You are on page 1of 18

Interdisciplinary detection science

www.rsc.org/analyst Volume 133  |  Number 6  |  June 2008  |  Pages 685–824

ISSN 0003-2654

PAPER
CRITICAL REVIEW Ganesh D. Sockalingum et al.
Christof M. Niemeyer et al. Detection of pathological aortic
Sensitivity by combination: immuno- tissues by infrared multispectral
PCR and related technologies imaging and chemometrics 0003-2654(2008)133:6;1-G
i-SECTION: CRITICAL REVIEW www.rsc.org/analyst | The Analyst

Sensitivity by combination: immuno-PCR


and related technologies
Michael Adler,a Ron Wacker*a and Christof M. Niemeyer*b,c
DOI: 10.1039/b718587c

The versatility of immunoassays for the detection of antigens can be combined with the
signal amplification power of nucleic acid amplification techniques in a broad range of
innovative detection strategies. This review summarizes the spectrum of both,
DNA-modification techniques used for assay enhancement and the resulting key
applications. In particular, it focuses on the highly sensitive immuno-PCR (IPCR) method.
This technique is based on chimeric conjugates of specific antibodies and nucleic acid
molecules, the latter of which are used as markers to be amplified by PCR or related
techniques for signal generation and read-out. Various strategies for the combination of
antigen detection and nucleic acid amplification are discussed with regard to their
laboratory analytic performance, including novel approaches to the conjugation of
antibodies with DNA, and alternative pathways for signal amplification and detection. A
critical assessment of advantages and drawbacks of these methods for a number of
applications in clinical diagnostics and research is conducted. The examples include the
detection of viral and bacterial antigens, tumor markers, toxins, pathogens, cytokines and
other targets in different biological sample materials.

1. Introduction: the power of polyclonal antibodies, as well as from our


DNA understanding of enzymatic amplification
systems. Our ability to link antibodies and
a
Chimera Biotec GmbH, Emil-Figge-Str. 76 A, Our increasing knowledge of the composi- enzymes to generate bio-molecular devices
D-44227, Dortmund, Germany. tion, structure and properties of biological has led to the variety of specific and sensi-
E-mail: wacker@chimera-biotec.com; Fax: +49
(0)231-9742-844 molecules allows us to take advantage tive assays, such as the so-called enzyme-
b
Technische Universität Dortmund, Fachbereich of the functionality of these molecules, linked-immuno-sorbent-assay (ELISA).1 In
Chemie, Lehrstuhl für Biologisch-Chemische to be implemented as versatile tools in ELISA, an antibody specifically recog-
Mikrostrukturtechnik, Otto-Hahn-Str. 6, a broad variety of novel analytical as- nizes the target (antigen) structure and
D-44227, Dortmund, Germany. E-mail: christof.
niemeyer@tu-dortmund.de; Fax: +49 says. As an example, the development the connected enzyme usually converts a
(0)231-755-7081 of immunological detection methods has low molecular weight chemical substrate
c
ISAS – Institute for Analytical Sciences, largely benefited from the elucidation of into a colored, fluorescent or lumines-
Bunsen-Kirchhoff-Strasse 11, 44139 Dortmund, structure and functionality of antibodies, cent product. Due to the catalytic nature
Germany
knowledge of how to raise mono- and of enzyme-mediated substrate conversion,

Dr Michael Adler (chemist and head of laboratory) and Dr


Ron Wacker (biologist and product manager) are employees at
Chimera Biotec GmbH, a company commercializing analytical
applications of DNA–protein conjugates. The company was
founded in 2000 by Dr Adler and Dr Christof M. Niemeyer.
Since 2002, Dr Niemeyer has been Professor of Chemistry at the
Technical University of Dortmund (Germany) where he holds
the chair of Biological and Chemical Microstructuring. Since
2005, he has held an additional appointment as a group leader
at the ISAS – Institute for Analytical Sciences, Dortmund. His
research interests concern the chemistry of semisynthetic DNA–
protein- and nanoparticle-conjugates and their applications in
life-sciences, catalysis and molecular nanotechnology.
Dr Ron Wacker, Dr Christof M. Niemeyer and
Dr Michael Adler (left to right)

702 | Analyst, 2008, 133, 702–718 This journal is © The Royal Society of Chemistry 2008
the binding event of the antibody oc-
curring at the level of few molecules is
amplified, thus becoming detectable at
the macroscopic level. The utilization of
enzymatic activity for detection of im-
munological binding events, however, is
no longer limited to the typical small-
molecule enzyme substrates. Nowadays,
an increasing number of research and
commercial applications concern the use
of macromolecular substrates, in partic-
ular, nucleic acid molecules. The combi-
nation of DNA-processing enzymes with
the general principle of ELISA method-
ology results in a range of novel ana-
lytic techniques which are based on the
unique properties of DNA as a functional
molecule (Fig. 1, 2, 3). Considering the
various different strategies used for DNA
processing as well as the broad field of
applications, it is helpful to provide an
overview on this rapidly growing field of
assay development (Fig. 4) to assist the
non-expert in selecting a suitable tech-
nique which meets the demands of a given
analytical challenge.
The idea of combining an antibody-
based binding event with the signal am-
plification power of PCR goes back to
the 1992’s with the initial description of
Fig. 1 Key steps of DNA-enhanced immunoassays. Note that additional details regarding
immuno-PCR (IPCR).2 The polymerase antibody–DNA coupling are summarized in (Fig. 3) while aspects regarding signal-generation
chain reaction (PCR)3,4 is a prime tool and DNA detection are further illustrated in (Fig. 6 and 7).
for taking advantage of the intrinsic prop-
erties of the DNA molecule to be used
as the substrate for signal amplification5
(Fig. 3A). The exponential amplifica-
tion of a DNA molecule and, there-
fore, the detection of even single target
molecules,6 is nowadays routinely con-
ducted by PCR, this level of sensitivity
is inaccessible for conventional ELISA
methods (Fig. 5). Furthermore, the infor-
mation stored within the DNA sequence,
the physicochemical stability of DNA
molecules and the number of enzymes and
techniques available for selective manipu-
lation and labeling significantly enhances
the applicability of DNA-based assays.
Fig. 2 The intrinsic properties of DNA molecules can be used to improve almost all fields
However, there are several major chal- of immunoassays. The scheme summarizes the range of different targets studied by exemplary
lenges to be solved in order to real- applications including IPCR with protein chimeras,20,21 Universal-IPCR,13 immuno-RCA,58
ize practical and routinely applicable im- PD-IPCR,44 ImperacerTM ,37,55 competitive assays for small molecules134 or proximity ligation.62
munoassays with the implementation of
the exponential nucleic-acid amplification
by PCR or related techniques (Fig. 1): and drawbacks and appropriate tech- tion leads to novel requirements for assay
(1) Coupling of immunoassay reagents niques have to be chosen according to the design, as well as new options in terms
and DNA markers: several strategies are demands of the intended application. of the scientific or diagnostic questions
available for linking antibody recognition (2) Assay adaptation, modification and which can be addressed by the assay. While
with nucleic acid amplification (Fig. 3B). application: the enormous sensitivity due technically the combination of antibody
Each alternative has its own advantages to the exponential nucleic acid amplifica- binding and enzymatic DNA amplification

This journal is © The Royal Society of Chemistry 2008 Analyst, 2008, 133, 702–718 | 703
critical assessment of the three aforemen-
tioned aspects to provide the reader with
expert knowledge regarding practical as-
say performance in this rapidly expanding
field of fundamental and applied research.
In the first part we briefly introduce dif-
ferent types of immunoassays which are
based on specific processing of the DNA
marker. While some approaches are still in
a proof-of-concept phase, other methods
have already reached the stage of routine
laboratory techniques. All techniques have
in common, a significant increase in the
sensitivity of the assay. In the second part,
we critically evaluate the potential advan-
tages and drawbacks of the established
methods, including a detailed discussion
and comparison of various different DNA
detection techniques. In the third part, we
spotlight selected applications from the
broad range of analytical targets (Fig. 2)
to discuss how advantages, in particular
applications, have been realized and how
problems have been circumvented, thus
illuminating the actual practical perfor-
mance of DNA-enhanced immunoassays.

2. Techniques: a journey
through the art of DNA-
enhanced immunoassays
Fig. 3 The evolution of immuno-PCR (IPCR): (A) the set-up of ELISA and IPCR is similar. The pioneering work on the use of DNA
Instead of an enzyme marker, such as alkaline phosphatase (left), IPCR uses amplification of
as an amplifiable label in immunoassays
attached DNA for signal generation (right). (B) Different strategies for coupling antibodies and
was presented in 1992 by Sano, Smith and
DNA: in the initial version of IPCR2 a Streptavidin (STV)–protein A chimeric fusion protein
was used for tagging the detection antibody with biotinylated DNA (I). In the universal
Cantor, who established an initial protocol
IPCR protocol,8–10,13,23 the signal generating complex is assembled in situ by subsequent for the IPCR method2 (Fig. 3B I). The key
incubation steps of biotinylated detection antibody, (strept-)avidin and biotinylated DNA; to IPCR was an inversion of conventional
either using a non-biotinylated primary and a species specific secondary antibody (II) or a ELISA protocols. ELISA uses antibody–
directly biotinylated primary antibody (III). The introduction of pre-assembled antibody– enzyme conjugates while the enzyme’s
DNA conjugates takes advantage of either species- or marker-specific secondary conjugates92 substrate is added subsequently as a freely
(IV) or direct conjugation of target-specific antibodies and DNA14 (V). Approaches such diffusing species. In IPCR the enzyme’s
as phage display mediated IPCR,44 tadpoles of antibodies and DNA,38 or native chemical substrate, i.e., a DNA template, is linked
ligation41–43 introduce elegant ways of coupling antibodies and DNA by circumventing artificial to the antibody while the enzyme is added
modifications such as biotin and complex crosslinking chemistries (VI). The linkage of multiple
subsequently (Fig. 3A). By amplifying the
antibodies and DNA molecules with particles, as used in the bio-barcode technology92 has led
DNA marker through PCR, the limit-of-
to polyvalent reagents, which allow one to connect single antibody–antigen binding events to
a larger number of DNA markers (VII). (C) Comparison of the multiple steps required for the detection (LOD) of a given ELISA is, in
in situ stepwise reagent assembly of the classical universal IPCR approach with the simplicity general, enhanced 100–10 000-fold (Fig. 5
of specific antibody–DNA conjugates. Note that each coupling step requires optimization of and 8). This simple and elegant approach
reaction parameters and leads to a loss in sensitivity. is still the most popular strategy for ul-
trasensitive protein detection by means of
DNA amplification, and nowadays it has
is still a modification of ELISA, the differ- to be coupled with suitable techniques to been established as a routine technique for
ences of both methods are the key for the allow fast, convenient and robust quantifi- applications in both fundamental research
proper choice and application of a suitable cation of the DNA amplification products and applied immunological diagnostics.8,10
assay. (Fig. 7). The true challenges for the practical ana-
(3) Signal amplification and assay read- A number of excellent review articles7–11 lyst, however, are found in the variety of
out: the products of ELISA signal am- have already addressed topics of the different techniques for reagent synthesis,
plification are typically easily detectable. emerging field of DNA-coupled im- signal generation and adaptation to the
Nucleic acid amplification, however, has munoassays. This article focuses on the analytical target under investigation.

704 | Analyst, 2008, 133, 702–718 This journal is © The Royal Society of Chemistry 2008
Fig. 4 The immuno-PCR “family tree”: historical development of the different immuno-PCR techniques during the past 15 years. While the
number of applications was limited during the initial decade of method refinement (approx. 100 different assays were reported—see Fig. 8
for a statistical analysis), the broad range of today’s applications (see Fig. 2 and 10) now underlines the potential of this method. As the first
milestones in IPCR development focused on the basic questions of antigen–DNA coupling and DNA detection (see also section 2.1 and 2.2),
recent innovations are aiming towards the improvement of assay handling and homogenous assay design, thereby rendering the initial research
method to a routine analytical technique.

2.1 First challenge: coupling of incubation step.15–17 Moreover, the protein reagents is the significant shortening of
antibodies with DNA A-STV fusion is unsuitable for two-sided the assay protocol and thus a reduction
immunoassays because it is impossible to of incomplete and/or non-specific binding
2.1.1 Modular approach/in-situ cou- completely suppress its binding to plate- events which usually occur in each incu-
pling. In order to physically link an- immobilized capture antibodies. Nonethe- bation step.24–26 It has been demonstrated
tibodies with DNA for IPCR appli- less, this reagent is suitable for applica- that the performance of pre-assembled
cations, various strategies have been tions which concern the detection of di- antibody–DNA conjugates exceeds that of
investigated.8,10,12 In general, two different rectly immobilized target molecules, such the stepwise assembled complexes in the
approaches were used in the majority of as single blastocytes in pre-implantation modular approach26 (Fig. 3C). However,
examples. These (i) either use a linker diagnostics.18–21 In a related approach, a the general drawbacks of these reagents
protein, such as streptavidin (STV) for bispecific fusion protein comprised of the stem from their complex syntheses and pu-
sequential coupling of (biotinylated) anti- polypeptide chains of carcinoembryonic rification procedures which require expert
bodies and (biotinylated) DNA in a multi- antigen (CEA) binding protein and strep- knowledge in protein chemistry.14,25,27–33
step protocol13 (Fig. 3B II & III and sec- tavidin was recently used in the detection With the development of refined protocols
tion 2, below) or (ii) employ pre-assembled of circulating CEA in human sera.22 The for the synthesis as well as the availabil-
and purified antibody–DNA conjugates “Universal IPCR” protocol of Zhou et al., ity of universal species-specific antibody–
in a single-step protocol14 (Fig. 3B IV, V the so far most popular and most reviewed DNA conjugates, which can be used
& VII and following paragraph). Sano’s approach for model applications8–10,13,23 similar to the common species-specific
initial IPCR was carried out following also follows the first strategy. antibody–enzyme conjugates in ELISA,34
the first strategy. The linker protein was the complex chemistry typically realized
a genetic fusion of STV and protein A, 2.1.2 Chemical crosslinking. Start- by heterobifunctional crosslinkers14 is no
thus allowing for the binding of the Fc ing from the “classical” IPCR proto- longer an issue for application of the
part of an IgG and subsequent tagging col, the potential of DNA as an ana- IPCR method.23,34,35 Application of dis-
with biotinylated dsDNA2 (Fig. 3B I). lytic tool was exploited since the early tinct ready-to-use IPCR-reagents together
While the use of this linker allows for 1990’s (Fig. 4). Nowadays, the use of with optimized protocols allow even inex-
simple labeling and high flexibility, the pre-assembled antibody–DNA conjugates perienced users to facilitate highly sensi-
multi-step assembly implies the disadvan- (Fig. 3B IV & V) is steadily increasing its tive quantitative immuno-PCR (qIPCR)23
tage of time consuming adjustments of foothold in the design of IPCR assays. assays with average routine LOD of sev-
reagent concentrations for each individual One major advantage of this class of eral 1000 molecules (corresponding to

This journal is © The Royal Society of Chemistry 2008 Analyst, 2008, 133, 702–718 | 705
targeted synthesis of functional IPCR
reagents.
Another biological inspired concept for
the conjugation of antibodies with DNA
was recently introduced by Guo et al. who
described a phage display-mediated IPCR
assay44 (Fig. 3 VI). Single chain variable
fragments (scFv) of IgG antibodies were
cloned and expressed on the surface of
phages45 and the complete phage was
used for binding to the target antigen.
Following the release of the DNA from the
immobilized phage, PCR amplification
enabled high sensitivity of this assay. This
was illustrated by detecting 10 pg ml−1
of purified Hantaan virus antigen NP or
1 ng ml−1 of the prion protein. This work
also indicated that IPCR is still feasible
even with low-affinity binders (i.e., the
scFv chains) thereby opening the door to
novel applications. In an recent continua-
tion, the usefulness of this technique for
the selection of recombinant antibodies
and their binding proteins in plasma cells
of multiple sclerosis CSF was demon-
strated by Yu et al.46 There, IPCR was used
as a screening tool with a sensitivity of as
few as 100 phage particles compared to a
detection limit of more than 104 particles
Fig. 5 Typical results of immuno-PCR (IPCR) experiments. (A) Comparison of IPCR, the
in conventional ELISA.
analogous conventional ELISA for the detection of Rotavirus antigens.108 Note the high
In a “minimalist” approach, target–
linearity and broad dynamic range of IPCR. (B) Comparison of different IPCR assay
techniques for the detection of human TNFa: the use of target-specific antibody–DNA protein binding nucleic acid aptamer
conjugates enables an increased sensitivity. The dark and light blue bars represent signals sequences47,48 can substitute antibodies,
obtained by sequential IPCR (see Fig. 3B III) and direct IPCR with pre-assembled antibody– thereby using oligonucleotides for both,
DNA conjugates (see Fig. 3B V), respectively. The red curve represents signals obtained in the target recognition and signal detection.
analogous ELISA. This strategy, which is elegant in its
simplicity as well as challenging in the
design of the aptamers, has been reviewed
sub-picogram amounts of typical pro- coupling of recognition proteins can be recently.9
teins) and a broad dynamic range of up to achieved without further modification.40
six orders of magnitude8,10 (Fig. 5), utilized Using the tadpoles, 32 000 molecules of 2.2 Second challenge: signal
e.g. in different pharmacokinetic clinical the model protein PA (protective antigen generation and detection
studies.36,37 Further advantages and draw- subunit of B. anthracis) were detected
backs of this approach are discussed below From “classical” nucleic acid analysis, a
in spiked human serum, purified by
in section 2. broad number of well-established quan-
gel-fractionation and further extracted
tification techniques are already available
with magnetic beads. The fractionation
for application in IPCR-based assays. The
2.1.3 Native/biomimetic strategies. was found to be necessary because
practical use of these techniques is there-
In parallel to these application-oriented the beads bound the tadpoles non-
fore, discussed in detail in section 3.2,
developments, the properties of DNA specifically, regardless of the protein
below. For the purpose of evaluating the
also were the key to novel approaches head.38 These initial problems suggested
special properties of DNA as a marker
in conjugate synthesis. As an example, that the complexity of the assay was
molecule in immunoanalytics, however, a
Burbulis et al.38 followed the pioneering shifted from the actual coupling step to
number of unique detection strategies are
work of Niemeyer’s group39 by employing the generation of molecules and assay
also especially interesting.
the biological protein splicing system conditions suitable for this approach.
for the coupling of DNA molecules Recent advances in the field of intein- 2.2.1 Transcription. In the “fluores-
with intein-tagged binding proteins. The mediated protein–DNA-coupling41–43 will cent amplification catalyzed by T7-poly-
resulting so-called “tadpole”-chimeras certainly support the general concept merase technique” (short: FACTT,
revealed full functionality in IPCR of merging coupling strategies adopted which was initially termed “immuno-
assays,38 thus demonstrating how direct from biological processes with the detection amplified by T7 RNA

706 | Analyst, 2008, 133, 702–718 This journal is © The Royal Society of Chemistry 2008
for the firefly luciferase protein. Forma-
tion of the immunocomplex was followed
by a one-step, cell-free translation expres-
sion step, which enabled the detection
of 3000 DNA molecules or 50 000 anti-
gen molecules, respectively. Therefore, this
method not only represents an additional
example how nature’s functional elements
can be utilized for in vitro diagnostics
but it also underlines the versatility of
DNA strands as a tool for innovative
immunoassay design.
2.2.3 Phosphorylation/dephosphory-
lation. Protein detection by DNA ampli-
fication is also possible by addressing
the functionality of the proteins directly.
Banin et al.53 introduced an alternative
approach to IPCR for the detection
of the enzyme alkaline phosphatase
(AP) using phosphorylated DNA as the
substrate. In the presence of AP, the
DNA substrate was dephosphorylated
and, thus, no longer a target for a k
exonuclease which otherwise degrades
the phosphorylated DNA. Therefore, the
presence of AP is confirmed when samples
incubated with phosphorylated DNA and
Fig. 6 Semi-homogenous DNA-enhanced immunoassays carried out in liquid phase reac- k exonuclease still yield signals in PCR
tions. (A) Bio-Barcode technology: magnetic beads coupled with antigen-specific capture amplification of this DNA substrate. This
antibodies (1) and gold particles135,136 coated with antigen-specific detection antibodies as assay was highly sensitive, allowing for the
well as a DNA-marker sequence (Bio-Barcodes) (2) are incubated with the target antigen detection of a few thousand molecules of
(3). Antigen-linked particles are isolated by magnetic separation (4) and the DNA marker AP. The approach was implemented into
is subsequently released by denaturation (5). Since each particle is coated with a multitude immunoassays by using specific antibody–
of DNA markers, many Bio-Barcodes are released for each positive binding event. In the
AP conjugates, thereby enabling the
final step (6), the released DNA marker is detected, e.g., by sequence-specific hybridization,90
detection of Chlamydia antigens with
colorimetric changes by hybridization with additional gold nanoparticle probes,72 or for
maximum sensitivity down to a few molecules, a subsequent conventional PCR step74 (B)
about a 1000-fold increase in sensitivity,
Magneto-IPCR: magnetosome particles functionalized with antigen-specific antibodies (7) as compared to conventional colorimetric
are incubated with the target antigen as well as an antigen-specific antibody–DNA conjugate ELISA-detection of the antibody–AP
(8). Following magnetic separation, magnetosome particles containing DNA are detected conjugate.53
directly by PCR (9). (C) DDI-IPCR: capture-antibodies, coupled with an oligonucleotide (10)
2.2.4 Nucleotide encoded informa-
as well as an antibody–DNA detection conjugate (8) are simultaneously incubated with the
target antigen. Conjugates of all three compounds are isolated by hybridization with a solid tion. The information content of the
phase-bound capture oligonucleotide (11) and the DNA marker is detected by PCR (9). DNA’s base sequence and/or the length
of the DNA marker allow for a multiplex
detection of several antibody–DNA con-
polymerase-IDAT”), DNA-labeled anti- breast cancer tumor markers in tissue jugates within a single experiment when
bodies were coupled to their target anti- samples.49,50 A potential drawback of the several antibodies are labeled with indi-
gens and the DNA marker was amplified technique results from special precautions vidual DNA markers (Fig. 9). The feasi-
by transcription using T7 polymerase. The which have to be taken into account bility of this approach was demonstrated
amplified RNA was then quantified by to prevent degradation of the inherently already in 1994 by Ebersole’s group in
addition of the RNA-intercalating dye Ri- unstable RNA by the ubiquitous RNases. the simultaneous detection of three dif-
boGreen and subsequent fluorescence de- ferent antigens (b-galactosidase, human
tection. Thus, enzymatic transcription was 2.2.2 Expression. The genetic infor- thyroid-stimulating hormone and human
used for signal amplification. Although mation stored in the DNA marker has chorionic gonadatropin).14,54 An approxi-
this assay lacks the typical exponential also been harnessed to express a detection mately 100-fold enhancement in sensitiv-
amplification characteristics of PCR, an enzyme as a means for signal genera- ity, as compared to conventional ELISA,
impressive 1000-fold increase in sensitivity tion. This approach, termed Expression was observed. Interestingly, despite the
was reported, e.g. in the detection of Immunoassay51,52 took advantage of a bi- striking possibilities of such a multiplex
prion aggregates in serum or her2/neu otinylated DNA marker which encoded approach, no further applications of

This journal is © The Royal Society of Chemistry 2008 Analyst, 2008, 133, 702–718 | 707
multiplex IPCR have been reported so far.
Possible explanations certainly include the
incompatibility with the widespread mod-
ular IPCR approach (Fig. 3B I–III), which
makes it impossible to individually label
certain antibodies with a distinct DNA
marker. Moreover, the general problem
of conventional multiplex PCR obviously
also applies to multiplexed IPCR assays.
The design of a set of DNA markers which
can be amplified simultaneously without
interference inevitably limits the amount
of simultaneously detectable targets.
The interference of simultaneously am-
plified DNA markers, however, can pur-
posefully be utilized for standardization
of IPCR assays when a second DNA
fragment is added to the assay as an
internal competitor. The calculation of
the ratio of products originating from
either the IPCR marker or the competi-
tor fragment was found to enhance the
signal-to-background ratio of the assay.
This approach had been used for quan-
titative antigen detection in plasma sam-
ples during a clinical phase I pharma-
cokinetic study of the anti-tumor drug
Aviscumine.8,55 As an additional advan-
tage, the generation of competitor signals,
which are inversely proportional to those
of DNA marker, simplifies the avoidance
of false-negative signals because correct
performance of the DNA-amplification
can be judged conveniently from the pres-
ence of competitor signals, even in the
absence of the target antigen. The use
of internal competitors, however, requires
methodologies for the independent de-
tection of the two PCR amplicons. To
this end, gel-electrophoresis8 as well as
sequence specific hybridization55 has been
Fig. 7 Comparison of the most prominent methods for the detection and quantification of used (Fig. 7).
DNA amplicons generated in DNA-enhanced immunoassays. (A) Intercalation fluorescence With respect to multiplexed DNA-
markers with increased specificity for double-stranded DNA, such as ethidiumbromide or enhanced immunoassays, the problems
SYBR greenTM , are used in gel-electrophoresis or real-time PCR analyses. Note that for
well-known for standard multiplex PCR
multiplex IPCR, it is necessary to separate amplicons of different length by gel-electrophoresis
have been effectively circumvented by
while multiplex real-time detection can not be performed using intercalation marks. (B)
Different types of sequence-specific fluorophore-labeled nucleic acid probes, e.g. TaqManTM or
a different type of DNA amplification,
ScorpionTM are typically used for real-time quantitative PCR. During elongation of primers, namely the rolling-circle amplification
the probes are modified and thereby, a fluorescent signal is generated. (C & D) Hybridization (RCA) of immobilized nucleic acids. In
assays for sequence-specific DNA-detection. Sensitivity can be increased by binding of multiple the immuno-RCA technology, an oligonu-
fluorophores to the amplified DNA by means of hybridization of fluorophore-labeled probes to cleotide primer, covalently attached to
products of RCA (C) or PCR (D) processes. In the case of immuno-RCA, the antibody–DNA– the detection antibody, is enzymatically
conjugate remains intact during DNA amplification and thus, a multitude of hybridization elongated by adding a circular single-
probes can bind to spots of microarrays, containing the immobilized antigen. In PCR-ELOSA strand DNA as the template56–59 (Fig. 7C).
(D), hapten-labeled amplicons, generated during PCR, are immobilized by means of surface- This method, recently reviewed in this
bound capture oligonucleotides and subsequent detection is carried out by using hapten-
journal,9 has been shown to be partic-
specific antibody–enzyme conjugates.
ularly well suited for multiplex analyses,
impressively demonstrated by the profiling
of 75 cytokines on microarrays,59 or the

708 | Analyst, 2008, 133, 702–718 This journal is © The Royal Society of Chemistry 2008
to an enzyme-amplified assay59 with the
advantage of the localized DNA amplifi-
cation product at the site of the antibody.
The approach is therefore well suited for
protein microarray applications.

2.3 Third challenge: assay


performance in the liquid phase
The standard platform for performing im-
munoassays is so far the solid phase, either
by applying typical ELISA microplates
or using microarrays. While this assay
format suffers from inefficient interphase
binding steps, a different strategy takes ad-
vantage of semi-homogenous (involving
a surface/bead for subsequent separation
of bound/no-bound compounds) or true
homogenous assay conditions. There, cou-
pling steps are carried out in the liquid
phase which leads to improved binding
kinetics and thus more efficient labeling
of antigens with antibodies and markers
(Fig. 6).
2.3.1 Simultaneous binding of more
than one antibody–DNA conjugate. In
the proximity ligation method61,62 two an-
tibodies, each of which binds to a specific
epitope of the antigen, are tagged with
individual short oligonucleotides. Enzy-
matic ligation of the two tags in the
presence of a third DNA oligomer leads to
the formation of a longer DNA which sub-
sequently functions as the target for PCR
or RCA amplification. A detailed review
of this technique was published recently.9
In the related approach of nucleic acid de-
tection immunoassay (NADIA),63 a short
Fig. 8 Statistical analysis of references reporting DNA-enhanced immunoassays: (A) self-priming overlap of two antibody-
summary of detection limits reported. The majority of examples revealed a maximum linked oligonucleotide sequences is used
sensitivity in the 0.016 amol–16 amol range (1000–100 000 molecules, respectively), thus
to initialize PCR amplification.
defining the standard performance of the method. Note that the broad detection range
A major challenge for proximity lig-
of about ≤10 molecules2,74,102 or single cells18 up to 1010 molecules in all cases involves a
significant improvement of the analogous ELISA.137,138 A typical LOD is found at approx. ation and NADIA concerns the careful
1000 molecules/sample, which is in accordance with the expected theoretical kinetics of selection of specific antibody pairs, which
immunoassays.13 (B) Overview of the N-fold improvement of conventional ELISA by the bind to a target in an appropriate distance
analogous IPCR. The sensitivity enhancement varies from 5-fold31 to up to 1 000 000 000- for the intended ligation. As a solution
fold82,86 depending on the design and optimization state of the assay as well as the performance to this intrinsic design problem, the use
of the antibodies. The majority of studies reported a 100–1000-fold improvement in LOD. of a population of polyclonal antibodies
(C) Overview of the linear dynamic range of IPCR applications. While conventional ELISA against a given target was suggested.64
typically reveals a dynamic range of two orders of magnitude in antigen concentration, IPCR This population is divided into two parts,
shows a significantly broader dynamic range (see also Fig. 5). In the majority of IPCR
each of which should include suitable
applications, the dynamic range comprised about four orders of magnitude.
binding partners for different epitopes due
to the diversity of binders in a polyclonal
serum. Indeed, this approach proved to
simultaneous detection of 11 cytokines in occurs in a linear fashion, which is by far be strikingly effective in the detection of
a bead-based assay.58 Drawbacks of this not as efficient as the almost exponential cytokines in a homogenous test format
approach concern the necessity of specif- amplification occurring in PCR. How- with femtomolar detection sensitivities.64
ically designed circular DNA templates60 ever, the gain in sensitivity obtained in However, the variability of polyclonal
as well as the fact that primer elongation immuno-RCA was found to be similar antibodies may also be a drawback for

This journal is © The Royal Society of Chemistry 2008 Analyst, 2008, 133, 702–718 | 709
routine applications because slight vari- means.78 The genetically controlled bio- available in typical biochemical or clinical
ations in affinity and specificity may not mineralization process ensures uniform laboratories. Therefore, combining these
always warrant long-term assay repro- particle shape, high magnetic susceptibil- two techniques in IPCR applications is
ducibility. Nevertheless, the linkage of ity, and a narrow size distribution which usually possible without the need for ad-
immunological reactivity and spatial in- can be regulated by choice of the organism ditional instrumentation, unlike the case
formation makes this technique certainly facilitating the magnetosome production. for many other highly sensitive techniques,
unique, especially for studying protein– It is clear from the aforementioned such as electro-chemiluminescence.84
protein interactions61,65–67 or monitoring examples that almost all intrinsic prop- As pointed out above, the availability
changes in protein conformation, for in- erties of the DNA molecule have already of pre-conjugated antibody–DNA conju-
stance as a consequence of splicing or been harnessed by IPCR and related tech- gates largely improves performance and
post-translational modifications.62 niques to improve the performance of handling issues of IPCR assays (Fig. 3C).
While the above described techniques conventional immunoassays. While some Fortunately, problems which often arose
take advantage of DNA as a marker examples are obviously more fascinating in the past from the chemical proce-
for signal amplification, the sequence- from a methodical point of view, quite dures to conjugate proteins with DNA,
specific recognition properties of DNA a number of innovative assay strategies can nowadays be circumvented by taking
can also be the harnessed to realize novel promise novel solutions to the analytical advantage of services and ready-to-use
immunoassay designs. By DNA-directed challenges faced in the daily laboratory reagents available from a growing number
immobilization (DDI), conjugates of an- routine. For a critical evaluation of the of suppliers. Therefore, users who intend
tibodies and short artificial DNA se- properties of theses assays, advantages to set-up an IPCR assay, should critically
quences were side-selectively immobilized and disadvantages of the various strate- evaluate whether it is worth the time and
at surfaces decorated with complemen- gies and applications are discussed in the cost effort to develop their own conjuga-
tary capture sequences.68,69 The combi- following section. tion approach or rather adopt reagents
nation of DDI and IPCR compatible and assays which have already proven their
antibody–DNA conjugates enabled an el- functional performance.
egant one-step assay for the detection
3. Advantages and disadvan-
of antigens by simultaneously incubating
tages: the price of sensitivity
3.2 Various ways of analyte detection:
the target-containing sample with DNA- A number of variations of the IPCR the key to quantitative IPCR
labeled capture- and detection-reagents70 method have been introduced to show
(Fig. 6C). the 1992’s initial protocol is applicable A drawback of the IPCR protocols, as
to today’s requirements of commercially compared to conventional ELISA, may
2.3.2 Immuno-PCR and bead tech- available reagent kits and routine diagnos- result from the assay’s handling effort
nology. A combination of capture- tics. Many of these variations have led to and duration. In particular, the post-
antibody-coated magnetic beads and a general increase in assay sensitivity, and assay quantification of the DNA ampli-
detection-antibody/detection-DNA coa- in some cases dramatic improvements of cons, which is mandatory for quantita-
ted gold nanoparticles71 (Fig. 3B VII) was greater than a 100 000-fold enhancement tive antigen detection, is often considered
recently introduced as the so-called Bio- in ELISA LOD have been reported.73,81–83 a major hurdle for switching from an
Barcode technology,72–75 summarized in In other cases, the IPCR principle was established ELISA to IPCR. ELISA is
Fig. 6A, and this method was also recently supplemented with additional beneficial essentially finished after the addition of
reviewed in more detail.9,10 Although the features, such as multiplex detection or the substrate because the enzymatic sig-
Bio-Barcode assays require specialized compatibility with automation. Here, we nal generation is conveniently detectable
equipment and tailored reagents, the po- will discuss some general considerations and measured after a given time with
tential of the nanoparticle-conjugates, e.g. which users might wish to take into ac- an appropriate reader. In IPCR, instead,
for multiplex detection76 or highly sensi- count when they strive to establish DNA- the DNA amplicons are essentially invis-
tive protein detection77 certainly justifies based immunoassays. ible, and thus they require specific means
this approach to homogenous assay tech- for detection and quantification. Gel-
nology. electrophoresis with DNA-intercalation
3.1 General considerations regarding
As a biogenic alternative to the artifi- labels (Fig. 7A) can be used for this
assay design and complexity
cial magnetic particles used in the Bio- task because it is available in almost
Barcode assays, antibody-functionalized To apply DNA-based immunoassays to an any (bio)chemical laboratory. Thus, this
biogenic magnetosome nanoparticles78 analytical question, one central point of technique has been used in many “clas-
have recently been applied in the magneto- consideration should concern the choice sical” applications of IPCR and related
immuno-PCR (M-IPCR) by Wacker of the appropriate method, which brings techniques.2,8,10,13,15,16,54,85–88 However, this
et al.79 (Fig. 6B). The appeal of this with it the ideal balance between com- approach has intrinsic disadvantages. It is
approach stems from the versatility of plexity of the assay and performance in hardly suitable for quantification purposes
the native particles, which can be pro- the intended target detection. One partic- because band intensity quantification usu-
duced by fermentation of the mag- ularly attractive aspect of combining an ally does not allow one to quantify DNA
netotactic bacterium Magnetospirillum immunoassay with PCR results from the over ranges spanning more than two or-
gryphiswaldense,80 and subsequently mod- fact that the equipment and knowledge ders of magnitude, unless time-consuming
ified with antibodies by synthetic chemical required for both techniques is usually dilution series or radiolabels are applied.

710 | Analyst, 2008, 133, 702–718 This journal is © The Royal Society of Chemistry 2008
Additionally, gel-electrophoresis is very by simply adding a PCR-mix containing variety of filters available for modern
cumbersome when it comes to large sam- the real-time probe, subsequently to the real-time cyclers also allows recording,
ple numbers, because it is difficult and immunoassay. typically, four different fluorescent signals
expensive to automate.27 It is essential that the microplate materi- simultaneously, thus enabling small scale
Due to these problems, alternative ways als used in such qIPCR assays are compat- multiplex detection14 (Fig. 9) or internal
of DNA quantification, initially devel- ible with the real-time PCR instrument. standardization55 to be conveniently facil-
oped in the course of PCR, have gained This means that, besides physical compati- itated.
acceptance in DNA-based immunoassays. bility, the microplate materials must have a Due to the aforementioned reasons, it
For example, labeled probes can be hy- certain degree of protein binding capacity, is not surprising that real-time PCR is
bridized with the PCR products, thus which is usually not the case for typical nowadays routinely used in DNA-based
leading to their sequence-specific tagging PCR microplates. Although the transfer immunoassays.26,29–31,36,66,83,92,99–101 After
with an appropriate detection molecule of the signal-generating immunocomplex solving initial challenges with respect to
(Fig. 7B–D). This principle has been into PCR cycler-compatible vessels is reproducibility and sensitivity, today’s
demonstrated for a variety of techniques, possible,29,30,96 the additional handling of performance of qIPCR is well beyond
including silver-staining, colorimetric,72,75 the contamination-susceptible PCR-mix that obtained by conventional read-out
scanometric,89 fluorescent56,61 or electrical is not recommended because it often leads systems. In particular, qIPCR reveals low
detection.90 Similarly, the PCR product to high background signals and a decrease errors and even an increase in sensitivity,
can be labeled with a hapten molecule ei- in precision of the assay. A number of when compared to conventional end-
ther during amplification (PCR-ELISA)27 first-generation real-time cyclers (e.g. early point-based IPCR read-out.27,70,92,26,92
or subsequent to PCR by hybridization versions of the light-cycler) are capil- Therefore, it seems appropriate to
(PCR-ELOSA).91,92 The hapten-labeled lary or single-vessel based instruments, postulate that real-time detection truly is
DNA is immobilized on a surface, often which therefore require the transfer step. the key to make the initial research-based
microplates, and then coupled with an Nowadays, microplates and modules in IPCR into a routine laboratory method.
antibody–enzyme conjugate which specifi- the standard 96-well format, specifically The resulting protocols for qIPCR23 are
cally binds to the hapten and allows DNA designed for IPCR with high protein similarly convenient to those of routine
detection by enzymatic signal amplifica- binding capacities and a cycler-compatible ELISA protocols with respect to assay
tion. It should be noted that such labeling format are available,92,97 and so are modern handling, simplicity and robustness.
by hybridization is mandatory in specific microplate-compatible cyclers. The use of
cases which require sequence-specific de- these products is recommended, in par- 3.3 From research to routine
tection of the PCR amplicons, as is the ticular, for routine and high-throughput applications: assay robustness and
case in microarray multiplex assays58 or in applications of IPCR. Fortunately, mod- precision
situ analyses.61 Although the throughput ern real-time cyclers are widely available
of these labeling-based assays is much due to the enormous popularity of real- While we have already discussed that
higher than that of electrophoresis, and time PCR for DNA analytics,98 and thus, assay precision depends on the read-out
they often reveal improved sensitivity and no additional equipment needs to be technique, a more in-depth discussion
precision, they still have the drawback purchased when users wish to establish of assay precision, robustness and back-
of being essentially two-step protocols. the real-time IPCR technique. The ground seemed helpful to provide deeper
Therefore, a simpler one-step detection
is needed to reduce assay duration and
to minimize error prone handling and
pipetting steps.
Real-time PCR, also often termed as
qPCR (quantitative PCR), is today’s dom-
inating technique for precise quantifica-
tion of DNA detection. In qPCR a flu-
orescent probe is added directly to the
amplification reaction and it emits flu-
orescent signals upon the generation of
PCR products93–95 (Fig. 7B). This tech-
nique makes any post-PCR quantification
steps obsolete and it minimizes the risk
of contamination of lab space by han-
dling PCR amplicons. Moreover, recent
advances in qPCR probes enable its use for
highly precise quantification of template Fig. 9 Multiplex and polyplex assays for the detection of several antigens in a single sample:
molecules. It is therefore straightforward in multiplex assays, different antigens (a and b) are tagged with different DNA sequences. In
to combine IPCR with a qPCR-based polyplex assays the sample is divided into small aliquots, each of which is analyzed individually
read-out.31 Consequently, IPCR can now by a target specific assay.
be performed as conveniently as an ELISA

This journal is © The Royal Society of Chemistry 2008 Analyst, 2008, 133, 702–718 | 711
insight into potential pitfalls of the IPCR recent study of different assay strategies generating a certain level of background
method. for the detection of human interleukin 6 signal. It was found useful to increase
A major source of increased error rates (hIL6),23 which included detailed proto- the specific coupling efficiency by using
stems from the number of handling steps cols and comparison of sequential incuba- tailored antibody–DNA conjugates,14,23,26
involved in the assay protocol. With each tion, indirect and direct qIPCR (Fig. 3). thus supplementing the blocking step with
additional step the coupling efficiency Sequential incubation as well as indirect increased specific signals.
decreases27 and thus the possibility for sandwich assay showed an LOD of 10
(iii) Dilution of the biological matrix.
errors increases. Multi-step protocols have pg ml−1 hIL6 in human serum (100-fold
Since the background signal is often
been reported as highly sensitive but they improvement compared to ELISA), while
caused by the presence of interfering com-
require careful, laborious adaptation to the use of tailored pre-conjugated reagents
pounds in the sample to be analyzed,
each variation in target antigen, biologi- led to an LOD of 100 fg ml−1 (10 000-fold
minimization of these disturbances was
cal sample, or antibodies.15,102–104 This ap- improvement compared to ELISA).23
achieved by dilution of the sample in
proach, therefore, certainly benefits from For selection of the appropriate assay
appropriate buffers, thus reducing the
expert knowledge of IPCR operation strategy, the main question, therefore, is
influence of the biological matrix8,23,55,108
and, therefore, it is not recommended indeed the intended application. While
(Fig. 10). Furthermore, it is recommended
for the inexperienced user who needs a sequential coupling protocols have the
to use a biological matrix identical to,
fast solution to a given analytical prob- drawback of being time consuming, their
or as similar as possible to, the sample
lem. With each deviation from the well flexibility and the ready availability of
material for the preparation of calibration
proven three-step standard ELISA proto- reagents make them attractive and well
curves in quantitative analyses. This can
col, “add target to microplate—incubate— suited for research applications. How-
also help to compensate for background
add detection conjugate—incubate—add ever, when maximum sensitivity, easy and
effects.23,55
substrate—measure”, potential advan- robust handling as well as fast routine
tages in sensitivity are inevitably linked to applications become a major issue, op- (iv) Standardization and simplifica-
increasing complexity, costs and possible timized protocols based on tailored pre- tion of assay handling. Finally, back-
error sources. conjugated detection reagents are clearly ground signals can result from handling
The advantage of simplified assay pro- advantageous.5 steps with potentially contaminating ma-
tocols is repeatedly confirmed by com- As the most prominent obstacle of terial, such as the transfer of DNA
parison of different assay protocols for IPCR, high background signals often pro- containing solutions30 or highly concen-
a given target. Using the exemplary de- hibit meaningful results. Typically, highly trated DNA stock preparations for in situ
tection of prostate specific antigen (PSA), sensitive assays amplify both, specific sig- coupling of antibody–DNA conjugates.13
Lind et al. compared the performance nals and non-specific background noise. Moreover, low signals and high back-
of IPCR-assays, carried out with either An increase in the signal-to-background ground values occur in IPCR assays when
pre-conjugated antibody–DNA reagents ratio is therefore most important to gen- the protein–DNA conjugate reagents are
or the sequential stepwise in situ cou- erate meaningful results. Various points not properly prepared and/or contami-
pling using STV to bridge biotinylated need to be optimized to reach this goal: nated with free binding proteins and/or
antibodies with biotinylated DNA.26 The marker DNA. To avoid these problems,
(i) Choice of highly specific antibod-
tumor marker PSA is an especially in- single-well solutions with plate materials
ies. The quality of the antibody is in-
teresting target for ultrasensitive diagnos- compatible with PCR cyclers as well as
deed an imperative for any successful im-
tics due to its properties as a biomarker low-concentrated, highly active antibody–
munoassay. Although the highly sensitive
for early detection of prostate cancer.105 DNA conjugates should be used to reduce
IPCR has been demonstrated to function
Therefore, a number of studies used this background signals.
even with weak binders which are usually
antigen to investigate the performance With the increase in kit-solutions and
not well suited for ELISA,44 the specificity
of DNA-enhanced immunoassays.9 Uni- service providers, the robustness and ac-
and affinity of the antibodies are the
formly, all methods revealed a significant cessibility of IPCR technology has sig-
key for the performance of the assay.27,92
(35–200 000-fold) increase in sensitivity, as nificantly increased and diversified in the
Therefore, IPCR assays do perform only
compared to conventional ELISA. Small past few years (Fig. 4). To estimate what
as well as the specificity of the antibodies
sample volumes as well as short assay one might expect from the developments
involved permits.23
duration were also typical for most as- of DNA-enhanced immunoassays typical
says. However, sequential IPCR and the (ii) Blocking of non-specific interac- results from the literature have been sum-
expression immunoassay both required tions. By the choice of an appropri- marized (Fig. 8). Analysis of the reported
significantly more time and handling ef- ate blocking reagent for coating surface data suggests that the average detection
fort. The increased number of incuba- materials and spiking of buffers, unspe- limit of IPCR is in the range of approx.
tion steps also led to lower sensitivities cific binding of assay reagents can be 1000 molecules/sample. This corresponds
in these assays. In contrast, the most minimized, thereby reducing the overall to an on average 100–1000-fold increase
sensitive detection was obtained by us- background signal.8,23,85 Owing to the ex- in the LOD of the corresponding con-
ing a standardized ImperacerTM IPCR tremely high sensitivity of PCR, it is ventional ELISA. In addition to the im-
kit106 and the nanoparticle-based Bio- impossible to reduce background signal provement of LOD, IPCR-based methods
Barcode.107 The outstanding performance completely, because single non-specifically usually reveal a significantly broader dy-
of tailored reagents was also verified in a bound DNA molecules are sufficient for namic range for the detection of analytes

712 | Analyst, 2008, 133, 702–718 This journal is © The Royal Society of Chemistry 2008
its ability to work with very small sample
volumes.

4.1 Improvement of sensitivity as the


key to early detection of
neurodegenerative diseases

The potential of largely improved sensi-


tivity becomes a major issue when well-
established assays with conventional sen-
sitivity require sample materials which
can only be accessed by stressful inva-
sive techniques or post-mortem sample
drawing procedures. In these cases, the
increase in assay sensitivity can open the
door to the analysis of alternative sample
materials which can be collected with less
effort. With the increasing societal im-
pact of neurodegenerative diseases, such
as scrapie, CJD/vCJD and Alzheimer’s
disease, the research on infectious agents
and biomarkers as prime targets for ultra-
sensitive detection come into focus. The
relevant biomarkers of neurodegenerative
diseases are typically present in the brain,
and their concentration is very low at early
stages of the disease.109 For obvious rea-
sons, a more easily accessible target matrix
than brain homogenate, such as blood
samples, would allow for in vivo/ante
mortem detection. Although the presence
of infectious agents for vCJD in blood has
already been confirmed,110,111 the LOD of
conventional analytical techniques (e.g. lu-
Fig. 10 Immuno-PCR combines the versatility of ELISA with increased sensitivity for a
minescence assays, western blots, capillary
broad range of analytical targets: the robust method allows access to almost all biological electrophoresis) were found to be insuffi-
matrices, including e.g. single cells20 and cell culture,139 tissue samples,140 saliva,141 stool,100,108 cient for the detection of these makers in
brain material,83 food99,100,120 and plant extracts, serum and blood50,55,88,142 from human or non-brain derived samples.112,113
animal organisms. The combination of this robustness and sensitivity with the almost unlimited The detection of 10–100 infectious units
number of targets detectable by immunoanalytics (see also Fig. 2) marks the potential of the of PrPSc by IPCR in infected hamster
IPCR method for the analytical scientist. brain homogenate by Constantine et al.83
in 2005 as well as specificity tests in
human tissue samples carried out by
(see Fig. 5 for typical results and Fig. 8C plications. Therefore, in the final part of Gofflot et al.97,114 in the same year rep-
for a statistical evaluation). Therefore, the this review, we will discuss selected appli- resent very important initial steps to-
nearly 15 years of method developments cations in detail. While general overviews wards the early detection and improved
clearly indicate that DNA-enhanced im- of IPCR applications and related tech- analysis of transmissible spongiform en-
munoassays are superior to conventional niques have been published elsewhere,8,10,12 cephalopathy. In addition, non-infectious
enzyme-amplified detection methods and the applications discussed here have been PrP was chosen as the target for the
hold the potential to help unravel impor- chosen to particularly demonstrate the demonstration of phage-display moder-
tant questions of health, clinical diagnos- diversity of fields where the combination ated IPCR.44 Moreover, Chang et al. ob-
tics, and fundamental research. of protein detection and DNA-based sig- served a 50% sensitivity and 100% speci-
nal amplification is being applied today ficity of illness-associated PrPSc in blood
4. Applications: putting (see Fig. 2). As discussed below, three for early stage and 100% sensitivity for
features of these assays are particularly im- late-stage asymptomatic infections in mice
innovative assay strategies to
portant for practical applications. These by using their so-called “Am-A-FACTT”
the test
include (i) the increase in sensitivity, (ii) the method,49 which combined PrP amplifi-
The touchstone for novel analytical meth- assay’s robustness against the disturbing cation and aggregation with a fluorescent
ods is their performance in practical ap- effects of the biological matrix, and (iii) detection method based on amplification

This journal is © The Royal Society of Chemistry 2008 Analyst, 2008, 133, 702–718 | 713
of antibody–DNA conjugates by T7-RNA formance of immunoassays, which have Barcode, only 1–30 ll sample material is
polymerase (see section 2.2.1). made ELISA the routine technology of required.74,108 This is especially important
It is somewhat surprising that the de- today’s analytical laboratories. in applications where only small amounts
tection of prion proteins by IPCR was not A recent work by Xiang et al. em- of the sample are available and/or acces-
attempted earlier in spite of the fact that ployed IPCR for screening of blad- sible, such as in biopsy, pediatrics, animal
abnormal PrPSc as a potential target was der cancer patients. The highly sensi- studies, or even single cell analytics.
already identified in 1991.115 However, as tive detection of mutated p53 protein in Another great potential for applications
for conventional immunoanalytics, IPCR serum of workers exposed to hazardous of the IPCR method results from the pos-
strongly depends on the performance of substances,118 demonstrated the usefulness sibility to split given samples into parts to
the antibodies. For prion proteins, the key of the method as a tool for disease moni- increase the amount of information acces-
detection antibody specific for PrPSc is still toring as well as for drug development. sible by conventional immunoanalytics.
missing, and therefore a proteinase K di- Due to the high sensitivity of IPCR This “polyplexing” differs from “multi-
gestion is required to distinguish between and related methods, background effects plexing” such that polyplexing describes
native PrP and PrPSc . Obviously, if this caused by interfering matrix components a strategy in which a sample, normally
digestion is incomplete, high background can often be simply diluted away using ap- following to dilution, is split into sev-
signals will occur, especially when highly propriate buffers, without significant de- eral individual assays for different targets,
sensitive assays are used.83 Therefore, the crease in the overall assay sensitivity.55,108 while in multiplexing several analytes are
usefulness of IPCR in these applications In addition to this purposeful dilution detected directly in a single experiment
strongly depends on further development of sample materials, there are also cases (Fig. 9). Owing to the high sensitivity of
of high quality antibodies. Nonetheless, where small amounts of antigen are DNA-enhanced immunoassays, 1 ll or
the implementation of real-time detection present in a very large sample volume. less of a given sample is often sufficient
has significantly increased the potential Chang et al. have impressively demon- for analysis and, thus, the IPCR method
of quantitative PrP-IPCR,83,97 including strated that the sensitivity of IPCR is is perfectly well suited for polyplexing
assays for PrPSc with novel antibodies.116 sufficient to detect a single E. coli cell in a analyses.
volume of 10 l by using a cell-released en- Polyplexing circumvents the general
zyme as the target for immunodetection.86 problems of multiplex assays, which
4.2 Robust performance enables
This result underlines the appeal of IPCR usually result from non-specific interac-
monitoring of rare biomarkers
for the detection of low-level contamina- tions of the various detection reagents
Another prime target for highly sensi- tion, which is particularly important for employed. Antibodies as well as DNA-
tive IPCR assays are marker proteins applications in the monitoring of food markers and their detection probes have to
indicative for virus infections either at allergens33 or toxins released by Clostrid- be carefully selected, designed, evaluated
the early (low virus load) stage of the ium botulinum,32,119 Bacillus thuringien- and modified to warrant for their non-
infection or else from complex matrices sis120 or Staphylococcus aureus.121 interfering performance in the assay.
which contain the marker in only very It is noteworthy that in these examples In this process the challenge increases
small amounts. Recent studies on viral method development was not the prime exponentially with an increasing number
antigen detection concerned, for instance, issue. Instead, well established IPCR pro- of simultaneous assays.122–124 In particular,
the detection of rotavirus antigen in pa- tocols were used to provide answers to the generation and selection of highly
tient stool samples,108 norovirus antigen in analytical questions, such as the time- specific and non interfering antibodies
food samples,100 or HIV antigen in plasma and temperature-dependent relationship with similar binding efficiencies and
samples.117 All these applications have in of toxin secretion by the microorgan- conditions for simultaneous application
common, an on average 1000-fold increase isms, because an outbreak of intoxica- in multiplex assays is a major problem
in sensitivity despite the fact that the IPCR tion in a general population can be in- which severely complicates design
assay was carried out in the presence of the duced by small numbers of microorgan- and applications of multiplex assays.
biological matrices. Although IPCR is of- isms not detectable with classical ana- Furthermore, it has been discussed above
ten expected to be highly susceptible to the lytical techniques,121 IPCR and related that the simultaneous detection of several
disturbing effects of various compounds techniques can help to warrant a new level DNA-markers by PCR-based assays
within complex matrices, these three ex- of environmental and food safety. brings with it additional complications. To
amples clearly indicate the robustness circumvent the problems of multiplexing
of this method in practical applications. IPCR, an example of a polyplex assay was
4.3 New assay strategies for small
Along this line, similar robust behavior reported by Case et al.102 which concerned
sample volumes
of IPCR in routine applications was ob- the parallel detection of the promutagenic
served in clinical phase I studies concern- The high sensitivity of IPCR can also be DNA base adduct O-6-methylguanosine,
ing a new anti-tumor drug including more utilized for analyzing very small sample its monomeric repair enzyme, O-6-
than 40 patients,37,55 or in a recent phar- volumes. While typical ELISA requires methylguanine-DNA methyltransferase
macokinetic study with 15 individuals.36 about 50–100 ll of sample material, the (MGMT), as well as a single peptide from
These examples demonstrate nicely, that increased sensitivity of DNA-enhanced the N-terminus of MGMT. An optimized
the introduction of DNA as a signal- immunoassays allows one to significantly sequential IPCR protocol was used for
generating marker in immunoassays does reduce the assay volume. For highly opti- the detection of the several structurally
not deteriorate robustness and stable per- mized methods like ImperacerTM or Bio- diverse antigens from the catalytic cycle

714 | Analyst, 2008, 133, 702–718 This journal is © The Royal Society of Chemistry 2008
of the cell protecting DNA-repair enzyme ing, including replication, transcription, gets will become increasingly important
MGMT, thus allowing the studying of translation, digestion and hybridization due to the progress in research and the
complex interaction effects within small has already been harnessed for the im- requirements of efficient diagnostics and
sample volumes. Similar detection of provement of immunoassays (section 2). A analytics. Another area which also calls
several antigens with a standardized thorough review of the literature suggests, for ultrasensitive analyses concerns the
IPCR method have been reported by Guo however, that the old truth of Occam’s monitoring of biological compounds such
et al.44 and Adler et al.92 These examples razor that the best approach is typically as contaminants in food, either by toxins
clearly demonstrate the potential of the simplest one, also applies to DNA- or genetically altered protein components.
polyplex IPCR assays for the analysis of enhanced immunoassays. Purity control is also of paramount im-
various targets in a given sample. With real-time PCR, a powerful tool for portance for clinical applications, such as
An additional important field of ap- quantitative DNA amplification and de- quality control of blood donations130 or
plication for the combined detection of tection is nowadays implemented in DNA- biological material for implantation. In
entire sets of antigens concerns the family enhanced immunoassays. This enables an this respect, IPCR was already proven
of cytokines, because cytokine profiling easy, robust and highly precise signal read- to be most valuable for the detection of
has been demonstrated to help under- out of these assays. The second major chal- low levels of viruses and infectious pro-
stand the inflammatory response in pa- lenge of DNA-enhanced immunoassays, teins (see above). In view of the growing
tients with trauma, shock, and sepsis by the actual conjugation of DNA marker importance of single cell analytics,131 the
studying specific cell-mediated immune with detection antibodies, has found sev- application of IPCR to analyze targets in
responses in blood.125 Typically, multi- eral different solutions. Moreover, from a single blastocytes132 suggests that this area
plexed microarray-based techniques have user’s point-of-view, it is nowadays solved of research will also benefit from DNA-
been used for organ failure assessment126 by service of commercial providers, as is enhanced immunoassays. Furthermore,
and mortality prediction.127 Comprehen- the case for conventional ELISA reagents. environmental monitoring and homeland
sive cytokine libraries have already been While the majority of challenges in general security issues provide additional chal-
analyzed by conventional multiplex ana- method development have been addressed lenging tasks for future applications.
lytics, in particular using microarrays, and by adjustment of assay strategy, three Another rewarding field for DNA-
the performance of immuno-RCA was major trends can be foreseen for future enhanced immunoassays is their combina-
impressively demonstrated by an 11-plex applications: tion with assays aimed at DNA/RNA an-
cytokine assay in serum.58 For this appli- (1) Homogenous single-phase assays alytes, the latter of which are already avail-
cation, however, extensive careful testing are becoming increasingly popular be- able in a variety of commercial products.
of the cross-reactivity of more than 130 cause this type of assay improves reagent The detection of the genetic information
analytes as well as systematic adaptation coupling, allows for single-step assays and or transcription products of a pathogenic
of the assay procedure was necessary. is well suited for automation. Neverthe- organism only allows for indirect con-
A polyplex assay of cytokines Il-2, Il- less, one major advantage is lost in this clusions on the actual distribution and
4, VGEF, PDGF-BB and insulin was type of assay, that is, they are no longer status of the organism. The most inter-
demonstrated by Gullberg et al. using conveniently compatible with established esting parts of it, that is shell particles for
the proximity ligation method64 while a ELISA protocols, reagents and instru- immunization or disease-inducing toxins,
number of other cytokines had already mentation. While certainly well suited for are not detectable by nucleic acid analysis
been studied in individual IPCR applica- stand-alone applications, it remains to be alone. Conventional immunoassays would
tions, such as TNFa,16,96,104,128 IL-18,17 IL- seen whether the limitation to specialized be helpful, but because their sensitivity is
6.23 In combination, these assays represent equipment will be a hindrance for broad not nearly in the range of PCR/RT-PCR,
a solid basis for the emerging field of flexibility and distribution in research labs they are typically not sensitive enough for
multiple protein analyses from a single and industry. early stage or low-level infections. With
sample. Therefore, the realization of poly- (2) Ultrasensitive multiplex and poly- the aid of DNA-enhanced immunoassays,
plex IPCR assays again nicely illustrates plex applications for the detection and protein detection can be linked to nu-
how the sensitivity of DNA-enhanced quantification of minute amounts of target cleic acid analysis, in order to get the
immunoassays can be used as a powerful antigens. With the increasing knowledge whole picture of a given disease pattern
tool for creative assay design in novel gained in proteomic research, monitoring or to monitor artificially induced protein
applications. of protein families and comprehensive sets expression.133 This should allow for more
of antigens will become a major challenge precise diagnosis and therapy. Indeed,
for research. Moreover, the monitoring of initial progress towards this goal indi-
5. Summary and outlook
complex medical relevant response pat- cated that both protein- and nucleic acid
The use of DNA as a signal enhancing terns for individual treatment also calls for analyses can supplement each other in a
tool in immunological diagnostics has large-scale protein detection, preferably meaningful way.81,117 Since, both protein
found a broad variety of applications in with ultimate sensitivity.129 and DNA detection can be carried out
recent years, demonstrating the versatility (3) Tailored reagents and standardized using the same real-time PCR instrument,
of the molecule as well as the creativity kit solutions for specific analytical chal- one can foresee that IPCR assays will be
of the researchers who invented novel lenges are likely to be the major applica- capable of linking information on proteins
strategies of assay design. Almost the tion of the ultrasensitive DNA-enhanced conveniently to that of nucleic acid target
entire repertoire of natural DNA process- immunoassays. New low abundant tar- molecules.

This journal is © The Royal Society of Chemistry 2008 Analyst, 2008, 133, 702–718 | 715
IPCR and related DNA-enhanced im- 17 D. Furuya, A. Yagihashi, T. Yajima, D. 40 G. P. Nolan, Nat. Methods, 2005, 2(1), 11–
munoassays should therefore not be con- Kobayashi, K. Orita, M. Kurimoto and 12.
N. Watanabe, J. Immunol. Methods, 2000, 41 M. Lovrinovic, M. Spengler, C. Deutsch
sidered only as a better performing alter- 238(1–2), 173–180. and C. M. Niemeyer, Mol. BioSyst., 2005,
native to existing assay technologies. In- 18 L. Quijada, D. Moreira, M. Soto, C. 1(1), 64–69.
stead, if correctly optimized and adapted, Alonso and J. M. Requena, BioTechniques, 42 M. Lovrinovic and C. M. Niemeyer,
they can be used as powerful tools to fill 1997, 24(4), 660–662. Biochem. Biophys. Res. Commun., 2005,
19 X. Ke and C. M. Warner, J. Reprod. 335(3), 943–948.
the gap between ultrasensitive nucleic acid Immunol., 2000, 46(1), 1–15. 43 M. Lovrinovic and C. M. Niemeyer, Chem-
detection and robust protein detection. 20 A. S. McElhinny, N. Kadow and C. M. BioChem, 2007, 8, 61–67.
This can supplement microarray tech- Warner, Mol. Hum. Reprod., 1998, 4(10), 44 Y. C. Guo, Y. F. Zhou, X. E. Zhang, Z. P.
966–971. Zhang, Y. M. Qiao, L. J. Bi, J. K. Wen,
niques with high sensitivity detection, and 21 A. S. McElhinny, G. E. Exley and C. M. M. F. Liang and J. B. Zhang, Nucleic Acids
also help to obtain significantly more in- Warner, Am. J. Reprod. Immunol., 2000, Res., 2006, 34(8), E62.
formation from a single biological sample. 44(1), 52–58. 45 G. P. Smith, Science, 1985, 228(4705),
In the coming years, we anticipate that 22 J. Ren, L. Ge, Y. Li, J. Bai, W. C. Liu and 1315–1317.
X. M. Si, Ann. N. Y. Acad. Sci., 2001, 945, 46 X. Yu, M. P. Burgoon, A. J. Shearer and
IPCR technology will further expand into 116–118. D. H. Gilden, J. Immunol. Methods, 2007,
a wide variety of scientific fields, enabling 23 C. M. Niemeyer, M. Adler and R. Wacker, 326(1–2), 33–40.
fundamentally new insights into biologi- Nat. Protoc., 2007, 2(8), 1918–1930. 47 E. N. Brody, M. C. Willis, J. D. Smith,
24 C. M. Niemeyer, M. Adler, S. Gao and L. S. Jayasena, D. Zichi and L. Gold, Mol.
cal systems as well as novel products for
Chi, Bioconjugate Chem., 2001, 12(3), 364– Diagn., 1999, 4(4), 381–388.
many areas of human society. 371. 48 S. Fredriksson, M. Gullberg, J. Jarvius, C.
25 C. M. Niemeyer, M. Adler, B. Pignataro, Olsson, K. Pietras, S. M. Gustafsdottir, A.
S. Lenhert, S. Gao, L. Chi, H. Fuchs and Ostman and U. Landegren, Nat. Biotech-
References D. Blohm, Nucleic Acids Res., 1999, 27(23), nol., 2002, 20(5), 473–477.
45530–45561. 49 B. Chang, X. Cheng, S. Yin, T. Pan, H.
1 Principles and Practice of Immunoassay, 26 K. Lind and M. Kubista, J. Immunol. Zhang, P. Wong, S. C. Kang, F. Xiao, H.
ed. C. P. Price and D. J. Newman, Methods, 2005, 304(1–2), 107–116. Yan, C. Li, L. L. Wolfe, M. W. Miller, T.
Macmillan Reference Ltd., London, UK, 27 C. M. Niemeyer, M. Adler and D. Wisniewski, M. I. Greene and M. S. Sy,
2nd edn, 1997. Blohm, Anal. Biochem., 1997, 246(1), 140– Clin. Vaccine Immunol., 2007, 14(1), 36–43.
2 T. Sano, C. L. Smith and C. R. Cantor, 145. 50 H. Zhang, X. Cheng, M. Richter and M. I.
Science, 1992, 258(5079), 120–122. 28 J. Sperl, V. Paliwal, R. Ramabhadran, B. Greene, Nat. Methods, 2006, 12(4), 473–
3 K. B. Mullis and F. Faloona, Methods Nowak and P. W. Askenase, J. Immunol. 477.
Enzymol., 1987, 155, 335–350. Methods, 1995, 186(2), 181–194. 51 N. H. Chiu and T. K. Christopoulos, Clin.
4 R. K. Saiki, S. Scharf, F. Faloona, K. B. 29 A. McKie, D. Samuel, B. Cohen and Chem., 1999, 45(11), 1954–1959.
Mullis, G. T. Horn, H. A. Erlich and N. N. A. Saunders, J. Immunol. Methods, 52 T. K. Christopoulos and N. H. Chiu, Anal.
Arnheim, Science, 1985, 230, 1350–1354. 2002, 261(1–2), 167–175. Chem., 1995, 67(23), 4290–4294.
5 J. Swanson, Genome Technol., 2007, 30 A. McKie, D. Samuel, B. Cohen and 53 S. Banin, S. M. Wilson and C. J. Stanley,
23–26, http://www.tataa.com/files/PDF/ N. A. Saunders, J. Immunol. Methods, Clin. Chem., 2004, 50(10), 1932–1934.
GT_20070901_Sep_2007.pdf. 2002, 270(1), 135–141. 54 R. D. Joerger, T. M. Truby, E. R. Hendrick-
6 S. Nakamura, S. Katamine, T. Yamamoto, 31 P. W. Sims, M. Vasser, W. L. Wong, P. M. son, R. M. Young and R. C. Ebersole, Clin.
S. K. Foung, T. Kurata, Y. Hirabayashi, K. Williams and Y. G. Meng, Anal. Biochem., Chem., 1995, 41(9), 1371–1377.
Shimada, S. Hino and T. Miyamoto, Virus 2000, 281(2), 230–232. 55 M. Adler, M. Langer, K. Witthohn,
Genes, 1993, 7(4), 325–338. 32 H. C. Wu, Y. L. Huang, S. C. Lai, Y. Y. K. Wilhelm-Ogunbiyi, P. Schoffski, P.
7 N. L. Rosi and C. A. Mirkin, Chem. Rev., Huang and M. F. Shaio, Lett. Appl. Micro- Fumoleau and C. M. Niemeyer, J. Pharm.
2005, 105(4), 1547–1562. biol., 2001, 32(5), 321–325. Biomed. Anal., 2005, 39(5), 972–982.
8 M. Adler, Adv. Clin. Chem., 2005, 39, 239– 33 N. Henterich, A. A. Osman, E. Mendez 56 B. Schweitzer, S. Wiltshire, J. Lambert, S.
292. and T. Mothes, Nahrung, 2003, 47(5), 345– O’Malley, K. Kukanskis, Z. Zhu, S. F.
9 H. Zhang, Q. Zhao, X. F. Li and X. C. Le, 348. Kingsmore, P. M. Lizardi and D. C. Ward,
Analyst, 2007, 132(8), 724–737. 34 M. Adler and C. M. Niemeyer, in Proc. Natl. Acad. Sci. U. S. A., 2000, 97(18),
10 C. M. Niemeyer, M. Adler and R. Wacker, DNA Amplification: Current Technologies 10113–10119.
Trends Biotechnol., 2005, 23(4), 208–216. and Applications, ed. V. V. Demidov 57 S. Wiltshire, S. O’Malley, J. Lambert, K.
11 J. Barletta, Mol. Aspects Med., 2006, 27(2– and N. E. Broude, Horizon Bioscience, Kukanskis, D. Edgar, S. F. Kingsmore and
3), 224–253. Norfolk, UK, 2004, pp. 293–312. B. Schweitzer, Clin. Chem., 2000, 46(12),
12 S. Schiavo, A. El-Shafey, N. Jordan, C. 35 M. Adler, in Immunoassays, ed. 1990–1993.
Orazine, W. Wang, X. Zhou, N. H. Chiu A. M. Raem and P. Rauch, Elsevier, 58 M. Mullenix, R. Dondero, D. Hirock,
and I. S. Krull, PharmaGenomics, 2004, Munich, Germany, 2007, pp. 70–78. M. Egholm, S. Kingsmore, and L. Perlee,
36–45. 36 R. Huber, J. Eisenbraun, B. Miletzki, M. DNA Amplification: Current Technolo-
13 H. Zhou, R. J. Fisher and T. S. Papas, Adler and C. Gleiter, Phytomedicine, 2007, gies and Applications, Horizon Bioscience,
Nucleic Acids Res., 1993, 21(25), 6038– 14(SVII), 30; M. Adler and J. Eisenbraun, Wymondham, Norfolk, UK, 2004, ch. 4.2,
6039. Phytomedicine, 2007, 14(SVII), 50. pp. 313–331.
14 E. R. Hendrickson, T. M. Hatfield Truby, 37 P. Schoffski, S. Riggert, P. Fumoleau, 59 B. Schweitzer, S. Roberts, B. Grimwade, W.
R. D. Joerger, W. R. Majarian and R. C. M. Campone, O. Bolte, S. Marreaud, D. Shao, M. Wang, Q. Fu, Q. Shu, I. Laroche,
Ebersole, Nucleic Acids Res., 1995, 23(3), Lacombe, B. Baron, M. Herold, H. Z. Zhou, V. T. Tchernev, J. Christiansen,
522–529. Zwierzina, K. Wilhelm-Ogunbiyi, H. M. Velleca and S. F. Kingsmore, Nat.
15 K. Sugawara, D. Kobayashi, K. Saito, Lentzen and C. Twelves, Ann. Oncology, Biotechnol., 2002, 20(4), 359–365.
D. Furuya, H. Araake, A. Yagihashi, T. 2004, 15(12), 1816–1824. 60 D. Liu, S. L. Daubendiek, M. A. Zillmann,
Yajima, K. Hosoda, T. Kamimura and N. 38 I. Burbulis, K. Yamaguchi, A. Gordon, R. K. Ryan and E. T. Kool, J. Am. Chem. Soc.,
Watanabe, Clin. Chim. Acta, 2000, 299(1– Carlson and R. Brent, Nat. Methods, 2005, 1996, 118(7), 1587–1594.
2), 45–54. 2(1), 31–37. 61 O. Soderberg, M. Gullberg, M. Jarvius, K.
16 M. Komatsu, D. Kobayashi, K. Saito, D. 39 M. Lovrinovic, R. Seidel, R. Wacker, H. Ridderstrale, K. J. Leuchowius, J. Jarvius,
Furuya, A. Yagihashi, H. Araake, N. Tsuji, Schroeder, O. Seitz, M. Engelhard, R. S. K. Wester, P. Hydbring, F. Bahram, L. G.
S. Sakamaki, Y. Niitsu and N. Watanabe, Goody and C. M. Niemeyer, Chem. Com- Larsson and U. Landegren, Nat. Methods,
Clin. Chem., 2001, 47(7), 1297–1301. mun., 2003, (7), 822–823. 2006, 3(12), 995–1000.

716 | Analyst, 2008, 133, 702–718 This journal is © The Royal Society of Chemistry 2008
62 O. Soderberg, K. J. Leuchowius, M. 84 M. M. Richter, Chem. Rev., 2004, 104(6), 109 R. Nakaoke, S. Sakaguchi, R. Atarashi,
Kamali-Moghaddam, M. Jarvius, S. 3003–3036. N. Nishida, K. Arima, K. Shigematsu and
Gustafsdottir, E. Schallmeiner, M. 85 E. Kakizaki, T. Yoshida, H. Kawakami, M. S. Katamine, Cell. Mol. Neurobiol., 2000,
Gullberg, J. Jarvius and U. Landegren, Oseto, T. Sakai and M. Sakai, Lett. Appl. 20(6), 717–730.
Genet. Eng. (N. Y.), 2007, 28, 85–93. Microbiol., 1996, 23(2), 101–103. 110 F. Houston, J. D. Foster, A. Chong, N.
63 E. Jablonski and T. Adams, Medical 86 T. C. Chang and S. H. Huang, J. Immunol. Hunter and C. J. Bostock, Lancet, 2000,
Device, 2006, IVDT 2006, http://www. Methods, 1997, 208(1), 35–42. 356(9234), 999–1000.
devicelink.com/ivdt/archive/06/11/009. 87 D. Furuya, R. Kaneko, A. Yagihashi, T. 111 N. Hunter, J. Foster, A. Chong, S.
html. Endoh, T. Yajima, D. Kobayashi, K. Yano, McCutcheon, D. Parnham, S. Eaton, C.
64 M. Gullberg, S. M. Gustafsdottir, E. E. Tsuda and N. Watanabe, Clin. Chem., MacKenzie and F. Houston, J. Gen. Virol.,
Schallmeiner, J. Jarvius, M. Bjarnegard, 2001, 47(8), 1475–1477. 2002, 83(11), 2897–2905.
C. Betsholtz, U. Landegren and S. 88 S. M. Chye, S. R. Lin, Y. L. Chen, L. Y. 112 K. Biffiger, D. Zwald, L. Kaufmann, A.
Fredriksson, Proc. Natl. Acad. Sci. U. S. A., Chung and C. M. Yen, Clin. Chem., 2004, Briner, I. Nayki, M. Purro, S. Bottcher,
2004, 101(22), 8420–8424. 50(1), 51–57. T. Struckmeyer, O. Schaller, R. Meyer,
65 U. Landegren, E. Schallmeiner, M. Nils- 89 H. D. Hill and C. A. Mirkin, Nat. Protoc., R. Fatzer, A. Zurbriggen, M. Stack, M.
son, S. Fredriksson, J. Baner, M. Gullberg, 2006, 1(1), 324–336. Moser, B. Oesch and E. Kubler, J. Virol.
J. Jarvius, S. Gustafsdottir, F. Dahl, O. 90 T. L. Chang, C. Y. Tsai, C. C. Sun, C. C. Methods, 2002, 101(1–2), 79–84.
Soderberg, O. Ericsson and J. Stenberg, Chen, L. S. Kuo and P. H. Chen, Biosens. 113 L. Cervenakova, P. Brown, S. Soukharev,
J. Mol. Recognit., 2004, 17(3), 194–197. Bioelectron., 2007, 22(12), 3139–3145. O. Yakovleva, H. Diringer, E. L. Saenko
66 S. M. Gustafsdottir, E. Schallmeiner, S. 91 M. Maia, H. Takahashi, K. Adler, R. K. and W. N. Drohan, Electrophoresis, 2003,
Fredriksson, M. Gullberg, O. Soderberg, Garlick and J. R. Wands, J. Virol. Methods, 24(5), 853–859.
M. Jarvius, J. Jarvius, M. Howell and U. 1995, 52(3), 273–286. 114 S. Gofflot, M. B. El, D. Zorzi, L.
Landegren, Anal. Biochem., 2005, 345(1), 92 M. Adler, R. Wacker and C. M. Niemeyer, Melen, S. Roels, D. Quatpers, J. Grassi, E.
2–9. Biochem. Biophys. Res. Commun., 2003, Vanopdenbosch, E. Heinen and W. Zorzi,
67 L. Zhu, H. Koistinen, P. Wu, A. Narva- 308(2), 240–250. J. Immunoassay Immunochem., 2004, 25(3),
nen, E. Schallmeiner, S. Fredriksson, U. 93 C. A. Heid, J. Stevens, K. J. Livak and P. M. 241–258.
Landegren and U. H. Stenman, Biol. Williams, Genome Res., 1996, 6(10), 986– 115 S. B. Prusiner, Science, 1991, 252(5012),
Chem., 2006, 387(6), 769–772. 994. 1515–1522.
68 C. M. Niemeyer, T. Sano, C. L. Smith and 94 U. Spengler and T. Laue, BioTechniques, 116 C. König, C. Engemann, U. Krummrei,
C. R. Cantor, Nucleic Acids Res., 1994, 1998, (1), 56–57. R. Wacker, and R. Hoffmann in:
22(25), 5530–5539. 95 A. Solinas, L. J. Brown, C. McKeen, J. M. Prion2005 - Between fundamentals and
69 C. M. Niemeyer, L. Boldt, B. Ceyhan and Mellor, J. Nicol, N. Thelwell and T. Brown, society’s needs, Neuroprion TSE forum,
D. Blohm, Anal. Biochem., 1999, 268(1), Nucleic Acids Res., 2001, 29(20), E96. 2005, 192, also: http://priontype.com/
54–63. 96 P. P. Sanna, F. Weiss, M. E. Samson, F. E. files/Poster_Prion2005.Adobe5.0.pdf.
70 C. M. Niemeyer, R. Wacker and M. Adler, Bloom and E. M. Pich, Proc. Natl. Acad. 117 J. M. Barletta, D. C. Edelman and N. T.
Nucleic Acids Res., 2003, 31(16), e90. Sci. U. S. A., 1995, 92(1), 272–275. Constantine, Am. J. Clin. Pathol., 2004,
71 C. S. Thaxton, D. G. Georganopoulou 97 S. Gofflot, M. Deprez, B. el Moualij, A. 122(1), 20–27.
and C. A. Mirkin, Clin. Chim. Acta, 2006, Osman, J. F. Thonnart, O. Hougrand, E. 118 C. Q. Xiang, C. L. Shen, Z. R. Wu, Y. Q.
363(1–2), 120–126. Heinen and W. Zorzi, Clin. Chem., 2005, Qin, Y. Y. Zhang, C. Z. Liu, J. G. Chen and
72 J. M. Nam, K. J. Jang and J. T. Groves, Nat. 51(9), 1605–1611. S. N. Zhang, J. Occup. Health, 2007, 49(4),
Protoc., 2007, 2(6), 1438–1444. 98 M. Kubista, J. M. Andrade, M. Bengtsson, 279–284.
73 J. M. Nam, S. I. Stoeva and C. A. Mirkin, A. Forootan, J. Jonak, K. Lind, R. 119 H. Y. Chao, Y. C. Wang, S. S. Tang and
J. Am. Chem. Soc., 2004, 126(19), 5932– Sindelka, R. Sjoback, B. Sjogreen, L. H. W. Liu, Toxicon, 2004, 43(1), 27–34.
5933. Strombom, A. Stahlberg and N. Zoric, 120 R. C. Allen, S. Rogelj, S. E. Cordova and
74 J. M. Nam, C. S. Thaxton and C. A. Mol. Aspects Med., 2006, 27(2–3), 95–125. T. L. Kieft, J. Immunol. Methods, 2006,
Mirkin, Science, 2003, 301(5641), 1884– 99 A. Fischer, C. von Eiff, T. Kuczius, K. 308(1–2), 109–115.
1886. Omoe, G. Peters and K. Becker, J. Mol. 121 A. Rajkovic, B. El-Moualij, M. Uytten-
75 J. M. Nam, A. R. Wise and J. T. Groves, Med., 2007, 85(5), 461–469. daele, P. Brolet, W. Zorzi, E. Heinen, E.
Anal. Chem., 2005, 77(21), 6985–6988. 100 P. Tian and R. Mandrell, J. Appl. Micro- Foubert and J. Debevere, Appl. Environ.
76 Y. Li and D. Luo, Expert Rev. Mol. Diagn., biol., 2006, 100(3), 564–574. Microbiol., 2006, 72(10), 6593–6599.
2006, 6(4), 567–574. 101 K. Lind and M. Kubista, BioRadiations, 122 M. C. Edwards and R. A. Gibbs, PCR
77 S. Tang, J. Zhao, J. J. Storhoff, P. J. Norris, 2002, 109, 32–33. Methods Appl., 1994, 3(4), S65–S75.
R. F. Little, R. Yarchoan, S. L. Stramer, 102 M. C. Case, A. D. Burt, J. Hughes, J. M. 123 O. Henegariu, N. A. Heerema, S. R.
T. Patno, M. Domanus, A. Dhar, C. A. Palmer, J. D. Collier, M. F. Bassendine, S. J. Dlouhy, G. H. Vance and P. H. Vogt,
Mirkin and I. K. Hewlett, J Acquired Yeaman, M. A. Hughes and G. N. Major, BioTechniques, 1997, 23(3), 504–511.
Immune Defic. Syndr., 2007, 46(2), 231– J. Immunol. Methods, 1999, 223(1), 93–106. 124 N. M. Cirino, K. A. Musser and C. Egan,
237. 103 M. Case, G. N. Major, M. F. Bassendine Expert Rev. Mol. Diagn., 2004, 4(6), 841–
78 B. Ceyhan, P. Alhorn, C. Lang, D. Schueler and A. D. Burt, Biochem. Soc. Trans., 1997, 857.
and C. M. Niemeyer, Small, 2006, 2, 1251– 25(2), 374S. 125 M. Lagrelius, P. Jones, K. Franck and
1255. 104 K. Saito, D. Kobayashi, M. Sasaki, H. H. Gaines, Cytokine, 2006, 33(3), 156–
79 R. Wacker, B. Ceyhan, P. Alhorn, D. Araake, T. Kida, A. Yagihashi, T. Yajima, 165.
Schueler, C. Lang and C. M. Niemeyer, H. Kameshima and N. Watanabe, Clin. 126 F. A. Bozza, J. I. Salluh, A. M. Japiassu,
Biochem. Biophys. Res. Commun., 2007, Chem., 1999, 45(5), 665–669. M. Soares, E. F. Assis, R. N. Gomes, M. T.
357(2), 391–396. 105 R. Lieberman, Am. J. Ther., 2004, 11(6), Bozza, H. C. Castro-Faria-Neto and P. T.
80 C. Lang, D. Schuler and D. Faivre, Macro- 501–506. Bozza, Crit. Care, 2007, 11(2), R49.
mol. Biosci., 2007, 7(2), 144–151. 106 M. Adler, S. Schulz and R. Wacker, Am. 127 P. R. Knight, A. Sreekumar, J. Siddiqui, B.
81 A. S. Mweene, T. Ito, K. Okazaki, E. Ono, Biotechnol. Lab., 2006, 24(1), 17. Laxman, S. Copeland, A. Chinnaiyan and
Y. Shimizu and H. Kida, J. Clin. Microbiol., 107 E. D. Goluch, J. M. Nam, D. G. D. G. Remick, Shock, 2004, 21(1), 26–30.
1996, 34(3), 748–750. Georganopoulou, T. N. Chiesl, K. A. 128 N. Watanabe, Rinsho Byori, 2001, 49(9),
82 H. Tsujii, Y. Okamoto, H. Nakano, K. Shaikh, K. S. Ryu, A. E. Barron, C. A. 829–833.
Watanabe and T. Naraki, Int. Hepatol. Mirkin and C. Liu, Lab Chip, 2006, 6(10), 129 H. M. Azzazy, M. M. Mansour and S. C.
Commun., 1995, 3(1000), 154. 1293–1299. Kazmierczak, Clin. Chem., 2006, 52(7),
83 J. M. Barletta, D. C. Edelman, W. E. 108 M. Adler, S. Schulz, R. Fischer and C. M. 1238–1246.
Highsmith and N. T. Constantine, J. Virol. Niemeyer, Biochem. Biophys. Res. Com- 130 C. Fournier-Wirth and J. Coste, Dev. Biol.,
Methods, 2005, 127(2), 154–164. mun., 2005, 333(4), 1289–1294. 2007, 127, 61–70.

This journal is © The Royal Society of Chemistry 2008 Analyst, 2008, 133, 702–718 | 717
131 R. Zhao, Clin. Lab. Sci., 2005, 18(4), 254– 136 J. J. Storhoff and C. A. Mirkin, Chem. Rev., Proc. Natl. Acad. Sci. U. S. A., 2001, 98(10),
262. 1999, 99, 1849–1862. 5497–5502.
132 A. S. McElhinny and C. M. Warner, 137 J. Ren, Z. Chen, S. J. Juan, X. Y. Yong, B. R. 140 Y. Cao, Methods Mol. Biol., 2002, 193,
BioTechniques, 1997, 23(4), 660–662. Pan and D. M. Fan, Cancer, 2000, 88(2), 191–196.
133 M. Adler et al., unpublished results. 280–285. 141 A. McKie, A. Vyse and C. Maple, Lancet
134 M. Adler, R. Wacker, E. Booltink, B. Manz 138 J. Ren, D. M. Fan and S. J. Zhou, Chung Infect. Dis., 2002, 2(1), 18–24.
and C. M. Niemeyer, Nat. Methods, 2005, Hua Chung Liu Tsa Chih, 1994, 16(4), 247– 142 M. Adler, M. Langer, K. Witthohn, J. Eck,
2(2), 147–149. 250. D. Blohm and C. M. Niemeyer, Biochem.
135 C. M. Niemeyer and B. Ceyhan, Angew. 139 H. T. Zhang, J. E. Kacharmina, K. Biophys. Res. Commun., 2003, 300(3), 757–
Chem., Int. Ed., 2001, 40, 3685–3688. Miyashiro, M. I. Greene and J. Eberwine, 763.

718 | Analyst, 2008, 133, 702–718 This journal is © The Royal Society of Chemistry 2008

You might also like