Vous êtes sur la page 1sur 7

Review TRENDS in Biotechnology Vol.25 No.

Biosensor developments: application


to prostate-specific antigen detection
Declan A. Healy1, Conor J. Hayes1, Paul Leonard1, Louise McKenna2 and
Richard O’Kennedy1
1
School of Biotechnology and Biomedical Diagnostics Institute, National Centre for Sensor Research, Dublin City University,
Dublin 9, Ireland
2
Forensic Science Laboratory, Garda Headquarters, Phoenix Park, Dublin 8, Ireland

Prostate-specific antigen (PSA) is the best serum Prostate cancer detection using serum PSA
marker currently available for the detection of prostate measurement
cancer and is the forensic marker of choice for deter- Trace levels of PSA are naturally found in the serum;
mining the presence of azoospermic semen in some however, PCa tumor growth usually leads to the release
sexual assault cases. Most current assays for PSA of high concentrations of PSA into the circulatory system
detection are processed on large analyzers at dedi- [6]. A PSA measurement above a cut-off value of 4.0 ng/ml
cated testing sites, which require that samples be sent (and more recently 2.5 ng/ml) is generally regarded as
away for testing. This leads to delays in patient man- positive and might indicate the need for a biopsy. PSA
agement and increased administration costs. The testing is also used to monitor the response of PCa patients
recent emphasis placed on the need for point-of-care to ablative therapy such as radical prostatectomy. In this
patient management has led to the development of situation PSA should be undetectable, and a subsequent
novel biosensor detection strategies that are suitable detectable level of PSA is a sign of disease recurrence. For
for the miniaturization of assays for various targets this reason, ultrasensitive assays that are capable of
including PSA. This review highlights the current and detecting concentrations of serum PSA in the low pg/ml
novel analytical technologies used for PSA detection, region have been sought to facilitate earlier detection of
which will benefit clinicians, patients and forensic recurrence.
workers in the future. Despite its wide use, PSA is not a cancer-specific
marker, and other non-cancerous diseases of the prostate,
Introduction such as benign prostatic hyperplasia (BPH), can also lead
Prostate cancer (PCa) accounts for 10% of all deaths to increased release of PSA into the circulation. The devel-
from cancer [1–4], and the indicator most widely used to opment of assays that are capable of distinguishing
detect it is serum prostate-specific antigen (PSA) levels: between different isoforms of PSA [1,7], such as the ratio
PSA is a serine protease that is produced by the prostate of free PSA to complexed PSA, has helped to improve the
epithelium to maintain liquefaction of seminal fluid [5]. distinction between PCa and BPH, particularly in the so-
The importance of PSA as an oncological marker is called ‘diagnostic grey zone’ of 4–10 ng/ml PSA [6–10].
partly because of the lack of real alternative markers
of PCa. Most PSA testing takes place at dedicated cen-
PSA detection in forensic samples
tralized laboratories using large, automated analyzers,
In addition to its use in the diagnosis of PCa, the detection of
requiring sample transportation, increased waiting
PSA has also become the method of choice for the forensic
times and increased administration and medical costs.
determination of the presence of semen, in the absence of
The availability of near-patient or point-of-care testing
sperm, in sexual assault cases. However, the detection of
(POCT) could help to reduce the number of clinic visits,
PSA in forensic samples necessitates different assay
decrease costs to the patient and the healthcare system,
requirements than its detection in clinical samples. First,
increase patient satisfaction and improve clinical out-
it is not the determination of absolute PSA levels that is
come. Recent advances in biosensor development, using
important in forensic science, rather the ability to detect
nanoparticles and nanostructures as integral com-
PSA in what are invariably ‘dirty’ samples: contamination
ponents, have brought POCT for PSA closer to reality.
with other body fluids or dirt, scarcity of the sample, the need
This review highlights some important biosensor formats
for extraction from different types of fabric – leading to low
that have recently been applied to PSA detection and
extraction efficiencies – and the decomposition of samples in
emphasizes the advantages or disadvantages of each
cadavers or following laundering of fabrics are all common
biosensor type with respect to its suitability for use in
challenges to PSA detection in forensic samples [11]. Second,
the POCT devices of the future.
owing to low concentrations of PSA being naturally present
in the tissues and body fluids of some females, PSA assays
Corresponding author: O’Kennedy, R. (Richard.OKennedy@dcu.ie). for forensic use require only limited sensitivity to avoid the
Available online 24 January 2007. danger of false-positive results.
www.sciencedirect.com 0167-7799/$ – see front matter ß 2007 Elsevier Ltd. All rights reserved. doi:10.1016/j.tibtech.2007.01.004
126 Review TRENDS in Biotechnology Vol.25 No.3

Table 1. Examples of some commercially available tests commonly used for PSA determinations in serum
Manufacturer Assay PSA isoform detected Lowest detection Expected range Sample Assay time Sample
limit (ng/ml) volume (mins) throughput
(ng/ml) required (ml) per hour
Abbott Diagnostics Architect Free PSA <0.008 0–30 NS 29 200
Total PSA <0.008 0–100 NS 29 200
AxSYM Free PSA <0.02 0–10 NS 19 62
Total PSA <0.04 0–50 NS 19 62
IMx Total PSA <0.04 0–50 NS 45 32 a
Diagnostic Products Immulite Total PSA 0.04 0–150 10 30 200
Corporation Free PSA 0.02 0–25 10 30 200
Third-generation PSA b 0.003 0–20 50 60 NS
Roche Elecsys Total PSA 0.002 0–100 5–50 18 85
Free PSA 0.01 0–50 5–50 18 85
Beckman Coulter Access-Hybritech Total PSA <0.008 0–150 25 20 NS
Free PSA <0.005 0–20 25 20 NS
Bayer Diagnostics ADVIA PSA-ACT 0.03 0–100 NS NS 240
a
Actual sample throughput is 24 samples in 45 min. Abbreviation: NS, not specified on product package insert. bThird-generation PSA assays are sensitive to 0.005 ng/ml,
compared with first-generation (0.5 ng/ml) and second-generation (0.05 ng/ml) assays.

Currently available PSA immunoassays centralized laboratory testing for clinical PSA samples
Currently, most PSA testing takes place at dedicated, because of their general lack of sensitivity and, at best,
centralized laboratories on large, automated high-through- semi-quantitative nature (Table 3). Despite this, PSA
put systems, and numerous analyzer-run PSA assays are immunostrip devices have been applied to PSA detection
currently available in the marketplace (Table 1). The in forensic semen samples, particularly in rape cases. The
advantages of such systems include low detection limits relatively low sensitivities of immunostrip devices help to
(in the region of 0.05–0.005 ng/ml), proven reliability and avoid the false-positive results caused by the low levels of
high-throughput of samples. However, an important dis- PSA that can be present in many female tissues and fluids.
advantage associated with the fact that these large sys- Although more suited for use with forensic samples than
tems are only found in dedicated laboratories (Table 2) is centralized immunoassay tests, membrane tests still suffer
the requirement for sample transportation to the testing from several problems related to specificity, false positives,
site: this results in delays in processing and reporting the interference from matrix components and the instability of
results back to the clinician. Thus, the time between the PSA [14–16].
decision to test and the therapeutic action taken on the test
result, termed the therapeutic turnaround time (TAT) [12], Biosensor development: a trend towards point-of-care
is often of the order of several weeks. The ideal situation, PSA measurement
where results are returned within a matter of minutes, An ongoing revolution in the area of medical diagnostics is
could be facilitated through the introduction of portable the development of biosensors that have the potential for
POCT devices. Patient experience would be revolutionized use in POCT devices. These would provide rapid and
through a reduction in the number of clinic visits, reliable quantitative results ‘anytime, anywhere’, for
decreased costs, increased patient satisfaction and example, clinics and hospital emergency departments. In
improved clinical outcome [13], similar to that seen for order for POCT to become a reality, current biological
other non-emergency diseases such as diabetes. Further- testing formats must be reduced to the size of handheld
more, the portability of such devices would have important devices that require only small amounts of sample and
implications for the possibility of high-quality testing of reagents, while maintaining a multi-analyte, high-
forensic samples at crime scenes. throughput, specific and sensitive assay. Recent advances
In an effort to provide a POCT alternative to centralized in biosensor development, using nanoparticles and nanos-
PSA testing, immunochromatographic membrane tests, tructures as integral components, have enabled the devel-
also known as immunostrip tests, have been developed. opment of analyte-detection methods in the nanoscale
The major advantage of immunostrip devices is their (Figure 1). These have obvious advantages for use in POCT
low cost, robust nature and ease of use; however, immuno- settings compared with traditional measurement tech-
strip devices have not proved to be a real alternative to niques. A biosensor is an analytical device that brings

Table 2. Limitations of current PSA testing technologies


Features of current PSA immunoassays Limitations
Run on large, high-throughput analyzers  High cost of analyzer and associated instrumentation
 Restricted to dedicated laboratories
 Up to 1 ml of sample required per test
Testing at dedicated laboratories  Requirement for sample transportation at high cost
 Requirement for storage until testing
 Several weeks from testing to results
Complicated technology  Requirement for highly trained technical personnel
Thousands of samples from different sources analyzed per day  High administration costs
 Potential for sample mix-ups
Each PSA isoform measured using different reagents  Many sets of reagents required, leading to high cost

www.sciencedirect.com
Review TRENDS in Biotechnology Vol.25 No.3 127

Table 3. Performance characteristics of some commercially available immunostrip PSA tests


PSA strip test Manufacturer Sample type Compared with PSA Sensitivity Specificity Refs
cut-off (ng/ml) (%) (%)
Biosign PSA test Princeton BioMeditech Corp, Serum Tandem-E 4.0 63 92 [43]
USA Abbott IMX 4.0 68 95
Chembio Chembio, USA Serum DPC Immulite 4.0 67 87 [44]
Medpro Medpro, UK Serum DPC Immulite 4.0 87 88 [44]
Coulter Hybritech 3.1 64 75 [45]
PSA semiquant test Seratec Diagnostica, Serum DPC Immulite 4.0 80 97 [44]
Germany
OneStep PSA test Syntron, Serum DPC Immulite 4.0 93 93 [44]
USA
Onco-screen FF Diagnostics, Serum Tandem-E 4.0 76 93 [46]
PSA test Germany
PSA strip test Cardimac GmBh, Whole blood Abbott IMX 4.0 91 84 [47]
Germany

together an immobilized biological sensing material and a the recognition element of the devices. Descriptions of all
transducer, to produce a quantifiable signal that is pro- the newly developed biosensor formats is outside the scope
portional to the concentration of the analyte. Biosensors of this short review; therefore, the following sections high-
comprise three distinct components: (i) a biorecognition light some of the more important signal transduction
element, usually an antibody, to recognize and bind the developments that have been applied to PSA biosensor
analyte or analytes of interest; (ii) a transducing element development and the trend towards the use of label-free
that converts the interactions of biomolecules (e.g. anti- modalities (Table 4). Particular emphasis is given to the
body and antigen) into a quantifiable signal; and (iii) a advantages or disadvantages of each signal transduction
readout system. The development of biosensors for the method with respect to multiplexing potential and its
measurement of PSA has almost exclusively used the suitability for incorporation into POCT devices. The suit-
binding of PSA by high-affinity intact antibodies to form ability of biosensors for the multiplexed analyses of several

Figure 1. Schematic representations of some biosensor formats applied to PSA detection. (a) Immuno-PCR: after PSA capture by anti-PSA antibodies on the biosensor
surface, a DNA-labeled detection antibody binds. After washing steps, DNA is amplified by real-time PCR, where during the exponential phase of the PCR the amount of
product formed reflects the amount of PSA in the sample. Adapted from [24]. (b) Biobarcode assay: PSA binds to anti-PSA antibodies coated onto magnetic microparticles.
Nanoparticle probes coated with anti-PSA antibody and barcode-DNA strands are used to bind immobilized PSA. Following washing, barcode DNA is dehybridized from the
nanoparticle probes and quantified. Adapted from [26]. (c) SERS: binding of PSA to anti-PSA antibodies functionalized with Raman reporter molecules (RRMs) and
immobilized onto gold microparticles leads to altered spectroscopic output of the RRMs, which can be used to quantify the concentration of PSA. Adapted from [21].
(d) Electrical detection of micro-cantilever bending: piezoresistive microcantilevers, functionalized with anti-PSA antibodies, bend owing to surface stress caused by PSA
capture. Micro-cantilever bending is measured by electrical detection. Adapted from [40].

www.sciencedirect.com
128 Review TRENDS in Biotechnology Vol.25 No.3

Table 4. Examples of biosensor formats applied to PSA detection


Signal transduction Assay format and/or nanostructure used Lowest detection Sample Assay time Refs
limit volume (mins)
required
(ml)
Time-resolved Europium (III) chelate-dyed polystyrene nanoparticles 0.21 ng/ml 20 240 [48]
fluorescence Silica-coated terbium (III) fluorescent nanoparticles 7.0 pg/ml 45 >180 [18]
Terbium complex-doped zirconia nanoparticles 0.4 ng/ml 45 >180 [20]
Europium(III) nanoparticle labels and streptavidin–biotin technology 0.83 pg/ml 5 >150 [19]
SERS Gold nanoparticles 1.0 pg/ml 40 550 [21]
Real-time Immuno-PCR Sandwich assay with DNA label on detection antibody 0.2 pg/ml a 5.5 >150 [24]
Immuno-RCA Sandwich assay with DNA label on detection antibody 0.1 pg/ml 10 180 [25]
Biobarcode Gold nanoparticles 1.0 fg/ml b 10 80 [26]
1.0–10.0 fg/ml 250 210 [27]
Enzyme and/or Lateral flow immunostrip containing an electrochemical transducer 3.0 ng/ml 11 10–30 [29]
impedance
Enzyme and/or Sandwich immunoassay on three-electrode system 0.25 ng/ml 10 60 [31]
amperometric
Surface plasmon Commercial SPR biochip with signal enhancement using a sandwich- 18.1 ng/ml 100 14 [35]
resonance assay format 1.0 ng/ml 130 6.5 [34]
0.15 ng/ml NS NS [49]
Surface plasmon Sandwich-immunoassay format on commercial SPR biochip 3 pg/ml 500 45 [50]
fluorescence
spectroscopy
Electrical Resonance frequency shift in a nanomechanical microcantilever 10 pg/ml NS NS [36]
100 ng/ml 20 60 [37]
Surface stress bending of piezoresistive self-sensing microcantilevers 10 ng/ml NS 10 [38]
Conductance change in silicon nanowire sensor chip 100 fg/ml NS 30 [40]
a
Sensitivity reported by authors as 4.8  105 PSA molecules, converted to pg/ml for consistency; bsensitivity reported as 30 aM, converted to fg/ml for consistency.
Abbreviation: NS, not specified by authors.

markers simultaneously will be particularly important for this is possible because Raman bands are sufficiently
distinguishing PCa from benign diseases, such as BPH, narrow to reduce the likelihood of spectral overlap when
and for facilitating better detection of PSA in forensic multiple labels are used.
samples, through the incorporation of antibodies against
epitopes of PSA that are more easily identified in contami- DNA labels as surrogate markers: from immuno-PCR to
nated samples. biobarcode assays
DNA tags have been used as signal amplification and
Label-based signal transduction transducing mechanisms in biosensor development for
Fluorescent labels the past 15 years. They have great potential for use in
Although fluorescence-dependent signal generation has multi-analyte detection systems because different specific
disadvantages with regard to its use in multiplexed assays, sequences can be associated with each individual target
fluorescence continues to be a major transduction modality analyte of interest. The first DNA label-based technique
in biosensors for the detection of PSA using optical readout was known as immuno-PCR [23]; and this used a secondary
systems. In particular, fluorescent nanoparticles, such as antibody labeled with a DNA molecule that could be
quantum dots and luminophore-doped nanoparticles, have amplified by PCR as a signal amplification method. More
provided an attractive field of research for biosensor devel- recently, real-time PCR, using fluorescence measurements
opment because they have high photostability and narrow to follow the DNA amplification, has been used in biosen-
emission peaks, enabling greater multiplexing potential. sors to detect 4.8  105 PSA molecules in a 5 ml sample –
With respect to PSA assay development, europium(III)- equivalent to a sensitivity of 0.2 pg/ml [24]. Although
and terbium (III)-doped fluorescent polystyrene and zirco- suitable for multiplex protein quantification, real-time
nia nanoparticles have been used for the detection of PSA immuno-PCR-based biosensors are currently hindered
to a concentration <1 pg/ml [17–20]. by long assay times (3–4 h) and the need for the incorp-
oration of multiple components and thermal cycling.
Surface-enhanced Raman spectroscopy Rolling-circle amplification (RCA) was used as a reporter
Surface-enhanced Raman scattering (SERS) has been used system for the sensitive detection of PSA, in an assay
in biosensor development as an alternative to fluorescence termed immuno–RCA. An oligonucleotide primer is
labeling. Raman dyes are used to label antibodies that are attached to a reporter antibody that is specific for the target
attached to gold nanoparticle probes; the light scattered analyte and is then amplified to generate an extended DNA
from the Raman reporter molecule (RRM) provides infor- molecule containing many copies of the circular DNA
mation about the vibrational quantum states of a molecule. sequence, which remains attached to the reporter antibody
SERS was used in a sandwich-immunoassay format for the for detection by fluorescent imaging. PSA has been detected
detection of PSA to a concentration of 1 pg/ml [21]. SERS at 0.1 pg/ml in a microspot array assay using this technique
was also used for the detection of multiple analytes [22]: [25].
www.sciencedirect.com
Review TRENDS in Biotechnology Vol.25 No.3 129

A highly sensitive, DNA-labeled, nanoparticle-based presented as far back as 1995 [30]. More recently, there
assay system that does not require signal amplification has been a trend towards the use of screen-printed electro-
has recently been developed [26]. This ‘biobarcode’ tech- des owing to their simple and low-cost fabrication and ease
nology relies on the use of magnetic beads that are func- of use. For example, amperometric detection of PSA at
tionalized with antibodies – to capture the target of 0.25 ng/ml using a three-electrode system has been
interest – and nanoparticle probes with both a DNA oli- reported [31].
gonucleotide sequence (the barcode) that is unique for each Although amperometric biosensors currently suffer
of the target proteins and a specific antibody that binds, in from the necessity for washing and separation of the free
a sandwich-assay format, to the target protein, already from the bound label, simplification of these biosensors
captured by the antibody on the magnetic beads. Following holds great promise for their incorporation into POCT
magnetic separation of the complex, the target protein devices of the future. Recent advances, such as the devel-
concentration is determined by measurement of the oligo- opment of a reagentless amperometric immunosensor
nucleotide (barcode) levels, which are directly related to based on direct electrochemistry enzyme labels and the
the levels of the target protein. PSA was detected at 1 fg/ml testing of microfluidic devices using amperometric detec-
(30 aM) concentration in a 10 mL sample, in a PCR-less tion strategies on microchip platforms, might be of great
biobarcode assay [26]. A recent modification to the barcode importance to POCT devices of the future. Furthermore,
assay for PSA detection has been reported [27]; here, a the suitability of amperometric biosensors to multiplex
brief exposure to silver stain increased the size and light- analyte detection has already been demonstrated, where
scattering properties of nanoparticle tracers, enabling PSA electrode arrays with multiple, individually addressable
to be detected at 1–10 fg/ml with low resolution optics and electrochemical transducers have been fabricated [32].
without the need for laser excitation. The high sensitivity
and huge multiplexing attributes of biobarcode assays are Label-free transduction
particularly suitable for use in POCT devices. Biosensors that use label-free transduction methods do not
require a reporter molecule to signal the presence of target
Electrochemical signal transduction analyte on the sensor surface, thereby enabling the use of
Electrochemical devices have received much attention in unmodified samples, with the possibility of ‘real-time’
the field of biosensor development because they provide a measurement. Examples of some label-free methods used
simple, inexpensive and accurate platform for the in PSA biosensors are described below.
measurement of the target analyte [28]. Electrochemical
biosensors determine the level of the analyte by detecting Surface plasmon resonance
the changes in either potential, current, capacitance, con- Many of the biosensors currently available on the market-
ductance or impedance that are caused by a specific bior- place use the optical properties of lasers to monitor the
ecognition reaction. interactions of biomolecules. Surface plasmon resonance
The development of a rapid, single-use, lateral flow, (SPR) is an optical–electrical technology that involves
nitrocellulose immunostrip test device, which is based the interaction of light with the electrons of a metal.
on capacitance measurement, has recently been reported The technique enables rapid, label-free monitoring of bio-
[29]. This device incorporates an electrochemical transdu- molecular reactions in real-time, and requires only a small
cer coated with a pH-sensitive polymer layer to enable amount of sample. SPR measures changes in the refractive
measurement of PSA to a sensitivity of 10 ng/ml: binding of index and thus in the resonance angle at which polarized
PSA to an immobilized anti-PSA antibody and an anti-PSA light is reflected from a surface, which is, in turn, related to
antibody–urease conjugate catalyzes the hydrolysis of ur- a change in layer thickness or mass upon antigen binding
ea, thereby generating ammonia, leading to an increase in to an antibody-coated gold layer [33].
the pH of the reaction solution. This leads to the degra- SPR-based detection of different PSA isoforms to 1 ng/
dation of the polymer layer coating the transducer, which ml using a commercial SPR biosensor (Biacore AB; http://
can be monitored as a change in the capacitance of the www.biacore.com/) has been reported by several groups
system. In addition to being robust and inexpensive to [34,35]. However, the Biacore system is expensive to pur-
make, this system has the advantage of requiring just one chase and unsuitable for applications that require port-
manual step: the addition of 11 ml of sample. The use of ability owing to its size. Recent advances in the
impedance- and capacitance-based immunosensors and miniaturization of SPR technology have made possible
immunostrip devices in future multiplexing POCT devices the development of portable systems that strengthen
might be particularly suited to forensic applications, the probability of SPR biosensor incorporation into future
although significant improvements in their sensitivities POCT devices. The Spreeta 2000 SPR biosensor elements,
will be required before they are used on clinical samples. manufactured by Texas Instruments (http://www.ti.com/),
Amperometric biosensors are based on the measure- enables the simultaneous label-free analysis of multiple
ment of the current associated with an electrochemically analytes in a portable format. Furthermore, biosensors
active species that is produced by, in general, an enzyme that use the SPR of noble metal nanoparticles and nanos-
label. Amperometric detection of enzyme labels has tructures, known as nanoSPR, for the optical transduction
the advantage of providing higher sensitivity than impe- of biorecognition events have been developed. In nanoSPR,
dance and capacitance methods and uses accurate sensors visible light induces the collective oscillations of the sur-
that are simple and inexpensive to produce. An example of face electrons and is responsible for the intense colors
an amperometric immunosensor for PSA detection was emitted by colloidal solutions of noble metals, including
www.sciencedirect.com
130 Review TRENDS in Biotechnology Vol.25 No.3

gold and silver [36]. In particular, nanoSPR can be applied in future POCT devices for the screening of numerous
to a chip format, in which nanoparticles with different diseases using a single blood sample.
optical signatures and coated with different antibody spe-
cificities [37] are immobilized in an array format for high- Future perspectives and conclusions
throughput screening of biomolecular binding interactions Recent developments in biosensor technology have enabled
in a real-time, label-free manner. the transition from centralized to near-patient PSA testing
to emerge from the realms of possibility and become closer
Electrical and electromechanical signal transduction to reality. In future, results will be available within min-
The advent of nanostructures that act as signal utes, thereby revolutionizing the patient experience
transducers in a label-free manner undoubtedly show through reduced hospital or clinic visits, decreased costs
the most promise for use in POCT devices of the future. and improved clinical outcomes. The use of nanoparticle-
Nanostructure-based assays will prove to be both robust based assays for PSA, comprising indirect protein ampli-
and inexpensive to produce owing to the elimination of the fication technologies such as immuno-PCR and biobarcode
need for sample preparation and labeling steps. assays, electrochemical assays based upon molecular
Micro-cantilever biosensors do not require a reporter probes, and assays using reporter molecules such as RRMs,
molecule to signal the presence of the target analyte on the offer higher sensitivities than traditional ELISAs on a
sensor surface, thereby enabling the use of unmodified miniaturized scale. The trend towards the use of label-free
samples, with the possibility of real-time measurement. biosensor formats, including SPR, microcantilever and
Microcantilevers are particularly suited to incorporation in nanowire-based assays, for PSA detection, enable the
POCT devices owing to their minute size (100 mm2), and use of unmodified samples, with the possibility of highly
lend themselves to the possibility of microcantilever sensitive real-time measurement. In addition to the need
arrays, enabling multiplex analysis of numerous markers. for multi-analyte-measuring capabilities, the success of a
Microcantilever biosensors can be operated in one of two particular biosensor format in future POCT devices will
modes: resonance response variation (microbalance depend on the ability to incorporate the format into devices
method) and cantilever bending (surface-stress method). that comprise simple and efficient sample handling and
Using the microbalance method, Kim et al. have reported readout systems. Such laboratory-on-a-chip, or micro-total
that electrical measurement of the resonant frequency analysis systems (mTAS), will incorporate the functions of
change of an anti-PSA-antibody-coated microcantilever a full-scale laboratory in a portable device that will provide
upon PSA binding enables detection of PSA in solution rapid and accurate sample analysis of minute sample
at concentrations as low as 10 pg/ml [38,39]. Unfortu- volumes in every environment.
nately, the microbalance method suffers from an undesir-
able resonant frequency change due to variations in the Acknowledgements
We thank the following for financial support: Enterprise Ireland (DAH),
viscosity of the medium and cantilever damping in a liquid
Cancer Research Ireland (CJH) and Science Foundation Ireland (PL).
environment [40]. Therefore, Kim’s group have sub-
sequently used electrical detection of cantilever bending References
due to surface-stress changes, to measure PSA-antibody 1 Ozen, H. and Sozen, S. (2006) PSA isoforms in prostate cancer
binding on the surface of micro-fabricated, self-sensing detection. Eur. Urol. Suppl. 5, 495
microcantilevers, with a reported sensitivity of 10 ng/ml 2 Aus, G. et al. (2005) EAU guidelines on prostate cancer. Eur. Urol. 48,
[40]. Microcantilever technology is particularly suited to 546–551
3 Zhu, Y. et al. (2006) Computer technology in detection and staging of
miniaturized biosensor development because it is a rapid, prostate carcinoma: a review. Med. Image Anal. 10, 178–199
sensitive, specific, cost-effective and portable technique 4 Saad, F. (2006) The treatment continuum in the management of
that does not require labeling or external excitation. In prostate cancer patients: what’s new? Eur. Urol. Suppl. 5, 539
addition, it has been demonstrated that microcantilevers 5 Nadler, R.B. et al. (1995) Effect of inflammation and benign prostatic
hyperplasia on elevated serum prostate-specific antigen levels. J. Urol.
can be used in an array format for the sensitive and
154, 407–413
simultaneous measurement of multiple unlabeled analytes 6 Stenman, U.H. et al. (1999) Prostate-specific antigen. Semin. Cancer
[41]. The incorporation of cantilever technology into micro- Biol. 9, 83–93
fluidic channels will reduce the amount of sample required. 7 Roddam, A.W. et al. (2005) Use of prostate-specific antigen (PSA)
Silicon nanowire field-effect sensors coated with anti- isoforms for the detection of prostate cancer in men with a PSA
level of 2–10 ng/ml: systematic review and meta-analysis. Eur. Urol.
body have been used for the real-time, highly sensitive
48, 386–399
detection of PSA [42]. Silicon nanowires act as signal 8 Catalona, W.J. et al. (2003) Serum pro-prostate-specific antigen
transducers through a change in conductance upon binding improves cancer detection compared to free and complexed prostate-
of a target analyte to the antibody receptor on the nanowire specific antigen in men with prostate-specific antigen of 2 to 4 ng/ml.
surface. They have been used by Zheng et al. to detect PSA J. Urol. 170, 2181–2185
9 Linton, H.J. et al. (2003) Benign prostate-specific antigen (BPSA) in
to a concentration of 0.9 pg/ml [42]. This group demon- serum is increased in benign prostate disease. Clin. Chem. 49, 253–259
strated the multiplexing ability of their nanowire sensor 10 Mikolajczyk, S.D. et al. (2004) Are multiple markers the future of
chip by simultaneously detecting both free PSA (fPSA) and prostate cancer diagnostics? Clin. Biochem. 37, 519–528
bound PSA (PSA–ACT bound) at pg/ml concentrations. 11 Johnson, E.D. and Kotowski, T.M. (1993) Detection of prostate-specific
Furthermore, the authors report the real-time multiplexed antigen by ELISA. J. Forensic Sci. 38, 250–258
12 Kost, G.J. (1995) Guidelines for point-of-care testing. Improving
detection of the cancer markers fPSA, carcinoembryonic patient outcomes. Am. J. Clin. Pathol. 104, S111–S127
antigen (CEA) and mucin-1 at concentrations in the range 13 Grieve, R. et al. (1999) Near-patient testing in diabetes clinics:
of 50–100 fg/ml – attributes that are likely to be important appraising the costs and outcomes. Health Technol. Assess. 3, 1–74
www.sciencedirect.com
Review TRENDS in Biotechnology Vol.25 No.3 131

14 Yokota, M. et al. (2001) Evaluation of prostate-specific antigen (PSA) 33 Wilson, W.D. (2002) Tech. Sight. Analyzing biomolecular interactions.
membrane test for forensic examination of semen. Leg. Med. (Tokyo) 3, Science 295, 2103–2105
171–176 34 Huang, L. et al. (2005) Prostate-specific antigen immunosensing
15 Khaldi, N. et al. (2004) Evaluation of three rapid detection methods for based on mixed self-assembled monolayers, camel antibodies and
the forensic identification of seminal fluid in rape cases. J. Forensic Sci. colloidal-gold enhanced sandwich assays. Biosens. Bioelectron. 21,
49, 749–753 483–490
16 Hochmeister, M.N. et al. (1999) Evaluation of prostate-specific antigen 35 Cao, C. et al. (2006) A strategy for sensitivity and specificity
(PSA) membrane test assays for the forensic identification of seminal enhancements in prostate-specific antigen–alpha-1-anti-chymotrypsin
fluid. J. Forensic Sci. 44, 1057–1060 detection based on surface plasmon resonance. Biosens. Bioelectron. 21,
17 Soukka, T. et al. (2001) Supersensitive time-resolved immunofluo- 2106–2113
rometric assay of free prostate-specific antigen with nanoparticle 36 Nath, N. and Chilkoti, A. (2004) Label-free colorimetric biosensing
label technology. Clin. Chem. 47, 1269–1278 using nanoparticles. J. Fluoresc. 14, 377–389
18 Ye, Z. et al. (2004) Preparation, characterization and time-resolved 37 Nicewarner-Pena, S.R. et al. (2001) Submicrometer metallic barcodes.
fluorometric application of silica-coated terbium(III) fluorescent Science 294, 137–141
nanoparticles. Anal. Chem. 76, 513–518 38 Lee, J.H. et al. (2005) Immunoassay of prostate-specific antigen (PSA)
19 Huhtinen, P. et al. (2004) Immunoassay of total prostate-specific using resonant frequency shift of piezoelectric nanomechanical
antigen using europium(III) nanoparticle labels and streptavidin– microcantilever. Biosens. Bioelectron. 20, 2157–2162
biotin technology. J. Immunol. Methods 294, 111–122 39 Hwang, K.S. et al. (2004) In situ quantitative analysis of a prostate-
20 Ye, Z. et al. (2005) Preparation, characterization and application of specific antigen (PSA) using a nanomechanical PZT cantilever. Lab
fluorescent terbium-complex-doped zirconia nanoparticles. J. Fluoresc. Chip 4, 547–552
15, 499–505 40 Wee, K.W. et al. (2005) Novel electrical detection of label-free disease
21 Grubisha, D.S. et al. (2003) Femtomolar detection of prostate-specific marker proteins using piezoresistive self-sensing micro-cantilevers.
antigen: an immunoassay based on surface-enhanced Raman Biosens. Bioelectron. 20, 1932–1938
scattering and immunogold labels. Anal. Chem. 75, 5936–5943 41 McKendry, R. et al. (2002) Multiple label-free biodetection and
22 Ni, J. et al. (1999) Immunoassay readout method using extrinsic quantitative DNA-binding assays on a nanomechanical cantilever
Raman labels adsorbed on immunogold colloids. Anal. Chem. 71, array. Proc. Natl. Acad. Sci. U. S. A. 99, 9783–9788
4903–4908 42 Zheng, G. et al. (2005) Multiplexed electrical detection of cancer
23 Sano, T. et al. (1992) Immuno-PCR: very sensitive antigen detection by markers with nanowire sensor arrays. Nat. Biotechnol. 23, 1294–
means of specific antibody–DNA conjugates. Science 258, 120–122 1301
24 Lind, K. and Kubista, M. (2005) Development and evaluation of three 43 Lein, M. et al. (1995) Rapid screening of PSA: evaluation of an
real-time immuno-PCR assemblages for quantification of PSA. immunochemical membrane strip test. Clin. Chem. 41, 1545–1547
J. Immunol. Methods 304, 107–116 44 Jung, K. et al. (1999) Rapid detection of elevated prostate-specific
25 Schweitzer, B. et al. (2000) Inaugural article: immunoassays with rolling antigen levels in blood: performance of various membrane strip
circle DNA amplification: a versatile platform for ultrasensitive antigen tests compared. Urology 53, 155–160
detection. Proc. Natl. Acad. Sci. U. S. A. 97, 10113–10119 45 Oberpenning, F. et al. (2003) Semi-quantitative immunochr-
26 Nam, J.M. et al. (2003) Nanoparticle-based bio-barcodes for the omatographic test for prostate-specific antigen in whole blood:
ultrasensitive detection of proteins. Science 301, 1884–1886 tossing the coin to predict prostate cancer? Eur. Urol. 43, 478–
27 Bao, Y.P. et al. (2006) Detection of protein analytes via nanoparticle- 484
based bio-barcode technology. Anal. Chem. 78, 2055–2059 46 Madersbacher, S. et al. (1996) Validation of a 10-minute dipstick test
28 Wang, J. (2006) Electrochemical biosensors: towards point-of-care for serum prostate-specific antigen. Eur. Urol. 30, 446–450
cancer diagnostics. Biosens. Bioelectron. 21, 1887–1892 47 Berg, W. et al. (1999) Possibility of improving the acceptance rate of
29 Fernandez-Sanchez, C. et al. (2004) Disposable noncompetitive early detection testing for prostate cancer with a one-step test for
immunosensor for free and total prostate-specific antigen based on prostate-specific antigen in whole blood. Urol. Int. 63, 102–106
capacitance measurement. Anal. Chem. 76, 5649–5656 48 Soukka, T. et al. (2003) Highly sensitive immunoassay of free prostate-
30 Meyerhoff, M.E. et al. (1995) Novel non-separation sandwich-type specific antigen in serum using europium(III) nanoparticle label
electrochemical enzyme immunoassay system for detecting marker technology. Clin. Chim. Acta 328, 45–58
proteins in undiluted blood. Clin. Chem. 41, 1378–1384 49 Besselink, G.A. et al. (2004) Signal amplification on planar and gel-type
31 Sarkar, P. et al. (2002) Amperometric biosensors for detection of the sensor surfaces in surface plasmon resonance-based detection of
prostate cancer marker (PSA). Int. J. Pharm. 238, 1–9 prostate-specific antigen. Anal. Biochem. 333, 165–173
32 Kojima, K. et al. (2003) Electrochemical protein chip with arrayed 50 Yu, F. et al. (2004) Surface plasmon fluorescence immunoassay of free
immunosensors with antibodies immobilized in a plasma-polymerized prostate-specific antigen in human plasma at the femtomolar level.
film. Anal. Chem. 75, 1116–1122 Anal. Chem. 76, 6765–6770

The ScienceDirect collection


ScienceDirect’s extensive and unique full-text collection covers more than 1900 journals, including
titles such as The Lancet, Cell, Tetrahedron and the full suite of Trends, Current Opinion and Drug
Discovery Today journals. With ScienceDirect, the research process is enhanced with unsurpassed
searching and linking functionality, all on a single, intuitive interface.

The rapid growth of the ScienceDirect collection is a result of the integration of several prestigious
publications and the ongoing addition to the Backfiles – heritage collections in a number of
disciplines. The latest step in this ambitious project to digitize all of Elsevier’s journals back to
volume one, issue one, is the addition of the highly cited Cell Press journal collection on
ScienceDirect. Also available online for the first time are six Cell titles’ long-awaited Backfiles,
containing more than 12,000 articles that highlight important historic developments in the field of
life sciences.

For more information, visit www.sciencedirect.com

www.sciencedirect.com

Vous aimerez peut-être aussi