also like